Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Cell Rep ; 37(1): 109767, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34610308

ABSTRACT

Cardiac metabolism is a high-oxygen-consuming process, showing a preference for long-chain fatty acid (LCFA) as the fuel source under physiological conditions. However, a metabolic switch (favoring glucose instead of LCFA) is commonly reported in ischemic or late-stage failing hearts. The mechanism regulating this metabolic switch remains poorly understood. Here, we report that loss of PHD2/3, the cellular oxygen sensors, blocks LCFA mitochondria uptake and ß-oxidation in cardiomyocytes. In high-fat-fed mice, PHD2/3 deficiency improves glucose metabolism but exacerbates the cardiac defects. Mechanistically, we find that PHD2/3 bind to CPT1B, a key enzyme of mitochondrial LCFA uptake, promoting CPT1B-P295 hydroxylation. Further, we show that CPT1B-P295 hydroxylation is indispensable for its interaction with VDAC1 and LCFA ß-oxidation. Finally, we demonstrate that a CPT1B-P295A mutant constitutively binds to VDAC1 and rescues LCFA metabolism in PHD2/3-deficient cardiomyocytes. Together, our data identify an oxygen-sensitive regulatory axis involved in cardiac metabolism.


Subject(s)
Carnitine O-Palmitoyltransferase/metabolism , Fatty Acids/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Procollagen-Proline Dioxygenase/metabolism , Voltage-Dependent Anion Channel 1/metabolism , Animals , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/deficiency , Carnitine O-Palmitoyltransferase/genetics , Diet, High-Fat , Fatty Acids/chemistry , Glucose/metabolism , Hydroxylation , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Lipid Peroxidation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mutagenesis, Site-Directed , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/genetics , Protein Binding , Voltage-Dependent Anion Channel 1/genetics
2.
Cell Death Differ ; 27(7): 2131-2142, 2020 07.
Article in English | MEDLINE | ID: mdl-31959916

ABSTRACT

Intestinal epithelia self-renew constantly and generate differentiated cells such as secretary goblet cells. The intestine goblet cells secrete gel-forming mucins that form mucus to create a barrier of defense. We reported previously that loss of prolyl hydroxylase (PHD) 3 led to disruption of the intestinal epithelial barrier function. However, the underlying mechanism remains elusive. Here, we demonstrate that PHD3 controls the generation of intestine goblet cell. We found that genetic ablation of Phd3 in mice intestine epithelial cells reduced the amount of goblet cells. Mechanistically, PHD3 bounds the E3 ubiquitin ligase HUWE1 and prevented HUWE1 from mediating ubiquitination and degradation of ATOH1, an essential driver for goblet cell differentiation. The prolyl hydroxylase activity-deficient variant PHD3(H196A) also prevented ATOH1 destruction. A genetic intestine epithelial PHD3(H196A)-knockin had no effect on ATOH1 expression or goblet cell amount in mice, suggesting that the PHD3 prolyl hydroxylase activity is dispensable for its ability to control ATOH1 expression and goblet cell generation. In dextran sulfate sodium (DSS)-induced experimental colitis, PHD3-knockout rather than PHD3(H196A)-knockin sensitized the mice to DSS treatment. Our results reveal an additional critical mechanism underlying the regulation of ATOH1 expression and goblet cell generation and highlight that PHD3 plays a role in controlling intestine goblet cell generation in a hydroxylase-independent manner.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Goblet Cells/metabolism , Intestines/cytology , Procollagen-Proline Dioxygenase/metabolism , Animals , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Dextran Sulfate , Epithelial Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Procollagen-Proline Dioxygenase/deficiency , Protein Binding , Protein Stability , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
4.
J Pathol ; 241(4): 547-558, 2017 03.
Article in English | MEDLINE | ID: mdl-27981571

ABSTRACT

Prolyl hydroxylase domain-containing proteins (PHDs) regulate the adaptation of cells to hypoxia. Pan-hydroxylase inhibition is protective in experimental colitis, in which PHD1 plays a prominent role. However, it is currently unknown how PHD1 targeting regulates this protection and which cell type(s) are involved. Here, we demonstrated that Phd1 deletion in endothelial and haematopoietic cells (Phd1f/f Tie2:cre) protected mice from dextran sulphate sodium (DSS)-induced colitis, with reduced epithelial erosions, immune cell infiltration, and colonic microvascular dysfunction, whereas the response of Phd2f/+ Tie2:cre and Phd3f/f Tie2:cre mice to DSS was similar to that of their littermate controls. Using bone marrow chimeras and cell-specific cre mice, we demonstrated that ablation of Phd1 in haematopoietic cells but not in endothelial cells was both necessary and sufficient to inhibit experimental colitis. This effect relied, at least in part, on skewing of Phd1-deficient bone marrow-derived macrophages towards an anti-inflammatory M2 phenotype. These cells showed an attenuated nuclear factor-κB-dependent response to lipopolysaccharide (LPS), which in turn diminished endothelial chemokine expression. In addition, Phd1 deficiency in dendritic cells significantly reduced interleukin-1ß production in response to LPS. Taken together, our results further support the development of selective PHD1 inhibitors for ulcerative colitis, and identify haematopoietic cells as their primary target. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Colitis, Ulcerative/drug therapy , Macrophages/metabolism , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Animals , Bone Marrow/drug effects , Bone Marrow/immunology , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Colon/drug effects , Colon/pathology , Dendritic Cells/drug effects , Dendritic Cells/pathology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Gene Deletion , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lipopolysaccharides , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/genetics
5.
Cell Death Dis ; 7: e2214, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27148687

ABSTRACT

Mitochondrial impairment induced by oxidative stress is a main characteristic of intrinsic cell death pathways in neurons underlying the pathology of neurodegenerative diseases. Therefore, protection of mitochondrial integrity and function is emerging as a promising strategy to prevent neuronal damage. Here, we show that pharmacological inhibition of hypoxia-inducible factor prolyl-4-hydroxylases (HIF-PHDs) by adaptaquin inhibits lipid peroxidation and fully maintains mitochondrial function as indicated by restored mitochondrial membrane potential and ATP production, reduced formation of mitochondrial reactive oxygen species (ROS) and preserved mitochondrial respiration, thereby protecting neuronal HT-22 cells in a model of glutamate-induced oxytosis. Selective reduction of PHD1 protein using CRISPR/Cas9 technology also reduced both lipid peroxidation and mitochondrial impairment, and attenuated glutamate toxicity in the HT-22 cells. Regulation of activating transcription factor 4 (ATF4) expression levels and related target genes may mediate these beneficial effects. Overall, these results expose HIF-PHDs as promising targets to protect mitochondria and, thereby, neurons from oxidative cell death.


Subject(s)
Hydroxyquinolines/pharmacology , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Neurons/drug effects , Procollagen-Proline Dioxygenase/genetics , Prolyl-Hydroxylase Inhibitors/pharmacology , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Adenosine Triphosphate/agonists , Adenosine Triphosphate/biosynthesis , Animals , Apoptosis/drug effects , CRISPR-Cas Systems , Cell Line , Gene Expression Regulation , Glutamic Acid/toxicity , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/genetics , Mice , Neurons/cytology , Neurons/metabolism , Oxidative Phosphorylation/drug effects , Oxidative Stress , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction
6.
Sci Rep ; 6: 24618, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-27094951

ABSTRACT

Obesity is associated with local tissue hypoxia and elevated hypoxia-inducible factor 1 alpha (HIF-1α) in metabolic tissues. Prolyl hydroxylases (PHDs) play an important role in regulating HIF-α isoform stability. In the present study, we investigated the consequence of whole-body PHD1 gene (Egln2) inactivation on metabolic homeostasis in mice. At baseline, PHD1-/- mice exhibited higher white adipose tissue (WAT) mass, despite lower body weight, and impaired insulin sensitivity and glucose tolerance when compared to age-matched wild-type (WT) mice. When fed a synthetic low-fat diet, PHD1-/- mice also exhibit a higher body weight gain and WAT mass along with glucose intolerance and systemic insulin resistance compared to WT mice. PHD1 deficiency led to increase in glycolytic gene expression, lipogenic proteins ACC and FAS, hepatic steatosis and liver-specific insulin resistance. Furthermore, gene markers of inflammation were also increased in the liver, but not in WAT or skeletal muscle, of PHD1-/- mice. As expected, high-fat diet (HFD) promoted obesity, hepatic steatosis, tissue-specific inflammation and systemic insulin resistance in WT mice but these diet-induced metabolic alterations were not exacerbated in PHD1-/- mice. In conclusion, PHD1 deficiency promotes hepatic steatosis and liver-specific insulin resistance but does not worsen the deleterious effects of HFD on metabolic homeostasis.


Subject(s)
Fatty Liver/genetics , Fatty Liver/metabolism , Insulin Resistance/genetics , Procollagen-Proline Dioxygenase/deficiency , Adiposity , Animals , Body Weight , Diet, High-Fat , Disease Models, Animal , Dyslipidemias/metabolism , Energy Metabolism , Fatty Liver/pathology , Glucose Intolerance/metabolism , Homeostasis , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Organ Specificity/genetics
7.
Bone ; 85: 15-22, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26808442

ABSTRACT

Lack of prolyl 3-hydroxylase 1 (P3H1) due to mutations in P3H1 results in severe forms of recessive osteogenesis imperfecta. In the present study, we investigated the bone tissue characteristics of P3H1 null mice. Histomorphometric analyses of cancellous bone in the proximal tibia and lumbar vertebra in 1-month and 3-month old mice demonstrated that P3H1 deficient mice had low trabecular bone volume and low mineral apposition rate, but normal osteoid maturation time and normal osteoblast and osteoclast surfaces. Quantitative backscattered electron imaging revealed that the bone mineralization density distribution was shifted towards higher values, indicating hypermineralization of bone matrix. It thus appears that P3H1 deficiency leads to decreased deposition of extracellular matrix by osteoblasts and increased incorporation of mineral into the matrix.


Subject(s)
Bone Matrix/physiology , Calcification, Physiologic/physiology , Procollagen-Proline Dioxygenase/deficiency , Analysis of Variance , Animals , Bone Density , Bone and Bones/metabolism , Bone and Bones/pathology , Genotype , Mice , Procollagen-Proline Dioxygenase/metabolism
8.
Cell Metab ; 23(2): 280-91, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26774962

ABSTRACT

The oxygen-sensing prolyl hydroxylase domain proteins (PHDs) regulate cellular metabolism, but their role in neuronal metabolism during stroke is unknown. Here we report that PHD1 deficiency provides neuroprotection in a murine model of permanent brain ischemia. This was not due to an increased collateral vessel network. Instead, PHD1(-/-) neurons were protected against oxygen-nutrient deprivation by reprogramming glucose metabolism. Indeed, PHD1(-/-) neurons enhanced glucose flux through the oxidative pentose phosphate pathway by diverting glucose away from glycolysis. As a result, PHD1(-/-) neurons increased their redox buffering capacity to scavenge oxygen radicals in ischemia. Intracerebroventricular injection of PHD1-antisense oligonucleotides reduced the cerebral infarct size and neurological deficits following stroke. These data identify PHD1 as a regulator of neuronal metabolism and a potential therapeutic target in ischemic stroke.


Subject(s)
Brain Ischemia/prevention & control , Cellular Reprogramming , Gene Deletion , Neurons/metabolism , Oxygen/metabolism , Procollagen-Proline Dioxygenase/metabolism , Stroke/prevention & control , Animals , Brain/blood supply , Brain/drug effects , Brain/pathology , Brain Ischemia/complications , Carbon/metabolism , Cellular Reprogramming/drug effects , Free Radical Scavengers/metabolism , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Injections, Intraventricular , Mice, Knockout , Neurons/drug effects , Neuroprotection/drug effects , Oligonucleotides/administration & dosage , Oligonucleotides/pharmacology , Oxidation-Reduction/drug effects , Pentose Phosphate Pathway/drug effects , Phenotype , Procollagen-Proline Dioxygenase/deficiency , Reactive Oxygen Species/metabolism , Stroke/complications
9.
Angiogenesis ; 19(2): 119-31, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26678600

ABSTRACT

In the adult, new vessels and red blood cells form in response to hypoxia. Here, the oxygen-sensing system (PHD-HIF) has recently been put into focus, since the prolyl-hydroxylase domain proteins (PHD) and hypoxia-inducible factors (HIF) are considered as potential therapeutic targets to treat ischemia, cancers or age-related macula degeneration. While the oxygen-sensing system (PHD-HIF) has been studied intensively in this respect, only little is known from developing vertebrate embryos since mutations within this pathway led to an early decease of embryos due to placental defects. During vertebrate embryogenesis, a progenitor cell called hemangioblast is assumed to give rise to blood cells and blood vessels in a process called hematopoiesis and vasculogenesis, respectively. Xenopus provides an ideal experimental system to address these processes in vivo, as its development does not depend on a functional placenta and thus allows analyzing the role of oxygen directly. To this end, we adopted a computer-controlled four-channel system, which allowed us to culture Xenopus embryos under defined oxygen concentrations. Our data show that the development of vascular structures and blood cells is strongly impaired under hypoxia, while general development is less compromised. Interestingly, suppression of Phd2 function using specific antisense morpholinos or a chemical inhibitor resulted in mostly overlapping vascular defects; nevertheless, blood cell was formed almost normally. Our results provide the first evidence that oxygen via Phd2 has a decisive influence on the formation of the vascular network during vertebrate embryogenesis. These findings may be considered in certain potential treatment concepts.


Subject(s)
Blood Vessels/embryology , Embryonic Development , Hypoxia/pathology , Neovascularization, Physiologic , Procollagen-Proline Dioxygenase/deficiency , Prolyl Hydroxylases/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/physiology , Animals , Blood Cells/metabolism , Cell Differentiation , Cell Lineage , Chronic Disease , Hematopoiesis , Hypoxia/embryology , Procollagen-Proline Dioxygenase/metabolism
10.
J Clin Invest ; 125(7): 2759-71, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26075818

ABSTRACT

Ischemic heart disease is the leading cause of heart failure. Both clinical trials and experimental animal studies demonstrate that chronic hypoxia can induce contractile dysfunction even before substantial ventricular damage, implicating a direct role of oxygen in the regulation of cardiac contractile function. Prolyl hydroxylase domain (PHD) proteins are well recognized as oxygen sensors and mediate a wide variety of cellular events by hydroxylating a growing list of protein substrates. Both PHD2 and PHD3 are highly expressed in the heart, yet their functional roles in modulating contractile function remain incompletely understood. Here, we report that combined deletion of Phd2 and Phd3 dramatically decreased expression of phospholamban (PLN), resulted in sustained activation of calcium/calmodulin-activated kinase II (CaMKII), and sensitized mice to chronic ß-adrenergic stress-induced myocardial injury. We have provided evidence that thyroid hormone receptor-α (TR-α), a transcriptional regulator of PLN, interacts with PHD2 and PHD3 and is hydroxylated at 2 proline residues. Inhibition of PHDs increased the interaction between TR-α and nuclear receptor corepressor 2 (NCOR2) and suppressed Pln transcription. Together, these observations provide mechanistic insight into how oxygen directly modulates cardiac contractility and suggest that cardiac function could be modulated therapeutically by tuning PHD enzymatic activity.


Subject(s)
Calcium-Binding Proteins/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Myocardium/metabolism , Procollagen-Proline Dioxygenase/metabolism , Receptors, Adrenergic, beta/metabolism , Animals , Calcium Signaling , Calcium-Binding Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Female , Humans , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Myocardial Contraction/physiology , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nuclear Receptor Co-Repressor 2/metabolism , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/genetics , Rats , Stress, Physiological , Thyroid Hormone Receptors alpha/metabolism
11.
J Biol Chem ; 290(27): 16964-78, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-26001784

ABSTRACT

Collagen prolyl 4-hydroxylases (C-P4H-I, C-P4H-II, and C-P4H-III) catalyze formation of 4-hydroxyproline residues required to form triple-helical collagen molecules. Vertebrate C-P4Hs are α2ß2 tetramers differing in their catalytic α subunits. C-P4H-I is the major isoenzyme in most cells, and inactivation of its catalytic subunit (P4ha1(-/-)) leads to embryonic lethality in mouse, whereas P4ha1(+/-) mice have no abnormalities. To study the role of C-P4H-II, which predominates in chondrocytes, we generated P4ha2(-/-) mice. Surprisingly, they had no apparent phenotypic abnormalities. To assess possible functional complementarity, we established P4ha1(+/-);P4ha2(-/-) mice. They were smaller than their littermates, had moderate chondrodysplasia, and developed kyphosis. A transient inner cell death phenotype was detected in their developing growth plates. The columnar arrangement of proliferative chondrocytes was impaired, the amount of 4-hydroxyproline and the Tm of collagen II were reduced, and the extracellular matrix was softer in the growth plates of newborn P4ha1(+/-);P4ha2(-/-) mice. No signs of uncompensated ER stress were detected in the mutant growth plate chondrocytes. Some of these defects were also found in P4ha2(-/-) mice, although in a much milder form. Our data show that C-P4H-I can to a large extent compensate for the lack of C-P4H-II in proper endochondral bone development, but their combined partial and complete inactivation, respectively, leads to biomechanically impaired extracellular matrix, moderate chondrodysplasia, and kyphosis. Our mouse data suggest that inactivating mutations in human P4HA2 are not likely to lead to skeletal disorders, and a simultaneous decrease in P4HA1 function would most probably be required to generate such a disease phenotype.


Subject(s)
Chondrocytes/enzymology , Extracellular Matrix/metabolism , Osteochondrodysplasias/enzymology , Procollagen-Proline Dioxygenase/deficiency , Animals , Apoptosis , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Collagen/biosynthesis , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Knockout , Osteochondrodysplasias/embryology , Osteochondrodysplasias/genetics , Osteochondrodysplasias/metabolism , Osteochondrodysplasias/physiopathology , Procollagen-Proline Dioxygenase/genetics
12.
Microvasc Res ; 97: 181-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25446011

ABSTRACT

BACKGROUND: There is an emerging focus on investigating innovative therapeutic molecules that can potentially augment neovascularization in order to treat peripheral arterial disease (PAD). Although prolyl hydroxylase domain proteins 1 and 3 (PHD1 and PHD3) may modulate angiogenesis via regulation of hypoxia inducible factor-1α (HIF-1α), there has been no study directly addressing their roles in ischemia-induced vascular growth. We hypothesize that PHD1(-/-) or PHD3(-/-) deficiency might promote angiogenesis in the murine hind-limb ischemia (HLI) model. STUDY DESIGN: Wild type (WT), PHD1(-/-) and PHD3(-/-) male mice aged 8-12weeks underwent right femoral artery ligation. Post-procedurally, motor function assessment and laser Doppler imaging were periodically performed. The mice were euthanized after 28days and muscles were harvested. Immunohistochemical analysis was performed to determine the extent of angiogenesis by measuring capillary and arteriolar density. VEGF expression was quantified by enzyme-linked immunosorbent assay (ELISA). Bcl-2 and HIF-1α were analyzed by immunofluorescence. Fibrosis was measured by picrosirius red staining. RESULTS: PHD1(-/-) and PHD3(-/-) mice showed significantly improved recovery of perfusion and motor function score when compared to WT after femoral artery ligation. These mice also exhibited increased capillary and arteriolar density, capillary/myocyte ratio along with decreased fibrosis compared to WT. VEGF, Bcl-2 and HIF-1α expression increased in PHD1(-/-) and PHD3(-/-) mice compared to WT. CONCLUSIONS: Taken together these results suggest that PHD1 and PHD3 deletions promote angiogenesis in ischemia-injured tissue, and may present a promising therapeutic strategy in treating PAD.


Subject(s)
Gene Deletion , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Neovascularization, Physiologic , Procollagen-Proline Dioxygenase/deficiency , Animals , Disease Models, Animal , Fibrosis , Hindlimb , Ischemia/genetics , Ischemia/pathology , Ischemia/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Muscle, Skeletal/pathology , Procollagen-Proline Dioxygenase/genetics , Protein Stability , Proto-Oncogene Proteins c-bcl-2/metabolism , Recovery of Function , Regional Blood Flow , Time Factors , Vascular Endothelial Growth Factor A/metabolism
13.
Nat Commun ; 5: 5582, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25420773

ABSTRACT

Solid tumours are exposed to microenvironmental factors such as hypoxia that normally inhibit cell growth. However, tumour cells are capable of counteracting these signals through mechanisms that are largely unknown. Here we show that the prolyl hydroxylase PHD3 restrains tumour growth in response to microenvironmental cues through the control of EGFR. PHD3 silencing in human gliomas or genetic deletion in a murine high-grade astrocytoma model markedly promotes tumour growth and the ability of tumours to continue growing under unfavourable conditions. The growth-suppressive function of PHD3 is independent of the established PHD3 targets HIF and NF-κB and its hydroxylase activity. Instead, loss of PHD3 results in hyperphosphorylation of epidermal growth factor receptor (EGFR). Importantly, epigenetic/genetic silencing of PHD3 preferentially occurs in gliomas without EGFR amplification. Our findings reveal that PHD3 inactivation provides an alternative route of EGFR activation through which tumour cells sustain proliferative signalling even under conditions of limited oxygen availability.


Subject(s)
Cell Proliferation , ErbB Receptors/metabolism , Glioblastoma/physiopathology , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia/physiopathology , Procollagen-Proline Dioxygenase/genetics , Animals , Apoptosis , Cell Line, Tumor , ErbB Receptors/genetics , Female , Gene Knockout Techniques , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Hypoxia/enzymology , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Male , Mice, Knockout , Oxygen/metabolism , Procollagen-Proline Dioxygenase/deficiency
14.
Biochem Biophys Res Commun ; 451(2): 288-94, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-25088999

ABSTRACT

Cells are frequently exposed to hypoxia in physiological and pathophysiological conditions in organisms. Control of energy metabolism is one of the critical functions of the hypoxic response. Hypoxia-Inducible Factor (HIF) is a central transcription factor that regulates the hypoxic response. HIF prolyl-hydroxylase PHDs are the enzymes that hydroxylate the α subunit of HIF and negatively regulate its expression. To further understand the physiological role of PHD3, proteomics were used to identify PHD3-interacting proteins, and pyruvate dehydrogenase (PDH)-E1ß was identified as such a protein. PDH catalyzes the conversion of pyruvate to acetyl-coA, thus playing a key role in cellular energy metabolism. PDH activity was significantly decreased in PHD3-depleted MCF7 breast cancer cells and PHD3(-/-) MEFs. PHD3 depletion did not affect the expression of the PDH-E1α, E1ß, and E2 subunits, or the phosphorylation status of E1α, but destabilized the PDH complex (PDC), resulting in less functional PDC. Finally, PHD3(-/-) cells were resistant to cell death in prolonged hypoxia with decreased production of ROS. Taken together, the study reveals that PHD3 regulates PDH activity in cells by physically interacting with PDC.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Procollagen-Proline Dioxygenase/metabolism , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Animals , Cell Death , Cell Hypoxia , Cells, Cultured , Enzyme Stability , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , MCF-7 Cells , Mice , Mice, Knockout , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/genetics , Protein Subunits , Pyruvate Dehydrogenase (Lipoamide)/chemistry , Pyruvate Dehydrogenase Complex/chemistry , RNA, Small Interfering/genetics
15.
J Leukoc Biol ; 96(3): 365-75, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24626957

ABSTRACT

On a molecular level, cells sense changes in oxygen availability through the PHDs, which regulate the protein stability of the α-subunit of the transcription factor HIF. Especially, PHD3 has been additionally associated with apoptotic cell death. We hypothesized that PHD3 plays a role in cell-fate decisions in macrophages. Therefore, myeloid-specific PHD3(-/-) mice were created and analyzed. PHD3(-/-) BMDM showed no altered HIF-1α or HIF-2α stabilization or increased HIF target gene expression in normoxia or hypoxia. Macrophage M1 and M2 polarization was unchanged likewise. Compared with macrophages from WT littermates, PHD3(-/-) BMDM exhibited a significant reduction in TUNEL-positive cells after serum withdrawal or treatment with stauro and SNAP. Under the same conditions, PHD3(-/-) BMDM also showed less Annexin V staining, which is representative for membrane disruption, and indicated a reduced early apoptosis. In an unbiased transcriptome screen, we found that Angptl2 expression was reduced in PHD3(-/-) BMDM under stress conditions. Addition of rAngptl2 rescued the antiapoptotic phenotype, demonstrating that it is involved in the PHD3-mediated response toward apoptotic stimuli in macrophages.


Subject(s)
Macrophages/cytology , Procollagen-Proline Dioxygenase/physiology , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Angiopoietins/biosynthesis , Angiopoietins/genetics , Angiopoietins/pharmacology , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/physiology , Bone Marrow Cells/cytology , Cell Hypoxia , Cells, Cultured , Gene Expression Regulation , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Macrophages/drug effects , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/enzymology , NF-kappa B/metabolism , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/genetics , Protein Processing, Post-Translational , Recombinant Fusion Proteins/pharmacology , S-Nitroso-N-Acetylpenicillamine/pharmacology , Staurosporine/pharmacology , Transcription, Genetic , Transcriptome
16.
Circulation ; 127(21): 2078-87, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23630130

ABSTRACT

BACKGROUND: Recent studies suggest that the oxygen-sensing pathway consisting of transcription factor hypoxia-inducible factor and prolyl hydroxylase domain proteins (PHDs) plays a critical role in glucose metabolism. However, the role of adipocyte PHD in the development of obesity has not been clarified. We examined whether deletion of PHD2, the main oxygen sensor, in adipocytes affects diet-induced obesity and associated metabolic abnormalities. METHODS AND RESULTS: To delete PHD2 in adipocyte, PHD2-floxed mice were crossed with aP2-Cre transgenic mice (Phd2(f/f)/aP2-Cre). Phd2(f/f)/aP2-Cre mice were resistant to high-fat diet-induced obesity (36.7±1.7 versus 44.3±2.0 g in control; P<0.01) and showed better glucose tolerance and homeostasis model assessment-insulin resistance index than control mice (3.6±1.0 versus 11.1±2.1; P<0.01). The weight of white adipose tissue was lighter (epididymal fat, 758±35 versus 1208±507 mg in control; P<0.01) with a reduction in adipocyte size. Macrophage infiltration into white adipose tissue was also alleviated in Phd2(f/f)/aP2-Cre mice. Target genes of hypoxia-inducible factor, including glycolytic enzymes and adiponectin, were upregulated in adipocytes of Phd2(f/f)/aP2-Cre mice. Lipid content was decreased and uncoupling protein-1 expression was increased in brown adipose tissue of Phd2(f/f)/aP2-Cre mice. Knockdown of PHD2 in 3T3L1 adipocytes induced a decrease in the glucose level and an increase in the lactate level in the supernatant with upregulation of glycolytic enzymes and reduced lipid accumulation. CONCLUSIONS: PHD2 in adipose tissue plays a critical role in the development of diet-induced obesity and glucose intolerance. PHD2 might be a novel target molecule for the treatment of obesity and associated metabolic abnormalities.


Subject(s)
Diet, High-Fat/adverse effects , Glucose Intolerance/etiology , Glucose Intolerance/physiopathology , Obesity/etiology , Obesity/physiopathology , Procollagen-Proline Dioxygenase/physiology , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue, White/blood supply , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Cells, Cultured , Disease Models, Animal , Glucose/metabolism , Glucose Intolerance/pathology , Glucose Transporter Type 4/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases , Lipid Metabolism/physiology , Macrophages/pathology , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/metabolism , Neovascularization, Physiologic/physiology , Obesity/pathology , Oxygen Consumption/physiology , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/genetics
17.
Arthritis Rheum ; 65(5): 1302-12, 2013 May.
Article in English | MEDLINE | ID: mdl-23334958

ABSTRACT

OBJECTIVE: To determine the effects of hypoxia on both anabolic and catabolic pathways of metabolism in human articular cartilage and to elucidate the roles played by hypoxia-inducible factors (HIFs) in these responses. METHODS: Normal human articular cartilage from a range of donors was obtained at the time of above-the-knee amputations due to sarcomas not involving the joint space. Fresh cartilage tissue explants and isolated cells were subjected to hypoxia and treatment with interleukin-1α. Cell transfections were performed on isolated human chondrocytes. RESULTS: Using chromatin immunoprecipitation, we found that hypoxia induced cartilage production in human tissue explants through direct binding of HIF-2α to a specific site in the master-regulator gene SOX9. Importantly, hypoxia also suppressed spontaneous and induced destruction of human cartilage in explant culture. We found that anticatabolic responses were predominantly mediated by HIF-1α. Manipulation of the hypoxia-sensing pathway through depletion of HIF-targeting prolyl hydroxylase-containing protein 2 (PHD-2) further enhanced cartilage responses as compared to hypoxia alone. Hypoxic regulation of tissue-specific metabolism similar to that in human cartilage was observed in pig, but not mouse, cartilage. CONCLUSION: We found that resident chondrocytes in human cartilage are exquisitely adapted to hypoxia and use it to regulate tissue-specific metabolism. Our data revealed that while fundamental regulators, such as SOX9, are key molecules both in mice and humans, the way in which they are controlled can differ. This is all the more important since it is upstream regulators such as this that need to be directly targeted for therapeutic benefit. HIF-specific hydroxylase PHD-2 may represent a relevant target for cartilage repair.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Adaptation, Physiological/physiology , Adolescent , Adult , Animals , Binding Sites , Cartilage, Articular/pathology , Cell Hypoxia/physiology , Cells, Cultured , Child , Chondrocytes/pathology , Female , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , Interleukin-1alpha/pharmacology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Procollagen-Proline Dioxygenase/deficiency , SOX9 Transcription Factor/metabolism , Swine , Transfection , Young Adult
18.
Matrix Biol ; 32(1): 39-44, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23186870

ABSTRACT

Prolyl 3-hydroxylase1 (P3H1) is a collagen modifying enzyme which hydroxylates certain prolines in the Xaa position of conventional GlyXaaYaa triple helical sequence. Recent investigations have revealed that mutations in the LEPRE1 (gene encoding for P3H1) cause severe osteogenesis imperfecta (OI) in humans. Similarly LEPRE1 knockout mice display an OI-like phenotype. Significant hearing loss is a common problem for people with osteogenesis imperfecta. Here we report that hearing of the P3H1 null mice is substantially affected. Auditory brainstem responses (ABRs) of the P3H1 null mice show an average increase of 20-30 dB in auditory thresholds. Three dimensional reconstructions of the mutant middle ear bones by Micro-scale X-ray computed tomography (Micro-CT) demonstrate abnormal morphology of the incudostapedial and incudomalleal joints. We establish the LEPRE1 knockout mouse as a valuable model system to investigate the mechanism of hearing loss in recessive OI.


Subject(s)
Ear Ossicles/abnormalities , Hearing Loss/genetics , Joints/abnormalities , Membrane Glycoproteins/genetics , Osteogenesis Imperfecta/genetics , Procollagen-Proline Dioxygenase/deficiency , Proteoglycans/genetics , Animals , Auditory Threshold , Evoked Potentials, Auditory, Brain Stem/physiology , Genes, Recessive/genetics , Hearing Loss/enzymology , Mice , Mice, Knockout , Osteogenesis Imperfecta/enzymology , X-Ray Microtomography
19.
Langenbecks Arch Surg ; 397(8): 1313-22, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22961008

ABSTRACT

PURPOSE: Liver regeneration after partial hepatectomy (PH) occurs in conditions of reduced oxygen supply. HIF prolyl hydroxylase enzymes (PHD1, PHD2, and PHD3) are oxygen sensors involved in adaptive response to hypoxia. Specific functions of these PHD enzymes in liver regeneration have, however, remained enigmatic. Here, we investigated the significance of PHD1 in liver regeneration following hepatectomy. METHODS: Liver regeneration was studied in PHD1-deficient (PHD1(-/-)) and wild type (WT) mice subjected to 80% hepatectomy. For in vitro analyses, hepatocytes were isolated from PHD1(-/-) and WT livers. Cell cycle progression was studied via FACS-based analysis of nuclear DNA profile. Transcription factor binding assays, qRT-PCR, and immunoblotting were applied to study the relevance of PHD1 downstream effectors during liver regeneration. RESULTS: Liver regeneration was significantly enhanced in PHD1(-/-) mice compared to WT littermates. This effect was due to enhanced proliferation rather than to hypertrophy of liver cells. Cell cycle progression was significantly enhanced, and transcriptional activity of the cell cycle regulator c-Myc was increased in PHD1-deficient hepatocytes. These changes coincided with increased expression of cyclin D2, a cell cycle-promoting c-Myc target, and decreased expression of the cell cycle-delaying c-Myc target p21. CONCLUSIONS: Loss of PHD1 enhances liver regeneration by boosting hepatocyte proliferation in a c-Myc-dependent fashion. PHD1 might, therefore, represent a potential target to facilitate liver regeneration after surgical resection.


Subject(s)
Hepatectomy , Hepatocytes/metabolism , Liver Regeneration/physiology , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cell Cycle , Cell Proliferation , Cells, Cultured , Hepatocytes/cytology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-myc/metabolism , Real-Time Polymerase Chain Reaction
20.
Stroke ; 43(10): 2748-56, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22933585

ABSTRACT

BACKGROUND AND PURPOSE: Numerous factors involved in the adaptive response to hypoxia, including erythropoietin and vascular endothelial growth factor are transcriptionally regulated by hypoxia-inducible factors (HIFs). During normoxia, prolyl-4-hydroxylase domain (PHD) proteins hydroxylate HIF-α subunits, resulting in their degradation. We investigated the effect of neuronal deletion of PHD2, the most abundant isoform in brain, for stroke outcome. METHODS: We generated neuron-specific Phd2 knockout mice and subjected animals to systemic hypoxia or transient middle cerebral artery occlusion. Infarct volume and cell death were determined by histology. HIF-1α, HIF-2α, and HIF target genes were analyzed by immunoblotting and real-time polymerase chain reaction, respectively. RESULTS: Neuron-specific ablation of Phd2 significantly increased protein stability of HIF-1α and HIF-2α in the forebrain and enhanced expression of the neuroprotective HIF target genes erythropoietin and vascular endothelial growth factor as well as glucose transporter and glycolysis-related enzymes under hypoxic and ischemic conditions. Mice with Phd2-deficient neurons subjected to transient cerebral ischemia exhibited a strong reduction in infarct size, and cell death of hippocampal CA1 neurons located in the peri-infarct region was dramatically reduced in these mice. Vessel density in forebrain subregions, except for caudate-putamen, was not altered in Phd2-deficient animals. CONCLUSIONS: Our findings denote that the endogenous adaptive response on hypoxic-ischemic insults in the brain is at least partly dependent on the activity of HIFs and identify PHD2 as the key regulator for the protective hypoxia response. The results suggest that specific inhibition of PHD2 may provide a useful therapeutic strategy to protect brain tissue from ischemic injury.


Subject(s)
Brain Injuries/prevention & control , Ischemic Attack, Transient/complications , Neurons/metabolism , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Injuries/metabolism , Brain Injuries/pathology , Cerebral Infarction/pathology , Female , Hypoxia/complications , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases , Infarction, Middle Cerebral Artery/complications , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Male , Mice , Mice, Knockout , Models, Animal , Procollagen-Proline Dioxygenase/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...