Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Rev Cancer ; 20(12): 710-726, 2020 12.
Article in English | MEDLINE | ID: mdl-33087883

ABSTRACT

2-Oxoglutarate-dependent dioxygenases (2OGDDs) are a superfamily of enzymes that play diverse roles in many biological processes, including regulation of hypoxia-inducible factor-mediated adaptation to hypoxia, extracellular matrix formation, epigenetic regulation of gene transcription and the reprogramming of cellular metabolism. 2OGDDs all require oxygen, reduced iron and 2-oxoglutarate (also known as α-ketoglutarate) to function, although their affinities for each of these co-substrates, and hence their sensitivity to depletion of specific co-substrates, varies widely. Numerous 2OGDDs are recurrently dysregulated in cancer. Moreover, cancer-specific metabolic changes, such as those that occur subsequent to mutations in the genes encoding succinate dehydrogenase, fumarate hydratase or isocitrate dehydrogenase, can dysregulate specific 2OGDDs. This latter observation suggests that the role of 2OGDDs in cancer extends beyond cancers that harbour mutations in the genes encoding members of the 2OGDD superfamily. Herein, we review the regulation of 2OGDDs in normal cells and how that regulation is corrupted in cancer.


Subject(s)
Dioxygenases/physiology , Ketoglutaric Acids/metabolism , Neoplasms/metabolism , Cell Hypoxia , Citric Acid Cycle , DNA Damage , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Neoplasms/drug therapy , Neoplasms/etiology , Reactive Oxygen Species/metabolism
2.
Proc Natl Acad Sci U S A ; 117(22): 12230-12238, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32414920

ABSTRACT

Tibetans have adapted to the chronic hypoxia of high altitude and display a distinctive suite of physiologic adaptations, including augmented hypoxic ventilatory response and resistance to pulmonary hypertension. Genome-wide studies have consistently identified compelling genetic signatures of natural selection in two genes of the Hypoxia Inducible Factor pathway, PHD2 and HIF2A The product of the former induces the degradation of the product of the latter. Key issues regarding Tibetan PHD2 are whether it is a gain-of-function or loss-of-function allele, and how it might contribute to high-altitude adaptation. Tibetan PHD2 possesses two amino acid changes, D4E and C127S. We previously showed that in vitro, Tibetan PHD2 is defective in its interaction with p23, a cochaperone of the HSP90 pathway, and we proposed that Tibetan PHD2 is a loss-of-function allele. Here, we report that additional PHD2 mutations at or near Asp-4 or Cys-127 impair interaction with p23 in vitro. We find that mice with the Tibetan Phd2 allele display augmented hypoxic ventilatory response, supporting this loss-of-function proposal. This is phenocopied by mice with a mutation in p23 that abrogates the PHD2:p23 interaction. Hif2a haploinsufficiency, but not the Tibetan Phd2 allele, ameliorates hypoxia-induced increases in right ventricular systolic pressure. The Tibetan Phd2 allele is not associated with hemoglobin levels in mice. We propose that Tibetans possess genetic alterations that both activate and inhibit selective outputs of the HIF pathway to facilitate successful adaptation to the chronic hypoxia of high altitude.


Subject(s)
Adaptation, Physiological , DNA-Binding Proteins/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Hypoxia/physiopathology , Loss of Function Mutation , Alleles , Altitude , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/genetics , Humans , Mice , Mice, Knockout , Phenotype , Selection, Genetic , Tibet
3.
Life Sci ; 242: 117167, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31838134

ABSTRACT

Recent studies suggested that prolyl hydroxylase 2 (PHD2) functions as an important regulator in vascular inflammation and Streptococcus pneumonia infection. However, whether PHD2 contributed to tumor progression prompted by intratumoral inflammation remains elusive. In this study, the effects of PHD2 in colon cancer were evaluated, and the underlying molecular mechanisms were investigated. The results showed that overexpressing PHD2 exerted proliferative and migratory inhibition in colon cancer cells. The expression of cell cycle and epithelial-mesenchymal transition (EMT)-associated proteins were changed: CyclinD1, CDK4, N-cadherin, and Vimentin were down-regulated, while E-cadherin was up-regulated in PHD2-overexpressing colon cancer cells. Moreover, in colon cancer xenograft mice, PHD2 overexpression suppressed tumor growth accompanied by decreased Ki67 expression. Importantly, we further demonstrated that overexpressing PHD2 attenuated inflammation in colon cancer xenograft mice through weakening accumulation of myeloid-derived suppressor cells (MDSCs) and M2-like tumor-associated macrophages (TAMs), as well as secretions of pro-inflammatory cytokines including G-CSF, TNF-α, IL-6, IL-8, IL-1ß, and IL-4. Mechanistically, PHD2 overexpression obviously suppressed NF-κB activity through decreasing phosphorylated IκB-α while increasing cytoplasmic NF-κB p65 levels in colon cancer. Our findings support the anti-cancer and anti-inflammatory roles of PHD2 and offer a preclinical proof of tumor progression regulated by cancer cells and inflammation.


Subject(s)
Colonic Neoplasms/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Inflammation/physiopathology , NF-kappa B/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Colonic Neoplasms/physiopathology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Inflammation/metabolism , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation
4.
Proc Natl Acad Sci U S A ; 116(48): 24006-24011, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31712437

ABSTRACT

Highland native Andeans have resided at altitude for millennia. They display high aerobic capacity (VO2max) at altitude, which may be a reflection of genetic adaptation to hypoxia. Previous genomewide (GW) scans for natural selection have nominated Egl-9 homolog 1 gene (EGLN1) as a candidate gene. The encoded protein, EGLN1/PHD2, is an O2 sensor that controls levels of the Hypoxia Inducible Factor-α (HIF-α), which regulates the cellular response to hypoxia. From GW association and analysis of covariance performed on a total sample of 429 Peruvian Quechua and 94 US lowland referents, we identified 5 EGLN1 SNPs associated with higher VO2max (L⋅min-1 and mL⋅min-1⋅kg-1) in hypoxia (rs1769793, rs2064766, rs2437150, rs2491403, rs479200). For 4 of these SNPs, Quechua had the highest frequency of the advantageous (high VO2max) allele compared with 25 diverse lowland comparison populations from the 1000 Genomes Project. Genotype effects were substantial, with high versus low VO2max genotype categories differing by ∼11% (e.g., for rs1769793 SNP genotype TT = 34.2 mL⋅min-1⋅kg-1 vs. CC = 30.5 mL⋅min-1⋅kg-1). To guard against spurious association, we controlled for population stratification. Findings were replicated for EGLN1 SNP rs1769793 in an independent Andean sample collected in 2002. These findings contextualize previous reports of natural selection at EGLN1 in Andeans, and support the hypothesis that natural selection has increased the frequency of an EGLN1 causal variant that enhances O2 delivery or use during exercise at altitude in Peruvian Quechua.


Subject(s)
Altitude , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Hypoxia/genetics , Oxygen/metabolism , Polymorphism, Single Nucleotide , Acclimatization , Adaptation, Physiological , Female , Gene Frequency , Genotype , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Indigenous Peoples , Male , Peru , Selection, Genetic , Stress, Physiological
5.
Cells ; 8(5)2019 04 26.
Article in English | MEDLINE | ID: mdl-31035491

ABSTRACT

All metazoans that utilize molecular oxygen (O2) for metabolic purposes have the capacity to adapt to hypoxia, the condition that arises when O2 demand exceeds supply. This is mediated through activation of the hypoxia-inducible factor (HIF) pathway. At physiological oxygen levels (normoxia), HIF-prolyl hydroxylases (PHDs) hydroxylate proline residues on HIF-α subunits leading to their destabilization by promoting ubiquitination by the von-Hippel Lindau (VHL) ubiquitin ligase and subsequent proteasomal degradation. HIF-α transactivation is also repressed in an O2-dependent way due to asparaginyl hydroxylation by the factor-inhibiting HIF (FIH). In hypoxia, the O2-dependent hydroxylation of HIF-α subunits by PHDs and FIH is reduced, resulting in HIF-α accumulation, dimerization with HIF-ß and migration into the nucleus to induce an adaptive transcriptional response. Although HIFs are the canonical substrates for PHD- and FIH-mediated protein hydroxylation, increasing evidence indicates that these hydroxylases may also have alternative targets. In addition to PHD-conferred alterations in protein stability, there is now evidence that hydroxylation can affect protein activity and protein/protein interactions for alternative substrates. PHDs can be pharmacologically inhibited by a new class of drugs termed prolyl hydroxylase inhibitors which have recently been approved for the treatment of anemia associated with chronic kidney disease. The identification of alternative targets of HIF hydroxylases is important in order to fully elucidate the pharmacology of hydroxylase inhibitors (PHI). Despite significant technical advances, screening, detection and verification of alternative functional targets for PHDs and FIH remain challenging. In this review, we discuss recently proposed non-HIF targets for PHDs and FIH and provide an overview of the techniques used to identify these.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Mixed Function Oxygenases/physiology , Prolyl-Hydroxylase Inhibitors/pharmacology , Animals , Humans , Hydroxylation , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
6.
Chem Biol Interact ; 303: 40-49, 2019 Apr 25.
Article in English | MEDLINE | ID: mdl-30817904

ABSTRACT

Tumor hypoxia is a well-known microenvironmental factor that causes cancer progression and resistance to cancer treatment. Proline hydroxylases (PHDs), a small protein family, belong to an evolutionarily conserved superfamily of dioxygenases, considered the central regulator of the molecular hypoxia response. Prolyl-4-hydroxylase 2 (PHD2), one member of PHDs family, regulates the stability of the hypoxia-inducible factor-1 alpha (HIF-1α) in response to oxygen availability. During hypoxia, the inhibition of PHD2 permits the accumulation of HIF-1α, allowing the cellular adaptation to oxygen limitation, causing activation of numerous genes, which enhances the angiogenesis, metastasis and invasiveness. Accurate regulation of oxygen homeostasis is essential, and which implies PHD2 may have a regulatory role in the pathogenesis of cancer. Although ample evidence exists for a positive correlation between HIFs and tumor formation, metastasis and poor prognosis, the function of the PHD2 in carcinogenesis is less well understood. Despite their original role as the oxygen sensors of the cell and many of the its functions are clearly conveyed through the HIF system, PHD2 is currently known to display HIF-independent and hydroxylase-independent functions in cancer cells and stroma in the control of different cellular pathways. In this review, we summarize the recent advances in the structure, regulation and functions of PHD2 in cancer microenvironment.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Tumor Microenvironment , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Oxygen/metabolism , Prolyl Hydroxylases , Signal Transduction
7.
Am J Physiol Lung Cell Mol Physiol ; 314(2): L256-L275, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29074488

ABSTRACT

Pulmonary vascular remodeling characterized by concentric wall thickening and intraluminal obliteration is a major contributor to the elevated pulmonary vascular resistance in patients with idiopathic pulmonary arterial hypertension (IPAH). Here we report that increased hypoxia-inducible factor 2α (HIF-2α) in lung vascular endothelial cells (LVECs) under normoxic conditions is involved in the development of pulmonary hypertension (PH) by inducing endothelial-to-mesenchymal transition (EndMT), which subsequently results in vascular remodeling and occlusive lesions. We observed significant EndMT and markedly increased expression of SNAI, an inducer of EndMT, in LVECs from patients with IPAH and animals with experimental PH compared with normal controls. LVECs isolated from IPAH patients had a higher level of HIF-2α than that from normal subjects, whereas HIF-1α was upregulated in pulmonary arterial smooth muscle cells (PASMCs) from IPAH patients. The increased HIF-2α level, due to downregulated prolyl hydroxylase domain protein 2 (PHD2), a prolyl hydroxylase that promotes HIF-2α degradation, was involved in enhanced EndMT and upregulated SNAI1/2 in LVECs from patients with IPAH. Moreover, knockdown of HIF-2α (but not HIF-1α) with siRNA decreases both SNAI1 and SNAI2 expression in IPAH-LVECs. Mice with endothelial cell (EC)-specific knockout (KO) of the PHD2 gene, egln1 (egln1EC-/-), developed severe PH under normoxic conditions, whereas Snai1/2 and EndMT were increased in LVECs of egln1EC-/- mice. EC-specific KO of the HIF-2α gene, hif2a, prevented mice from developing hypoxia-induced PH, whereas EC-specific deletion of the HIF-1α gene, hif1a, or smooth muscle cell (SMC)-specific deletion of hif2a, negligibly affected the development of PH. Also, exposure to hypoxia for 48-72 h increased protein level of HIF-1α in normal human PASMCs and HIF-2α in normal human LVECs. These data indicate that increased HIF-2α in LVECs plays a pathogenic role in the development of severe PH by upregulating SNAI1/2, inducing EndMT, and causing obliterative pulmonary vascular lesions and vascular remodeling.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Endothelial Cells/pathology , Epithelial-Mesenchymal Transition , Hypertension, Pulmonary/etiology , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Animals , Cells, Cultured , Endothelial Cells/metabolism , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Vascular Remodeling
8.
Kidney Int ; 92(6): 1370-1383, 2017 12.
Article in English | MEDLINE | ID: mdl-28847650

ABSTRACT

Hypoxia in the embryo is a frequent cause of intra-uterine growth retardation, low birth weight, and multiple organ defects. In the kidney, this can lead to low nephron endowment, predisposing to chronic kidney disease and arterial hypertension. A key component in cellular adaptation to hypoxia is the hypoxia-inducible factor pathway, which is regulated by prolyl-4-hydroxylase domain (PHD) dioxygenases PHD1, PHD2, and PHD3. In the adult kidney, PHD oxygen sensors are differentially expressed in a cell type-dependent manner and control the production of erythropoietin in interstitial cells. However, the role of interstitial cell PHDs in renal development has not been examined. Here we used a genetic approach in mice to interrogate PHD function in FOXD1-expressing stroma during nephrogenesis. We demonstrate that PHD2 and PHD3 are essential for normal kidney development as the combined inactivation of stromal PHD2 and PHD3 resulted in renal failure that was associated with reduced kidney size, decreased numbers of glomeruli, and abnormal postnatal nephron formation. In contrast, nephrogenesis was normal in animals with individual PHD inactivation. We furthermore demonstrate that the defect in nephron formation in PHD2/PHD3 double mutants required intact hypoxia-inducible factor-2 signaling and was dependent on the extent of stromal hypoxia-inducible factor activation. Thus, hypoxia-inducible factor prolyl-4-hydroxylation in renal interstitial cells is critical for normal nephron formation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Kidney/growth & development , Procollagen-Proline Dioxygenase/physiology , Renal Insufficiency/genetics , Anemia/blood , Anemia/drug therapy , Anemia/etiology , Animals , Cell Hypoxia/physiology , Clinical Trials, Phase III as Topic , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Hydroxylation/physiology , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Kidney/cytology , Kidney/metabolism , Kidney Diseases/complications , Kidney Diseases/drug therapy , Mice , Molecular Targeted Therapy/methods , Mutation , Organ Size/physiology , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Procollagen-Proline Dioxygenase/genetics , Renal Insufficiency/mortality , Renal Insufficiency/pathology , Stromal Cells/metabolism
9.
Hemodial Int ; 21 Suppl 1: S110-S124, 2017 06.
Article in English | MEDLINE | ID: mdl-28449418

ABSTRACT

A classic response to systemic hypoxia is the increase in red blood cell production. This response is controlled by the prolyl hydroxylase domain/hypoxia-inducible factor (HIF) pathway, which regulates a broad spectrum of cellular functions. The discovery of this pathway as a key regulator of erythropoiesis has led to the development of small molecules that stimulate the production of endogenous erythropoietin and enhance iron metabolism. This review provides a concise overview of the cellular and molecular mechanisms that govern HIF-induced erythropoietic responses and provides an update on clinical experience with compounds that target HIF-prolyl hydroxylases for anemia therapy.


Subject(s)
Erythropoiesis/drug effects , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Iron/metabolism , Prolyl-Hydroxylase Inhibitors/therapeutic use , Anemia/drug therapy , Barbiturates/adverse effects , Barbiturates/therapeutic use , Clinical Trials as Topic , Erythropoietin/biosynthesis , Glycine/adverse effects , Glycine/analogs & derivatives , Glycine/therapeutic use , Humans , Isoquinolines/adverse effects , Isoquinolines/therapeutic use , Picolinic Acids/adverse effects , Picolinic Acids/therapeutic use , Renal Dialysis/adverse effects
10.
Mol Biol Cell ; 28(2): 261-269, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27881662

ABSTRACT

Autosomal dominant polycystic kidney disease is caused by mutations in the genes encoding polycystin-1 (PC1) and polycystin-2 (PC2), which form an ion channel complex that may mediate ciliary sensory processes and regulate endoplasmic reticulum (ER) Ca2+ release. Loss of PC1 expression profoundly alters cellular energy metabolism. The mechanisms that control the trafficking of PC1 and PC2, as well as their broader physiological roles, are poorly understood. We found that O2 levels regulate the subcellular localization and channel activity of the polycystin complex through its interaction with the O2-sensing prolyl hydroxylase domain containing protein EGLN3 (or PHD3), which hydroxylates PC1. Moreover, cells lacking PC1 expression use less O2 and show less mitochondrial Ca2+ uptake in response to bradykinin-induced ER Ca2+ release, indicating that PC1 can modulate mitochondrial function. These data suggest a novel role for the polycystins in sensing and responding to cellular O2 levels.


Subject(s)
TRPP Cation Channels/metabolism , TRPP Cation Channels/physiology , Animals , Endoplasmic Reticulum/metabolism , Humans , Hypoxia , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , LLC-PK1 Cells , Mitochondria/metabolism , Oxygen/metabolism , Protein Transport/physiology , Swine
11.
Oncotarget ; 7(34): 54317-54328, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27527871

ABSTRACT

Prolyl hydroxylase domain protein 2 (PHD2) is a key oxygen sensor, setting low steady-state level of hypoxia-inducible factor-α (HIF-α). Here, we showed that treatment of cobalt chloride (CoCl2), a hypoxia mimic, in HK-2 tubular epithelial cells induced PHD2 and HIF-1/2α expression as well as cell apoptosis and autophagy activation. Three methyladenine (3-MA), the autophagy inhibitor, blocked autophagy and protected HK-2 cells from CoCl2. Significantly, siRNA knockdown of PHD2 also protected HK-2 cells from CoCl2,possibly via increasing HIF-1α expression. Reversely, HIF-1α siRNA knockdown almost abolished cytoprotection by PHD2 siRNA in CoCl2-treated HK-2 cells. In vivo, pretreatment with a PHD inhibitor L-mimosine remarkably attenuated mice renal ischemia-reperfusion injuries. Molecularly, L-mimosine inhibited apoptosis and inflammatory responses in injured mice kidneys. Together, our results suggest that PHD2 silence or inhibition protects human renal epithelial cells and mice kidney from hypoxia injuries.


Subject(s)
Cell Hypoxia , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Kidney/drug effects , Mimosine/therapeutic use , Prolyl-Hydroxylase Inhibitors/therapeutic use , Reperfusion Injury/prevention & control , Animals , Apoptosis/drug effects , Cobalt/pharmacology , Cytoprotection , Epithelial Cells/drug effects , Humans , Kidney/blood supply , Mice
12.
Physiol Rep ; 4(5)2016 Mar.
Article in English | MEDLINE | ID: mdl-26997627

ABSTRACT

Type 2 diabetes is associated with impaired nutrient-regulated anaplerosis and insulin secretion in pancreatic ß-cells. One key anaplerotic substrate that may be involved in regulating insulin release is α-ketoglutarate (αKG). Since prolyl hydroxylase domain proteins (PHDs) can metabolize cytosolic αKG, we sought to explore the role of this enzyme in the regulation of ß-cell function. The oxygen-sensing PHDs regulate the stability of hypoxia-inducible factor 1α (HIF1α) as well as other proline-containing proteins by catalyzing the hydroxylation of proline residues. This reaction is dependent on sufficient levels of oxygen, iron, and αKG. In the present study, we utilized both pharmacological and genetic approaches to assess the impact of inhibiting PHD activity on ß-cell function. We demonstrate that ethyl-3,4-dihydroxybenzoate (EDHB), a PHD inhibitor, significantly blunted glucose-stimulated insulin secretion (GSIS) from 832/13 clonal cells, rat, and human islets. EDHB reduced glucose utilization, ATP/ADP ratio, and key TCA cycle intermediates such as pyruvate, citrate, fumarate, and malate. siRNA-mediated knockdown of PHD1 and PHD3 inhibited GSIS, whereas siRNA-mediated knockdown of PHD2 had no effect on GSIS. Taken together, the current results demonstrate an important role for PHDs as mediators of islet insulin secretion.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Cell Line , Gene Knockdown Techniques/methods , Humans , Insulin Secretion , Protein Structure, Tertiary/physiology , Rats
13.
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
14.
Nephron Clin Pract ; 127(1-4): 70-4, 2014.
Article in English | MEDLINE | ID: mdl-25343825

ABSTRACT

Oxygen is vital to mammalian survival. Oxygen deprivation, defined as hypoxia, elicits adaptive responses in cells and tissues, a process regulated by proteins known as hypoxia-inducible factors (HIF). Animal studies have provided compelling data to demonstrate a pivotal role for the HIF pathway in the pathogenesis of acute kidney injury (AKI) that have led to initial human clinical trials examining this pathway in ischemia-reperfusion injury in various organ systems, including the kidney. HIF are master regulators and mediate adaptive responses to low oxygen in tissues and cells. This review will summarize recent key advances in the field highlighting preclinical and clinical studies relevant to the HIF pathway in the pathophysiology of AKI.


Subject(s)
Acute Kidney Injury/metabolism , Cell Hypoxia , Acetazolamide/therapeutic use , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Adaptation, Physiological , Altitude , Animals , Barbiturates/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/physiology , Clinical Trials, Phase II as Topic , Gene Expression Regulation , Glycine/analogs & derivatives , Glycine/therapeutic use , Humans , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Hypoxia-Inducible Factor 1/physiology , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Intercellular Signaling Peptides and Proteins/physiology , Kidney/blood supply , Mice , Rats , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Transcription Factors/antagonists & inhibitors , Transcription Factors/physiology , Transcription, Genetic , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/physiology
15.
Mol Biol Cell ; 25(18): 2788-96, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25079693

ABSTRACT

Actin filaments play an essential role in cell movement, and many posttranslational modifications regulate actin filament assembly. Here we report that prolyl hydroxylase 3 (PHD3) interacts with nonmuscle actin in human cells and catalyzes hydroxylation of actin at proline residues 307 and 322. Blocking PHD3 expression or catalytic activity by short hairpin RNA knockdown or pharmacological inhibition, respectively, decreased actin prolyl hydroxylation. PHD3 knockdown increased filamentous F-actin assembly, which was reversed by PHD3 overexpression. PHD3 knockdown increased cell velocity and migration distance. Inhibition of PHD3 prolyl hydroxylase activity by dimethyloxalylglycine also increased actin polymerization and cell migration. These data reveal a novel role for PHD3 as a negative regulator of cell motility through posttranslational modification of nonmuscle actins.


Subject(s)
Actins/metabolism , Cell Movement , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Amino Acid Sequence , HEK293 Cells , HeLa Cells , Humans , Hydroxylation , Molecular Sequence Data , Polymerization , Proline/metabolism , Protein Multimerization , Protein Processing, Post-Translational
16.
Yao Xue Xue Bao ; 49(2): 151-7, 2014 Feb.
Article in Chinese | MEDLINE | ID: mdl-24761603

ABSTRACT

Prolyl-4-hydroxylase domain (PHDs) family is one of the most important regulatory factors in hypoxic stress. PHD2 plays a critical role in cells and tissues adaptation to the low oxygen environment. Its hydroxylation activity regulates the stability and transcriptional activity of the hypoxia-inducible factor 1 (HIF-1), which is the key factor in response to hypoxic stress. Subsequently, PHD2 acts as an important factor in oxygen homeostasis. Studies have shown that PHD2, through its regulation on HIF-1, plays an important role in the post-ischemic neovascularization. Furthermore, under hypoxic condition, PHD2 also regulates other pathways that positively regulate angiogenesis factors HIF-1 independently. Moreover, recently, several evidences have also shown that PHD2 also affects tumor growth and metastasis in a tumor microenvironment. Based on these facts, PHD2 have been considered as a potential therapeutic target both in treating ischemic diseases and tumors. Here, we review the molecular regulation mechanism of PHD2 and its physiological and pathological functions. We focus on the role of PHD2 in both therapeutic angiogenesis for ischemic disease and tumor angiogenesis, and the current progress in utilizing PHD2 as a therapeutic target.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases , Neoplasms/blood supply , Neovascularization, Pathologic , Vascular Diseases/pathology , Animals , Humans , Hydroxylation , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Tumor Microenvironment , Vascular Diseases/therapy
17.
FASEB J ; 28(6): 2455-65, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24558194

ABSTRACT

Intervertebral disc degeneration is the leading cause of chronic back pain. Recent studies show that raised level of SDC4, a cell-surface heparan sulfate (HS) proteoglycan, plays a role in pathogenesis of disc degeneration. However, in nucleus pulposus (NP) cells of the healthy intervertebral disc, the mechanisms that control expression of SDC4 and its physiological function are unknown. Hypoxia induced SDC4 mRNA and protein expression by ~2.4- and 4.4-fold (P<0.05), respectively, in NP cells. While the activity of the SDC4 promoter containing hypoxia response element (HRE) was induced 2-fold (P<0.05), the HRE mutation decreased the activity by 40% in hypoxia. Transfections with plasmids coding prolyl-4-hydroxylase domain protein 2 (PHD2) and ShPHD2 show that hypoxic expression of SDC4 mRNA and protein is regulated by PHD2 through controlling hypoxia-inducible factor 1α (HIF-1α) levels. Although overexpression of HIF-1α significantly increased SDC4 protein levels, stable suppression of HIF-1α and HIF-1ß decreased SDC4 expression by 50% in human NP cells. Finally, suppression of SDC4 expression, as well as HS function, resulted in an ~2-fold increase in sex-determining region Y (SRY)-box 9 (Sox9) mRNA, and protein (P<0.05) and simultaneous increase in Sox9 transcriptional activity and target gene expression. Taken together, our findings suggest that in healthy discs, SDC4, through its HS side chains, contributes to maintenance of the hypoxic tissue niche by controlling baseline expression of Sox9.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Intervertebral Disc/metabolism , Syndecan-4/biosynthesis , Animals , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , NF-kappa B/physiology , RNA, Messenger/metabolism , Rats , SOX9 Transcription Factor/biosynthesis
18.
Int J Cancer ; 134(4): 849-58, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-23913502

ABSTRACT

The tumor microenvironment plays a pivotal role during cancer development and progression. The balance between suppressive and cytotoxic responses of the tumor immune microenvironment has been shown to have a direct effect on the final outcome in various human and experimental tumors. Recently, we demonstrated that the oxygen sensor HIF-prolyl hydroxylase-2 (PHD2) plays a detrimental role in tumor cells, stimulating systemic growth and metastasis in mice. In our current study, we show that the conditional ablation of PHD2 in the hematopoietic system also leads to reduced tumor volume, intriguingly generated by an imbalance between enhanced cell death and improved proliferation of tumor cells. This effect seems to rely on the overall downregulation of protumoral as well as antitumoral cytokines. Using different genetic approaches, we were able to confine this complex phenotype to the crosstalk of PHD2-deficient myeloid cells and T-lymphocytes. Taken together, our findings reveal a multifaceted role for PHD2 in several hematopoietic lineages during tumor development and might have important implications for the development of tumor therapies in the future.


Subject(s)
Carcinoma, Lewis Lung/prevention & control , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Melanoma, Experimental/prevention & control , Myeloid Cells/pathology , T-Lymphocytes/pathology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Bone Marrow/metabolism , Bone Marrow/pathology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Cell Movement , Cell Proliferation , Cytokines/genetics , Cytokines/metabolism , Disease Progression , Flow Cytometry , Gene Expression Profiling , Immunoenzyme Techniques , Integrases/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
19.
J Physiol ; 591(14): 3565-77, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23690557

ABSTRACT

Oxygen-dependent prolyl hydroxylation of hypoxia-inducible factor (HIF) by a set of closely related prolyl hydroxylase domain enzymes (PHD1, 2 and 3) regulates a range of transcriptional responses to hypoxia. This raises important questions about the role of these oxygen-sensing enzymes in integrative physiology. We investigated the effect of both genetic deficiency and pharmacological inhibition on the change in ventilation in response to acute hypoxic stimulation in mice. Mice exposed to chronic hypoxia for 7 days manifest an exaggerated hypoxic ventilatory response (HVR) (10.8 ± 0.3 versus 4.1 ± 0.7 ml min(-1) g(-1) in controls; P < 0.01). HVR was similarly exaggerated in PHD2(+/-) animals compared to littermate controls (8.4 ± 0.7 versus 5.0 ± 0.8 ml min(-1) g(-1); P < 0.01). Carotid body volume increased (0.0025 ± 0.00017 in PHD2(+/-) animals versus 0.0015 ± 0.00019 mm(3) in controls; P < 0.01). In contrast, HVR in PHD1(-/-) and PHD3(-/-) mice was similar to littermate controls. Acute exposure to a small molecule PHD inhibitor (PHI) (2-(1-chloro-4-hydroxyisoquinoline-3-carboxamido) acetic acid) did not mimic the ventilatory response to hypoxia. Further, 7 day administration of the PHI induced only modest increases in HVR and carotid body cell proliferation, despite marked stimulation of erythropoiesis. This was in contrast with chronic hypoxia, which elicited both exaggerated HVR and cellular proliferation. The findings demonstrate that PHD enzymes modulate ventilatory sensitivity to hypoxia and identify PHD2 as the most important enzyme in this response. They also reveal differences between genetic inactivation of PHDs, responses to hypoxia and responses to a pharmacological inhibitor, demonstrating the need for caution in predicting the effects of therapeutic modulation of the HIF hydroxylase system on different physiological responses.


Subject(s)
Carotid Body/pathology , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Hypoxia/physiopathology , Pulmonary Ventilation/physiology , Animals , Carotid Body/physiopathology , Hyperplasia/physiopathology , Hypoxia-Inducible Factor 1/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...