Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Mol Immunol ; 149: 48-58, 2022 09.
Article in English | MEDLINE | ID: mdl-35724581

ABSTRACT

The key players of the hypoxic response are the hypoxia-inducible factors (Hif), whose α-subunits are tightly regulated by Prolyl-4-hydroxylases (PHD), predominantly by PHD2. Monocytes/Macrophages are involved in atherosclerosis but also restenosis and were found at hypoxic and sites of the lesion. Little is known about the role of the myeloid PHD2 in atherosclerosis and neointima formation. The study aimed to investigate the consequences of a myeloid deficiency of PHD2 in the process of neointima formation using an arterial denudation model. LysM-cre mice were crossed with PHD2fl/fl, PHD2fl/fl/Hif1αfl/fl and PHD2fl/fl/Hif2αfl/fl to get myeloid specific knockout of PHD2 and the Hif-α subunits. Denudation of the femoral artery was performed and animals were fed a western type diet afterwards with analysis of neointima formation 5 and 35 days after denudation. Increased neointima formation in myeloid PHD2 knockouts was observed, which was blunted by double-knockout of PHD2 and Hif1α whereas double knockout of PHD2 and Hif-2α showed comparable lesions to the PHD2 knockouts. Macrophage infiltration was comparable to the neointima formation, suggesting a more inflammatory reaction, and was accompanied by increased intimal VEGF-A expression. Collagen-content inversely correlated to the extent of neointima formation suggesting a destabilization of the plaque. This effect might be triggered by macrophage polarization. Therefore, in vitro results showed a distinct expression pattern in differentially polarized macrophages with high expression of Hif-1α, VEGF and MMP-1 in proinflammatory M1 macrophages. In conclusion, the results show that myeloid Hif-1α is involved in neointima hyperplasia. Our in vivo and in vitro data reveal a central role for this transcription factor in driving plaque-vascularization accompanied by matrix-degradation leading to plaque destabilization.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Femoral Artery , Hypoxia-Inducible Factor-Proline Dioxygenases , Macrophages , Neointima , Plaque, Atherosclerotic , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Femoral Artery/injuries , Femoral Artery/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Macrophages/metabolism , Mice , Neointima/genetics , Neointima/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , Procollagen-Proline Dioxygenase/genetics
2.
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
3.
Cell Metab ; 32(2): 215-228.e7, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32663458

ABSTRACT

Rapid alterations in cellular metabolism allow tissues to maintain homeostasis during changes in energy availability. The central metabolic regulator acetyl-CoA carboxylase 2 (ACC2) is robustly phosphorylated during cellular energy stress by AMP-activated protein kinase (AMPK) to relieve its suppression of fat oxidation. While ACC2 can also be hydroxylated by prolyl hydroxylase 3 (PHD3), the physiological consequence thereof is poorly understood. We find that ACC2 phosphorylation and hydroxylation occur in an inverse fashion. ACC2 hydroxylation occurs in conditions of high energy and represses fatty acid oxidation. PHD3-null mice demonstrate loss of ACC2 hydroxylation in heart and skeletal muscle and display elevated fatty acid oxidation. Whole body or skeletal muscle-specific PHD3 loss enhances exercise capacity during an endurance exercise challenge. In sum, these data identify an unexpected link between AMPK and PHD3, and a role for PHD3 in acute exercise endurance capacity and skeletal muscle metabolism.


Subject(s)
Fats/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Muscle, Skeletal/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Cell Line , Exercise Tolerance , Female , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidation-Reduction
4.
PLoS One ; 15(5): e0233261, 2020.
Article in English | MEDLINE | ID: mdl-32413092

ABSTRACT

The transcription factor Hypoxia-inducible factor 1 (HIF-1) plays a pivotal role in tissue regeneration. HIF-1 is negatively controlled by O2-dependent prolyl hydroxylases with a predominant role of prolyl hydroxylase 2 isoform (Phd2). Transgenic mice, hypomorphic for this isoform, accumulate more HIF-1 under normoxic conditions. Using these mice, we investigated the influence of Phd2 and HIF-1 on the regenerative capability of skeletal muscle tissue after myotrauma. Phd2-hypomorphic and wild type mice (on C57Bl/6 background) were grouped with regeneration times from 6 to 168 hours after closed mechanic muscle trauma to the hind limb. Tissue samples were analysed by immuno-staining and real-time PCR. Bone marrow derived macrophages of wild type and Phd2-hypomorphic mice were isolated and analysed via flow cytometry and quantitative real-time PCR. Phd2 reduction led to a higher regenerative capability due to enhanced activation of myogenic factors accompanied by induction of genes responsible for glucose and lactate metabolism in Phd2-hypomorphic mice. Macrophage infiltration into the trauma areas in hypomorphic mice started earlier and was more pronounced compared to wild type mice. Phd2-hypomorphic mice also showed higher numbers of macrophages in areas with sustained trauma 72 hours after myotrauma application. In conclusion, we postulate that the HIF-1 pathway is activated secondary to a Phd2 reduction which may lead to i) higher activation of myogenic factors, ii) increased number of positive stem cell proliferation markers, and iii) accelerated macrophage recruitment to areas of trauma, resulting in faster muscle tissue regeneration after myotrauma. With the current development of prolyl hydroxylase domain inhibitors, our findings point towards a potential clinical benefit after myotrauma.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Muscle, Skeletal/physiology , Regeneration/physiology , Soft Tissue Injuries/physiopathology , Animals , Cell Proliferation/physiology , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Random Allocation , Vascular Endothelial Growth Factor A/metabolism
6.
Nature ; 565(7740): 511-515, 2019 01.
Article in English | MEDLINE | ID: mdl-30651640

ABSTRACT

Endochondral ossification, an important process in vertebrate bone formation, is highly dependent on correct functioning of growth plate chondrocytes1. Proliferation of these cells determines longitudinal bone growth and the matrix deposited provides a scaffold for future bone formation. However, these two energy-dependent anabolic processes occur in an avascular environment1,2. In addition, the centre of the expanding growth plate becomes hypoxic, and local activation of the hypoxia-inducible transcription factor HIF-1α is necessary for chondrocyte survival by unidentified cell-intrinsic mechanisms3-6. It is unknown whether there is a requirement for restriction of HIF-1α signalling in the other regions of the growth plate and whether chondrocyte metabolism controls cell function. Here we show that prolonged HIF-1α signalling in chondrocytes leads to skeletal dysplasia by interfering with cellular bioenergetics and biosynthesis. Decreased glucose oxidation results in an energy deficit, which limits proliferation, activates the unfolded protein response and reduces collagen synthesis. However, enhanced glutamine flux increases α-ketoglutarate levels, which in turn increases proline and lysine hydroxylation on collagen. This metabolically regulated collagen modification renders the cartilaginous matrix more resistant to protease-mediated degradation and thereby increases bone mass. Thus, inappropriate HIF-1α signalling results in skeletal dysplasia caused by collagen overmodification, an effect that may also contribute to other diseases involving the extracellular matrix such as cancer and fibrosis.


Subject(s)
Bone Diseases/metabolism , Bone Diseases/pathology , Chondrocytes/metabolism , Collagen/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Animals , Cartilage/metabolism , Extracellular Matrix/metabolism , Glucose/metabolism , Glutamine/metabolism , Growth Plate/metabolism , Hydroxylation , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Ketoglutaric Acids/metabolism , Lysine/metabolism , Male , Mice , Osteogenesis , Oxidation-Reduction , Proline/metabolism
7.
J Biol Chem ; 294(10): 3760-3771, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30617181

ABSTRACT

Most clear cell renal cell carcinomas (ccRCCs) have inactivation of the von Hippel-Lindau tumor suppressor protein (pVHL), resulting in the accumulation of hypoxia-inducible factor α-subunits (HIF-α) and their downstream targets. HIF-2α expression is particularly high in ccRCC and is associated with increased ccRCC growth and aggressiveness. In the canonical HIF signaling pathway, HIF-prolyl hydroxylase 3 (PHD3) suppresses HIF-2α protein by post-translational hydroxylation under sufficient oxygen availability. Here, using immunoblotting and immunofluorescence staining, qRT-PCR, and siRNA-mediated gene silencing, we show that unlike in the canonical pathway, PHD3 silencing in ccRCC cells leads to down-regulation of HIF-2α protein and mRNA. Depletion of other PHD family members had no effect on HIF-2α expression, and PHD3 knockdown in non-RCC cells resulted in the expected increase in HIF-2α protein expression. Accordingly, PHD3 knockdown decreased HIF-2α target gene expression in ccRCC cells and expression was restored upon forced HIF-2α expression. The effect of PHD3 depletion was pinpointed to HIF2A mRNA stability. In line with these in vitro results, a strong positive correlation of PHD3 and HIF2A mRNA expression in ccRCC tumors was detected. Our results suggest that in contrast to the known negative regulation of HIF-2α in most cell types, high PHD3 expression in ccRCC cells maintains elevated HIF-2α expression and that of its target genes, which may enhance kidney cancer aggressiveness.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Renal Cell/pathology , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Kidney Neoplasms/pathology , Cell Line, Tumor , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Silencing , Glucose Transporter Type 1/genetics , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Protein Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Nat Commun ; 9(1): 5426, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30575721

ABSTRACT

Prolyl hydroxylase domain protein 2 (PHD2) is a well-known master oxygen sensor. However, the role of PHD2 in tumor initiation remains controversial. We find that during the transition of human nevi to melanoma, the expression of PHD2 protein is significantly decreased and lower expression PHD2 in melanoma is associated with worse clinical outcome. Knockdown of PHD2 leads to elevated Akt phosphorylation in human melanocytes. Mice with conditional melanocyte-specific expression of Phd2lox/lox (Tyr::CreER;Phd2lox/lox) fail to develop pigmented lesions. However, deletion of Phd2 in combination with expression of BRafV600E in melanocytes (Tyr::CreER;Phd2lox/lox;BRafCA) leads to the development of melanoma with 100% penetrance and frequent lymph node metastasis. Analysis of tumor tissues using reverse phase protein arrays demonstrates that Phd2 deletion activates the AKT-mTOR-S6 signaling axis in the recovered tumors. These data indicate that PHD2 is capable of suppressing tumor initiation largely mediated through inhibiting of the Akt-mTOR signaling pathway in the melanocyte lineage.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Melanocytes/metabolism , Melanoma/etiology , Proto-Oncogene Proteins B-raf/genetics , Animals , Cell Line, Tumor , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Melanoma/metabolism , Mice, Transgenic , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
9.
Cell Physiol Biochem ; 48(3): 1003-1011, 2018.
Article in English | MEDLINE | ID: mdl-30036883

ABSTRACT

BACKGROUND/AIMS: Liver disease is a leading cause of high mortality and morbidity worldwide. The aim of the present study is to investigate the regulatory role of prolyl hydroxylase-2 (PHD2)-hypoxia-inducible factor-2a (HIF-2α) axis on nonalcoholic fatty liver disease (NAFLD) and to explore the potential mechanisms by which endothelial (EC)-specific PHD2 deficiency regulates hepatic steatosis and fibrosis. METHODS: In the endothelial-specific PHD2 knockout (PHD2ECKO) mouse fed with normal diet or high fat diet (HFD), liver lipid accumulation and fibrosis were measured by Oil Red O and Masson trichrome staining. The fat and body weight (FW/BW) ratio and glucose tolerance were measured. The expression of HIF-2α, atrial natriuretic peptide (ANP), angiopoietin-2 (Ang-2), and transforming growth factor-b (TGF-ß) were analyzed by western blot analysis. RESULTS: The steatosis and fibrosis were significantly increased in the PHD2ECKO mice. FW/BW ratio was significantly increased in the PHD2ECKO mice. Moreover, knockout of endothelial PHD2 resulted in an impairment of glucose tolerance in mice. Western blot analysis showed that the expression of HIF-2α in liver tissues was not significantly increased. Interestingly, the expression of ANP was decreased, and Ang-2 and TGF-ß levels were significantly increased in the liver of PHD2ECKO mice. The FW/BW ratio was also significantly increased in the PHD2ECKO mice fed with HFD for 16 weeks. Feeding HFD resulted in a significant increase in hepatic steatosis in the control PHD2f/f mice, but did not further enhance hepatic steatosis in the PHD2ECKO mice. CONCLUSIONS: We concluded that the endothelial PHD2 plays a critical role in hepatic steatosis and fibrosis, which may be involved in the regulation of ANP and Ang-2/TGF-ß signaling pathway, but not the HIF-2α expression.


Subject(s)
Endothelium/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Animals , Atrial Natriuretic Factor/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Diet, High-Fat , Glucose Tolerance Test , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/veterinary , Phosphofructokinase-2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribonuclease, Pancreatic/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
10.
Sci Rep ; 7(1): 17220, 2017 12 08.
Article in English | MEDLINE | ID: mdl-29222481

ABSTRACT

Hypoxia-inducible factor 1α (HIF1α) induces the expression of several hundred genes in hypoxia aiming at restoration of oxygen homeostasis. HIF prolyl-4-hydroxylases (HIF-P4Hs) regulate the stability of HIF1α in an oxygen-dependent manner. Hypoxia is a common feature in inflammation and cancer and the HIF pathway is closely linked with the inflammatory NF-κB and tumor suppressor p53 pathways. Here we show that genetic inactivation or chemical inhibition of HIF-P4H-1 leads to downregulation of proinflammatory genes, while proapoptotic genes are upregulated. HIF-P4H-1 inactivation reduces the inflammatory response under LPS stimulus in vitro and in an acute skin inflammation model in vivo. Furthermore, HIF-P4H-1 inactivation increases p53 activity and stability and hydroxylation of proline 142 in p53 has an important role in this regulation. Altogether, our data suggest that HIF-P4H-1 inhibition may be a promising therapeutic candidate for inflammatory diseases and cancer, enhancing the reciprocal negative regulation of the NF-κB and p53 pathways.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , NF-kappa B/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Caspase 3/metabolism , Cell Line , Down-Regulation , Gene Silencing , Humans , Hydroxylation , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Macrophages/cytology , Macrophages/metabolism , Mice , Proteolysis
11.
J Pathol ; 242(3): 322-333, 2017 07.
Article in English | MEDLINE | ID: mdl-28418093

ABSTRACT

Osteogenic-angiogenic coupling is promoted by the hypoxia-inducible factor 1-alpha (HIF-1α) transcription factor, provoking interest in HIF activation as a therapeutic strategy to improve osteoblast mineralization and treat pathological osteolysis. However, HIF also enhances the bone-resorbing activity of mature osteoclasts. It is therefore essential to determine the full effect(s) of HIF on both the formation and the bone-resorbing function of osteoclasts in order to understand how they might respond to such a strategy. Expression of HIF-1α mRNA and protein increased during osteoclast differentiation from CD14+ monocytic precursors, additionally inducing expression of the HIF-regulated glycolytic enzymes. However, HIF-1α siRNA only moderately affected osteoclast differentiation, accelerating fusion of precursor cells. HIF induction by inhibition of the regulatory prolyl-4-hydroxylase (PHD) enzymes reduced osteoclastogenesis, but was confirmed to enhance bone resorption by mature osteoclasts. Phd2+/- murine osteoclasts also exhibited enhanced bone resorption, associated with increased expression of resorption-associated Acp5, in comparison with wild-type cells from littermate controls. Phd3-/- bone marrow precursors displayed accelerated early fusion, mirroring results with HIF-1α siRNA. In vivo, Phd2+/- and Phd3-/- mice exhibited reduced trabecular bone mass, associated with reduced mineralization by Phd2+/- osteoblasts. These data indicate that HIF predominantly functions as a regulator of osteoclast-mediated bone resorption, with little effect on osteoclast differentiation. Inhibition of HIF might therefore represent an alternative strategy to treat diseases characterized by pathological levels of osteolysis. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Bone Resorption/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Osteoclasts/physiology , Osteogenesis/physiology , Prolyl Hydroxylases/physiology , Animals , Cancellous Bone/physiology , Cell Adhesion/physiology , Cell Differentiation/physiology , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Leukocytes, Mononuclear/pathology , Mice , RNA, Messenger/metabolism
12.
Trends Cardiovasc Med ; 27(4): 229-236, 2017 05.
Article in English | MEDLINE | ID: mdl-28089339

ABSTRACT

Pulmonary arterial hypertension (PAH) is a lung vascular disease characterized with a progressive increase of pulmonary vascular resistance and obliterative pulmonary vascular remodeling resulting in right heart failure and premature death. In this brief review, we document the recent advances in identifying genetically modified murine models of PH, with a focus on the recent discovery of the mouse model of Tie2 Cre-mediated deletion of prolyl hydroxylase 2, which exhibits progressive obliterative vascular remodeling, severe PAH, and right heart failure, thus recapitulating many of the features of clinical PAH. We will also discuss the translational potential of recent findings arising from experimental studies of murine PH models.


Subject(s)
Arterial Pressure , Hypertension, Pulmonary/enzymology , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Pulmonary Artery/enzymology , Vascular Remodeling , Animals , Antihypertensive Agents/therapeutic use , Arterial Pressure/drug effects , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , Disease Models, Animal , Disease Progression , Enzyme Activation , Enzyme Activators/therapeutic use , Genetic Predisposition to Disease , Heart Failure/enzymology , Heart Failure/physiopathology , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Integrases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Receptor, TIE-2/genetics , Severity of Illness Index , Vascular Remodeling/drug effects
13.
Exp Neurol ; 286: 93-106, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27720797

ABSTRACT

Episodes of cerebral hypoxia/ischemia increase the risk of dementia, which is associated with impaired learning and memory. Previous studies in rodent models of dementia indicated a favorable effect of the hypoxia-inducible factor (HIF) targets VEGF (vascular endothelial growth factor) and erythropoietin (Epo). In the present study we thus investigated whether activation of the entire adaptive HIF pathway in neurons by cell-specific deletion of the HIF suppressor prolyl-4-hydroxylase 2 (PHD2) improves cognitive abilities in young (3months) and old (18-28months) mice suffering from chronic brain hypoperfusion. Mice underwent permanent occlusion of the left common carotid artery, and cognitive function was assessed using the Morris water navigation task. Under conditions of both normal and decreased brain perfusion, neuronal PHD2 deficiency resulted in improved and faster spatial learning in young mice, which was preserved to some extent also in old animals. The loss of PHD2 in neurons resulted in enhanced hippocampal mRNA and protein levels of Epo and VEGF, but did not alter local microvascular density, dendritic spine morphology, or expression of synaptic plasticity-related genes in the hippocampus. Instead, better cognitive function in PHD2 deficient animals was accompanied by an increased number of neuronal precursor cells along the subgranular zone of the dentate gyrus. Overall, our current pre-clinical findings indicate an important role for the endogenous oxygen sensing machinery, encompassing PHDs, HIFs and HIF target genes, for proper cognitive function. Thus, pharmacological compounds affecting the PHD-HIF axis might well be suited to treat cognitive dysfunction and neurodegenerative processes.


Subject(s)
Cognition Disorders/etiology , Cognition Disorders/genetics , Gene Expression Regulation/genetics , Hypoxia, Brain/complications , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/pathology , Brain/ultrastructure , Cerebrovascular Circulation/genetics , Cerebrovascular Circulation/physiology , Disease Models, Animal , Escape Reaction/physiology , Hypoxia, Brain/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Locomotion/genetics , Locomotion/physiology , Male , Maze Learning/physiology , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Psychomotor Performance/physiology , Reaction Time/genetics , Reaction Time/physiology , Silver Staining , Statistics, Nonparametric
14.
J Bone Miner Res ; 31(10): 1877-1887, 2016 10.
Article in English | MEDLINE | ID: mdl-27082941

ABSTRACT

The main oxygen sensor hypoxia inducible factor (HIF) prolyl hydroxylase 2 (PHD2) is a critical regulator of tissue homeostasis during erythropoiesis, hematopoietic stem cell maintenance, and wound healing. Recent studies point toward a role for the PHD2-erythropoietin (EPO) axis in the modulation of bone remodeling, even though the studies produced conflicting results. Here, we used a number of mouse strains deficient of PHD2 in different cell types to address the role of PHD2 and its downstream targets HIF-1α and HIF-2α in bone remodeling. Mice deficient for PHD2 in several cell lineages, including EPO-producing cells, osteoblasts, and hematopoietic cells (CD68:cre-PHD2f/f ) displayed a severe reduction of bone density at the distal femur as well as the vertebral body due to impaired bone formation but not bone resorption. Importantly, using osteoblast-specific (Osx:cre-PHD2f/f ) and osteoclast-specific PHD2 knock-out mice (Vav:cre- PHD2f/f ), we show that this effect is independent of the loss of PHD2 in osteoblast and osteoclasts. Using different in vivo and in vitro approaches, we show here that this bone phenotype, including the suppression of bone formation, is directly linked to the stabilization of the α-subunit of HIF-2, and possibly to the subsequent moderate induction of serum EPO, which directly influenced the differentiation and mineralization of osteoblast progenitors resulting in lower bone density. Taken together, our data identify the PHD2:HIF-2α:EPO axis as a so far unknown regulator of osteohematology by controlling bone homeostasis. Further, these data suggest that patients treated with PHD inhibitors or EPO should be monitored with respect to their bone status. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Bone Resorption/metabolism , Erythropoietin/biosynthesis , Hematopoietic Stem Cells/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Osteoblasts/metabolism , Osteoclasts/metabolism , Animals , Bone Density/genetics , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Resorption/genetics , Bone Resorption/pathology , Erythropoietin/genetics , Hematopoietic Stem Cells/pathology , Mice , Mice, Knockout , Osteoblasts/pathology , Osteoclasts/pathology
15.
Neurobiol Dis ; 91: 221-35, 2016 07.
Article in English | MEDLINE | ID: mdl-27001147

ABSTRACT

Hypoxia inducible factors (HIFs) mediate the endogenous adaptive responses to hypoxia. HIF prolyl 4-hydroxylase domain proteins (PHD) are important suppressors of the HIF pathway. Recently, we demonstrated that neuron-specific deletion of Phd2 reduces cerebral tissue damage in the very acute phase of ischemic stroke. In the present study, we investigated whether neuronal Phd2 ablation is likewise beneficial for stroke recovery, and aimed to identify underlying cellular mechanisms. Mice underwent permanent occlusion of the distal middle cerebral artery (pdMCAO) for either 7days (sub-acute stage) or 30days (chronic stage). One week after pdMCAO the infarct size of Phd2-deficient mice was significantly reduced as compared to wild-type (WT) mice. Accordingly, Phd2-deficient animals showed less impaired sensorimotor function. Neuronal loss of Phd2 upregulated vascular endothelial growth factor (VEGF) and significantly increased microvascular density along the infarct border in the sub-acute stage of stroke. Phd2-deficient mice showed reduced expression of pro-inflammatory cytokines and increased numbers of resting microglia/macrophages and reactive astrocytes within peri-infarct regions in comparison to WT littermates. Finally, brain tissue protection and increased angiogenesis upon sub-acute ischemic stroke was completely absent in Phd2 knockout mice that were additionally deficient for both Hif1a and Hif2a. Our findings suggest that lack of PHD2 in neurons improves histological and functional long-term outcome from ischemic stroke at least partly by amplifying endogenous adaptive neovascularization through activation of the HIF-VEGF axis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Ischemia/enzymology , Brain Ischemia/physiopathology , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Recovery of Function/physiology , Stroke/enzymology , Stroke/physiopathology , Animals , Brain Ischemia/genetics , Disease Models, Animal , Hypoxia/pathology , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Male , Mice , Mice, Knockout , Neurons/metabolism , Stroke/genetics , Stroke/pathology
16.
Skelet Muscle ; 6: 5, 2016.
Article in English | MEDLINE | ID: mdl-26949511

ABSTRACT

BACKGROUND: Hypoxia exposure is known to induce an alteration in skeletal muscle fiber-type distribution mediated by hypoxia-inducible factor (HIF)-α. The downstream pathway of HIF-α leading to fiber-type shift, however, has not been elucidated. The calcineurin pathway is one of the pathways responsible for slow muscle fiber transition. Because calcineurin pathway is activated by vascular endothelial growth factor (VEGF), one of the factors induced by HIF-1α, we hypothesized that the stabilization of HIF-1α may lead to slow muscle fiber transition via the activation of calcineurin pathway in skeletal muscles. To induce HIF-1α stabilization, we used a loss of function strategy to abrogate Prolyl hydroxylase domain protein (PHD) 2 responsible for HIF-1α hydroxylation making HIF-1α susceptible to ubiquitin dependent degradation by proteasome. The purpose of this study was therefore to examine the effect of HIF-1α stabilization in PHD2 conditional knockout mouse on skeletal muscle fiber-type transition and to elucidate the involvement of calcineurin pathway on muscle fiber-type transition. RESULTS: PHD2 deficiency resulted in an increased capillary density in skeletal muscles due to the induction of vascular endothelial growth factor. It also elicited an alteration of skeletal muscle phenotype toward the type I fibers in both of the soleus (35.8 % in the control mice vs. 46.7 % in the PHD2-deficient mice, p < 0.01) and the gastrocnemius muscle (0.94 vs. 1.89 %, p < 0.01), and the increased proportion of type I fibers appeared to correspond to the area of increased capillary density. In addition, calcineurin and nuclear factor of activated T cell (NFATc1) protein levels were increased in both the gastrocnemius and soleus muscles, suggesting that the calcineurin/NFATc1 pathway was responsible for the type I fiber transition regardless of PGC-1α, which responded minimally to PHD2 deficiency. Indeed, we found that tacrolimus (FK-506), a calcineurin inhibitor, successfully suppressed slow fiber-type formation in PHD2-deficient mice. CONCLUSIONS: Taken together, stabilized HIF-1α induced by PHD2 conditional knockout resulted in the transition of muscle fibers toward a slow fiber type via a calcineurin/NFATc1 signaling pathway. PHD2 conditional knockout mice may serve as a model for chronic HIF-1α stabilization as in mice exposed to low oxygen concentration.


Subject(s)
Calcineurin/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Muscle Fibers, Slow-Twitch/enzymology , Muscle, Skeletal/enzymology , NFATC Transcription Factors/metabolism , Signal Transduction , Animals , Calcineurin Inhibitors/pharmacology , Capillaries/metabolism , Cell Hypoxia , Cell Line , Genotype , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Mice, Knockout , Muscle Fibers, Slow-Twitch/drug effects , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Neovascularization, Physiologic , Phenotype , Protein Stability , Signal Transduction/drug effects , Tacrolimus/pharmacology , Time Factors , Vascular Endothelial Growth Factor A/metabolism
17.
EMBO Rep ; 16(10): 1394-408, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26323721

ABSTRACT

Cancer-associated fibroblasts (CAFs) interact with tumour cells and promote growth and metastasis. Here, we show that CAF activation is reversible: chronic hypoxia deactivates CAFs, resulting in the loss of contractile force, reduced remodelling of the surrounding extracellular matrix and, ultimately, impaired CAF-mediated cancer cell invasion. Hypoxia inhibits prolyl hydroxylase domain protein 2 (PHD2), leading to hypoxia-inducible factor (HIF)-1α stabilisation, reduced expression of αSMA and periostin, and reduced myosin II activity. Loss of PHD2 in CAFs phenocopies the effects of hypoxia, which can be prevented by simultaneous depletion of HIF-1α. Treatment with the PHD inhibitor DMOG in an orthotopic breast cancer model significantly decreases spontaneous metastases to the lungs and liver, associated with decreased tumour stiffness and fibroblast activation. PHD2 depletion in CAFs co-injected with tumour cells similarly prevents CAF-induced metastasis to lungs and liver. Our data argue that reversion of CAFs towards a less active state is possible and could have important clinical implications.


Subject(s)
Cell Hypoxia , Fibroblasts/physiology , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mammary Neoplasms, Experimental/pathology , Neoplasm Metastasis , Stromal Cells/physiology , Amino Acids, Dicarboxylic/pharmacology , Animals , Breast/cytology , Cell Adhesion Molecules/genetics , Cell Movement/drug effects , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Mammary Neoplasms, Experimental/physiopathology , Mice , Myosin Type II/metabolism , Neoplasm Invasiveness , Organ Culture Techniques , Tumor Cells, Cultured
18.
Circ Res ; 117(1): 65-79, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26089364

ABSTRACT

Hypoxia inducible factors (HIFs) are α/ß heterodimeric transcription factors that direct multiple cellular and systemic responses in response to changes in oxygen availability. The oxygen sensitive signal is generated by a series of iron and 2-oxoglutarate-dependent dioxygenases that catalyze post-translational hydroxylation of specific prolyl and asparaginyl residues in HIFα subunits and thereby promote their destruction and inactivation in the presence of oxygen. In hypoxia, these processes are suppressed allowing HIF to activate a massive transcriptional cascade. Elucidation of these pathways has opened several new fields of cardiovascular research. Here, we review the role of HIF hydroxylase pathways in cardiac development and in cardiovascular control. We also consider the current status, opportunities, and challenges of therapeutic modulation of HIF hydroxylases in the therapy of cardiovascular disease.


Subject(s)
Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Adaptation, Physiological , Altitude , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/enzymology , Cardiovascular System/enzymology , Cell Hypoxia , Heart/embryology , Heart Defects, Congenital/embryology , Heart Defects, Congenital/enzymology , Humans , Hydroxylation , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Iron/physiology , Ischemic Preconditioning, Myocardial , Mice , Mixed Function Oxygenases/physiology , Oxygen/metabolism , Polycythemia/enzymology , Polycythemia/genetics , Protein Isoforms , Protein Processing, Post-Translational , Repressor Proteins/physiology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
19.
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
20.
PLoS One ; 9(12): e115974, 2014.
Article in English | MEDLINE | ID: mdl-25546437

ABSTRACT

Oxygen sensor prolyl hydroxylases (PHDs) play important roles in the regulation of HIF-α and cell metabolisms. This study was designed to investigate the direct role of PHD2 in high fat-diet (HFD)-induced cardiac dysfunction. In HFD fed mice, PHD2 expression was increased without significant changes in PHD1 and PHD3 levels in the heart. This was accompanied by a significant upregulation of myeloid differentiation factor 88 (MYD88) and NF-κB. To explore the role of PHD2 in HFD-induced cardiac dysfunction, PHD2 conditional knockout mice were fed a HFD for 16 weeks. Intriguingly, knockout of PHD2 significantly reduced MYD88 and NF-κb expression in HFD mouse hearts. Moreover, knockout of PHD2 inhibited TNFα and ICAM-1 expression, and reduced cell apoptosis and macrophage infiltration in HFD mice. This was accompanied by a significant improvement of cardiac function. Most importantly, conditional knockout of PHD2 at late stage in HFD mice significantly improved glucose tolerance and reversed cardiac dysfunction. Our studies demonstrate that PHD2 activity is a critical contributor to the HFD-induced cardiac dysfunction. Inhibition of PHD2 attenuates HFD-induced cardiac dysfunction by a mechanism involving suppression of MYD88/NF-κb pathway and inflammation.


Subject(s)
Diet, High-Fat/adverse effects , Heart/physiopathology , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Angiogenesis Inducing Agents/metabolism , Animals , Apoptosis , Blood Glucose/metabolism , Body Weight , Feeding Behavior , Glucose Tolerance Test , Heart Function Tests , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Myeloid Differentiation Factor 88/metabolism , Transcription Factor RelA/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...