Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jul 25.
Published in final edited form as: Hemodial Int. 2017 Jun;21(Suppl 1):S110–S124. doi: 10.1111/hdi.12567

HIF-prolyl hydroxylases as therapeutic targets in erythropoiesis and iron metabolism

Volker H HAASE 1,2,3
PMCID: PMC5526677  NIHMSID: NIHMS876502  PMID: 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.

Keywords: Anemia, erythropoietin, iron, hypoxia-inducible factor, prolyl hydroxylase

INTRODUCTION

A classic response to systemic hypoxia is the increased production of red blood cells (rbc). This association was first noted in the second half of the 19th century by Bert and Jourdanet,13 who studied the physiologic effects of hypoxia. The search for the humoral factor that mediated this effect resulted in the purification of the glycoprotein hormone erythropoietin (EPO) in 1977, which was followed by the cloning of the EPO gene in 1985.46 About one decade later, Semenza purified hypoxia-inducible factor (HIF) as the heterodimeric transcription factor responsible for the hypoxic induction of EPO and other oxygen-sensitive genes.711 The laboratories of Ratcliffe and Kaelin then established that hydroxylation of specific proline residues within the HIF-α subunit was required for its inactivation.12,13 This observation led to the identification of prolyl hydroxylase domain (PHD)-2, a 2-oxoglutarate (2OG)-dependent dioxygenase that utilizes molecular oxygen to carry out this hydroxylation reaction. PHD2 promotes HIF-α degradation under normoxia, whereas its inhibition results in HIF activation in most cell types.14 Due to their seminal contributions to understanding the molecular basis of cellular oxygen sensing in metazoans, Semenza, Ratcliffe, and Kaelin received the prestigious Albert Lasker award in 2016.

THE PHD/HIF OXYGEN-SENSING PATHWAY

The PHD/HIF axis is a critically important oxygen-sensing pathway that mediates tissue adaptation to low oxygen environments primarily through the transcriptional regulation of gene expression. The key components of this pathway and their regulation by hypoxia are discussed in this section.

HIF transcription factors

HIFs are basic pleiotropic helix-loop-helix transcription factors that belong to the PAS (PER/aryl hydrocarbon receptor nuclear translocator [ARNT]/single minded) family of transcription factors. They consist of two subunits, an oxygen-sensitive α-subunit and a constitutively expressed β-subunit, which is often referred to as the ARNT.1517 The HIF-a subunit, which is continuously synthesized, is rapidly degraded in the presence of molecular oxygen. Three HIF-α-subunits have been identified, HIF-1α, HIF-2α (also known as EPAS1), and HIF-3α. Under hypoxic conditions, HIF-α is no longer degraded and translocates to the nucleus where it forms a heterodimer with HIF-β and activates gene transcription.

HIF-1 and HIF-2, the most extensively studied HIF transcription factors, facilitate oxygen delivery and cellular adaptation to hypoxia by regulating a wide spectrum of hypoxia responses such as angiogenesis, anaerobic glucose metabolism, mitochondrial biogenesis, and others.18 The list of genes that are directly HIF-regulated is large and several hundred high stringency HIF binding sites have been identified across the genome.19 Although HIF-1 and HIF-2 share many transcriptional targets, certain genes are not co-regulated. For example, HIF-1 regulates anaerobic glycolysis,20 whereas EPO production and certain iron genes are HIF-2-controlled.2126

HIF-prolyl hydroxylases

Under normoxic conditions, HIF-α is targeted for rapid degradation via the hydroxylation of specific proline residues (Figure 1). Hydroxylated HIF-α is tagged for proteasomal degradation by the von Hippel-Lindau (VHL) tumor suppressor protein, which functions as the substrate recognition component of an E3 ubiquitin ligase.28,29 Under hypoxic conditions, HIF-prolyl hydroxylation is reduced, HIF-α is no longer degraded and translocates to the nucleus where it heterodimerizes with HIF-β and activates gene transcription (Figure 1). Any reduction in HIF-proline hydroxylation or impairment in VHL function decreases HIF degradation and results in increased HIF target gene expression. For example, patients with certain inactivating VHL mutations are predisposed to the development of CNS hemangioblastomas, clear cell renal cancer, and pheochromocytomas, tumors which are characterized by increased HIF-regulated gene expression.30 Furthermore, certain mutations that affect the ability of cells to effectively hydroxylate HIF-α are associated with abnormal regulation of HIF activity and result in the development of polycythemia.31

Figure 1.

Figure 1

Schematic overview of the PHD (prolyl hydroxylase domain)/hypoxia-inducible factor (HIF) pathway. Although the oxygen-sensitive α-subunit of HIF is constitutively synthesized, it is rapidly degraded under normoxic conditions. Under hypoxia, however, cellular HIF-α levels build up and HIF-α translocates to the nucleus, where it forms a heterodimer with HIF-β. Proteasomal degradation of HIF-α is mediated by the pVHL-E3-ubiquitin ligase complex and requires HIF-α prolyl-4-hydroxylation by oxygen- and iron-dependent PHD dioxygenases (PHD1-3). The decarboxylation of 2-oxoglurate (2OG) produces hydroxylated HIF-α, succinate and CO2. PHD or von Hippel-Lindau inhibition results in increased transcription of HIF-regulated genes such as vascular endothelial growth factor (VEGF), erythropoietin (EPO), phosphoglycerate kinase 1 (PGK1), lactate dehydrogenase (LDH), and other genes involved in the regulation of hypoxia responses, including cellular metabolism and mitochondrial function, inflammation, vascular function, and oxidative stress and other responses. Shown is the chemical structure of a HIF-PHD inhibitor (PHI) capable of effectively stimulating the production of endogenous EPO in hemodialysis patients.27

Oxygen-dependent hydroxylation of HIF-α is carried out by PHD1, PHD2, and PHD3, which function as oxygen sensors in the HIF pathway.32 PHDs belong to a large family of 2OG-dependent dioxygenases with more than 60 members. Because these dioxygenases utilize molecular oxygen for hydroxylation, they link oxygen, intermediary, and amino acid metabolism to various cellular processes that include HIF regulation/hypoxia responses, collagen synthesis, epigenetic gene regulation, and fatty acid metabolism.33

Small molecules, such as reactive oxygen species, nitric oxide, and the Krebs cycle intermediates succinate and fumarate inhibit PHD catalytic activity leading to HIF-α stabilization and activation of HIF transcriptional programs.32 The latter is clinically relevant in patients with fumarate hydratase deficiency who are predisposed to the development of the hereditary leiomyomatosis renal cell cancer syndrome, which is characterized by increased HIF activity in affected tissues.34,35 Structural analogs of 2OG that block 2OG and HIF-α from accessing the PHD catalytic center and thus reversibly inhibit HIF-α hydroxylation are now under clinical investigation for renal anemia and other indications.36 Current clinical experience with these compounds is discussed below.

THE PHD/HIF AXIS IN ERYTHROPOIESIS AND IRON METABOLISM

The PHD/HIF axis coordinates hypoxia responses across multiple cell types and tissues. In addition to renal and hepatic EPO production, HIF controls iron uptake and utilization and facilitates erythroid progenitor maturation and proliferation in the bone marrow. Key aspects of renal and hepatic EPO regulation and the mechanisms by which HIF controls iron homeostasis are discussed in this section.

Oxygen-dependent EPO production in the kidney

The kidney is the main source of EPO in adults, where it is synthesized by perivascular fibroblasts and pericytes.36 In chronic kidney injury, renal perivascular interstitial cells and pericytes are also major cellular sources of myofibroblasts, which are interstitial cells that promote fibrosis and progression of CKD through excessive collagen production. Therefore, perivascular interstitial cells and pericytes provide an important cellular link between fibrosis and anemia development in CKD.36

Renal EPO mRNA expression and plasma EPO levels correlate directly with the number of EPO-producing cells (EPC) in the kidney.37 Renal EPC are exquisite oxygen sensors that respond to small changes in tissue pO2 with HIF-2α stabilization and increased EPO transcription.25,38 In contrast to Hep3B hepatoma cells, which were used to purify HIF-1, the hypoxic induction of EPO in the kidney is HIF-2-dependent. This paradigm is based on genetic studies in mice, the analysis of human mutations, and on immunohistochemical examinations of rodent and human tissues.39

Our laboratory has recently shown that renal EPC are derived from forkhead box D1-expressing stroma, which surrounds the cap mesenchyme during kidney development and gives rise to essentially all nonendothelial renal interstitial cells including pericytes and perivascular fibroblasts, mesangial cells, and vascular smooth muscle cells. Interestingly, the ability to produce EPO is shared between brain and renal pericytes, which are both of neural crest origin.38,40 PHD2 is the main prolyl hydroxylase that controls HIF activity under normoxic conditions. Its genetic inactivation in renal interstitial cells results in HIF-2 activation and EPO production in ~40% of cortical and outer medullary renal interstitial cells leading to severe polycythemia.38

HIF-2 also controls EPO synthesis in hepatocytes, which are the main source of EPO production during embryonic development. Although the liver does not contribute to plasma EPO levels in adults under baseline conditions, EPO synthesis in the adult liver can be reactivated by moderate to severe hypoxia, VHL, or PHD inhibition.27,41,42 Furthermore, in the setting of end stage renal disease (ESRD) a significant fraction of plasma EPO is liver-derived.43

HIF coordinates erythropoiesis with iron metabolism

Stimulation of erythropoietic activity as a result of HIF-2-induced EPO synthesis increases iron demand in the bone marrow. To meet these requirements, iron uptake and utilization has to be adjusted (Figure 2). HIF-2 increases the transcription of divalent metal transporter 1 (DMT1) and duodenal cytochrome b (DCYTB) and thus promotes iron uptake as demonstrated in animal models of iron-deficiency and hemochromatosis.26,44,45 DMT1 transports iron into the cytoplasm of cells, and DCYTB reduces ferric iron to its ferrous form (Fe2+) before it is taken up from the gut lumen into intestinal cells via DMT1 (Figure 2). In addition, HIF regulates the transcription of transferrin, which transports iron in its ferric form (Fe3+) in blood, transferrin receptor TFR1,4648 ceruloplasmin, which oxidizes Fe2+ to Fe3+ and is also important for iron transport,49 heme-oxygenase-1, which is involved in the recycling of iron from phagocytosed erythrocytes,50 and ferroportin (FPN),51 the only known cellular iron exporter.

Figure 2.

Figure 2

HIF-dependent regulation of iron metabolism. Schematic overview of HIF-regulated genes involved in iron metabolism shown in red. In the gut, duodenal cytochrome b (DCYTB) reduces ferric iron (Fe3+) to Fe2+, which enters enterocytes via the divalent metal transporter-1 (DMT1). DCYTB and DMT1 are both bona fide HIF-2-regulated genes. Release of iron into the circulation occurs via ferroportin (FPN), which is hepcidin-regulated but also HIF-inducible. In blood, iron is transported in complex with transferrin (TF) to the liver, cells of the reticulo-endothelial system (RES), bone marrow and other organs. Increased erythropoietic activity in the bone marrow produces growth differentiation factor 15 (GDF15) and erythroferrone, which have been shown suppress hepcidin in hepatocytes. Inflammatory cytokines stimulate hepcidin production in the liver and lead to reduced ferroportin surface expression and hypoferremia.

FPN is targeted for degradation by hepcidin, a small 25-amino-acid peptide, produced by hepatocytes. Its production is increased by iron and inflammatory cytokines such as interleukin 6. Because hepcidin suppresses FPN cell surface expression, high plasma hepcidin levels are associated with reduced intestinal iron uptake, and impaired release of iron from internal stores.52 In patients with advanced CKD plasma hepcidin levels are frequently elevated and contribute to the pathogenesis of renal anemia as increased hepcidin leads to functional iron-deficiency.36,53

Systemic HIF activation suppresses hepcidin production in the liver enhancing iron uptake and mobilization.54,55 Although initial studies suggested a direct role for HIF-1 in the suppression of hepcidin transcription,56 subsequent analysis of mouse models with global or liver-specific HIF activation supported the notion that hepcidin suppression was dependent on EPO-mediated stimulation of erythropoiesis, i.e., that HIF does not act as a direct repressor of hepcidin transcription in the liver.57,58 This notion is supported by genome-wide studies in cell lines.19

The relationship between HIF and iron metabolism is bidirectional, as iron itself can modulate HIF activation. The gene encoding HIF-2α contains an iron response element in its 5′ UTR, which binds iron regulatory proteins (IRPs).59 Similar to the regulation of ferritin translation, the binding of IRPs to the HIF2A 5′ UTR has been shown to inhibit HIF-2α translation and thus decreases HIF-2α protein levels in the presence of low intracellular iron.6062 According to this model, renal EPO production would be relatively reduced in iron-deficiency anemia compared to other types of anemia with normal or elevated plasma iron levels.

In addition to regulating iron metabolism, hypoxia and the PHD/HIF pathway stimulate erythropoiesis through direct effects on the bone marrow via stimulation of EPO receptor expression, hemoglobin synthesis,6367 and modulation of stem cell maintenance, lineage differentiation, and maturation.68,69

HIF-PROLYL HYDROXYLASES AS DRUG TARGETS IN RENAL ANEMIA

Anemia associated with CKD is due to a relative deficiency in renal EPO production, concomitant functional, and/or absolute iron deficiency and resistance to EPO signaling, which is frequently seen in the setting of inflammation.36 Pharmacologic activation of HIF signaling has the potential to provide a more comprehensive and physiologic approach to the treatment of renal anemia compared to therapy with recombinant EPO preparations alone. Recent clinical studies of compounds that target the PHD/HIF pathway are discussed below.

Rational for therapeutic targeting of the HIF system

The administration of erythropoiesis stimulating agents (ESAs), such as first-generation recombinant human EPO (rhEPO) preparations epoetin alfa and epoetin beta, and second-generation epoetins darbepoetin alfa and epoetin beta pegol, is current standard of care for patients with renal anemia.7072 ESAs are effective in maintaining hemoglobin levels in target range in most patients and their use has reduced the need for red blood cell transfusions. Despite its clinical success, ESA therapy in advanced CKD is associated with an increased risk for cardiovascular events.7375 Although the underlying mechanisms are not clearly understood, ESA-associated cardiovascular events are most likely due to (a) intermittent supra-physiologic plasma EPO levels following the injections of high doses of ESAs to reach target hemoglobin levels, (b) hemoglobin oscillations, and (c) excursions of hemoglobin levels beyond target range.7680

The identification of oxygen- and iron-dependent HIF-PHDs as key regulators of erythropoiesis has catalyzed the development of novel therapeutic agents, which are referred to as HIF-PHD inhibitors (HIF-PHIs). HIF-PHIs reversibly inhibit PHD catalysis and induce a transient increase in the expression of HIF-regulated genes, including renal, and hepatic EPO.27 However, the increase of plasma EPO in CKD patients successfully treated with HIF-PHIs is significantly lower than in patients treated with injectable rhEPO.81,82 Although clinical data are not yet available, HIF-PHI therapy holds the promise to reduce the cardiovascular risk associated with conventional ESA therapy. HIF-PHIs have also beneficial effects on iron homeostasis, as they lower hepcidin and ferritin and increase total iron binding capacity (TIBC) in patients with CKD.8186 Furthermore, pending additional investigation, HIF-PHIs have the potential to protect renal EPCs from loss of EPO production in chronic kidney injury, as suggested by preclinical studies.87 Owing to their combined action on EPO synthesis and iron metabolism, and their ability to stimulate erythropoiesis without excessively raising plasma EPO levels, HIF-PHIs have the potential to become therapeutic alternatives to conventional ESA therapy.

HIF-PHIs in clinical trials

Several HIF-PHIs have been developed and are currently undergoing clinical investigation in nondialysis-dependent (NDD) CKD patients, EPO-naïve dialysis-dependent (DD) patients, and DD patients who were previously treated with ESAs (Table 1). Peer-reviewed clinical data from randomized controlled trials are available for three orally administered compounds, daprodustat (GSK-1278863), roxadustat (FG-4592), and vadadustat (AKB-6548) all of which have progressed to phase 3 clinical evaluations (Table 2). Others are currently in preclinical, phase 1 or 2 clinical trials for renal anemia or are being evaluated for other indications: Molidustat (BAY 85-3934, Bayer)93; JTZ-951 (Japan Tobacco, Inc./Akros Pharma, Inc.)96; JNJ-42905343 (Janssen)99; DS-1093 (Daiichi Sankyo, Inc., no longer investigated for anemia); Zyan1 (Cadila Healthcare Ltd.).97,98 In addition to stimulating erythropoiesis, HIF-PHIs have been shown to produce nonerythropoietic effects, such as lowering cholesterol and reducing blood pressure.81,82,84,93,100

Table 1.

HIF-PHIs in clinical development for the treatment of anemia

Compound Ref. Development status HIF-a stabilization HIF-PHD targets
AKB-6548
(Vadadustat)
Akebia Therapeutics
86,88,89 Phase 3 HIF-2α > HIF-1α PHD3 > PHD2
GSK-1278863
(Daprodustat)
GlaxoSmithKline
81,9092 Phase 3 HIF-1α and HIF-2α PHD2 and PHD3
FG-4592
(Roxadustat)
Fibrogen/Astellas Pharma/AstraZeneca
82,85 Phase 3 HIF-1α and HIF-2α PHD1, 2 and 3
BAY 85-3934
(Molidustat)
Bayer Pharma
9395 Phase 2 HIF-1α and HIF-2α PHD2 > PHD1/PHD3
JTZ-951
Japan Tabacco, Inc./Akros Pharma, Inc.
96 Phase 2 Not published Not published
Zyan1
Cadila Healthcare
97,98 Phase 1 Not published Not published
JNJ-42905343
Janssen Pharmaceutica
99 Pre-clinical HIF-1α and HIF-2α PHD1, 2 and 3
DS-1093
Daiichi Sankyo, Inc.
Discontinued for anemia, under evaluation for other indications Not published Not published

The above table is based on a review of the literature and other publicly available resources such as patent applications. A direct comparison of half maximal inhibitory concentrations (IC50) of various compounds for HIF-PHDs, FIH, and other dioxygenases has not yet been published.

Table 2.

Summary of peer-reviewed clinical studies discussed in this review

Compound Ref. Pat Durat weeks Comp Fe suppl Hb Ferritin TIBC TSAT Hepcidin Chol VEGF
AKB-6548
Vadadustat
86 NDD 20 Placebo Oral Fe allowed + No change No effect No change
88,89
(Meeting abstracts)
DD convers. 16 None, 3 dose cohorts i.v. Fe permitted + n.r.$ n.r.$ n.r.$ n.r.$ n.r.$ n.r.$
GSK-1278863
Daprodustat
81 NDD 4 Placebo Not specified + * * No clear change#
DD 4 rhEPO Not specified +
5 mg only

(5 mg)
* No change (5 mg) No change (5 mg) No clear change#
FG-4592
Roxadustat
83 NDD 4 Placebo i.v. Fe prohibited, oral Fe allowed +
Dose-dep.
&
Also placebo
n.r. n.r.
85 DD
Incident patients
12 None One i.v., 2 oral and one no-iron cohort +
Iron-indep.
n.r n.r
84 NDD 16–24 None, 6-cohort dose-finding study i.v. Fe prohibited, oral Fe allowed +
Starting dose-dep.
↓, for BL Ferritin
≥100 ng/mL
n.r.
Initially
n.r.
82 DD
convers.
6 or 19 rhEPO Oral Fe allowed, i.v. Fe as rescue + No change
At 6 wks
No change
At 19 wks
n.r.

+ denotes effectiveness in changing or maintaining hemoglobin (Hb);

*

denotes trends, small, or statistically nonsignificant changes;

#

denotes that high interpatient variability was observed;

&

not statistically significant compared to placebo;

$

not reported in the meeting abstracts, manuscript containing iron data in preparation;

BL, base line; convers., conversion from ESA therapy to HIF-PHI; Comp, Comparator; n.r., not reported; Pat, either dialysis-dependent (DD) or nondialysis-dependent (NDD) CKD patients, incident patients are newly initiated EPO-naïve hemodialysis or peritoneal dialysis patients.

Daprodustat, Roxadustat, Vadadustat, and Molidustat are discussed in greater detail below. Vadadustat is the least potent inhibitor of PHD2, as the concentration needed to inhibit 50% of its activity in vitro (IC50) is the highest among the four compounds.

Daprodustat (GSK-1278863)

Daprodustat was developed by GlaxoSmithKline as an oral, once-daily HIF-stabilizing agent and is currently in phase 3 clinical trials for renal anemia. It contains a characteristic carbonylglycine side chain that is structurally analogous to 2OG. Preclinical studies demonstrated that daprodustat inhibits PHD2 and PHD3 leading to both HIF-1α and HIF-2α stabilization.101 Holdstock et al. conducted a phase 2a study in 72 CKD patients not undergoing dialysis and not receiving rhEPO, and in 82 patients on hemodialysis previously treated with stable doses of rhEPO.81 Participants in this 4-week study were randomized to either receive 0.5, 2, or 5 mg of once-daily daprodustat. Oral administration of daprodustat led to a dose-dependent increase in hemoglobin levels. In the NDD study group, a hemoglobin increase of 0.95 g/dL was observed with 5 mg daprodustat compared to a hemoglobin reduction of 0.23 g/dL in the placebo group. In the DD group, 5 mg of daprodustat was required to maintain hemoglobin levels in target range, whereas 0.5 and 2 mg were not effective. Median peak plasma EPO concentrations in the HIF-PHI-treated groups were much lower compared to DD patients receiving rhEPO (424.9.8 U/L vs. 24.7 U/L in the 5 mg DD group). Daprodustat treatment increased TIBC in both NDD and DD groups, whereas transferrin saturation (TSAT) was decreased in NDD patients. Hepcidin decreased in NDD patients in a dose-dependent manner, whereas no change (<0.5%) was observed in the DD group treated with 5 mg of daprodustat compared to the rhEPO control group. In addition to the effects on erythropoiesis and iron metabolism, Holdstock et al. reported dose-dependent decreases in total cholesterol, HDL-, and directly-measured LDL-cholesterol levels. Overall, the most common reported side effect was nausea in the NDD group and anemia in the DD arm (0.5 and 2.0 mg groups). Serious adverse events were reported in five patients of the NDD cohort (hypoglycemia, acute pancreatitis, acute renal failure, and azotemia) and four patients in the DD group (abnormal liver function tests, acute respiratory failure, constipation, and gastrointestinal hemorrhage). Deaths were not reported. Brigandi et al. reported a dose-dependent increase in plasma EPO and hemoglobin levels and a dose-dependent decrease in plasma hepcidin in a separate phase 2a study (daprodustat dose range from 10 to 100 mg).90 The most common side effect in this study was also nausea, which was mainly observed in the high dose cohorts (50 and 100 mg).

Roxadustat (FG-4592)

Roxadustat is FibroGen’s lead compound and has, in collaboration with Astellas Pharma and AstraZeneca, entered several large phase 3 cardiovascular outcomes trials. FG-4592 is structurally related to FG-2216, which is no longer used in humans due to a single case of fatal hepatic necrosis that was not attributed to the drug.102 FG-2216 and FG-4592 are both hydroxy-quinoline-based compounds that differ structurally from each other by the addition of a phenoxy-substituent to the quinoline core of FG-2216.

Four major clinical studies have been published with Roxadustat. In the first study, Besarab et al. investigated the effects of roxadustat in 116 NDD-CKD patients treated orally BIW or TIW for 28 days at doses ranging from 0.7 to 2.0 mg/kg. This study demonstrated a dose-dependent increase in hemoglobin and decrease in plasma hepcidin, ferritin and TSAT were decreased at the end of study.83

Provenzano et al. published two dose-finding studies in NDD-CKD patients and in long-term hemodialysis patients on rhEPO therapy.82,84 In the NDD study, 145 patients with hemoglobin ≤10.5 g/dL were enrolled and randomized into six different dosage regimen and treated with roxadustat for 16–24 weeks. 92% of roxadustat-treated patients responded with an increase in hemoglobin of 1 g/dL from baseline and hemoglobin level of ≥11.0 g/dL. This response was independent of baseline C-reactive protein levels and iron repletion status. Erythropoietic responses were associated with decreased hepcidin levels and iron supplementation was not needed to maintain hemoglobin. Furthermore, mean total cholesterol levels were reduced. Although 35 patients experienced serious adverse events in this study, including 5 deaths (2 cardiopulmonary arrests, one pulmonary embolism, one cerebral infarction with pulmonary edema, and one unwitnessed death), none of these events were considered to be drug-related.84 The second study was an active comparator study that consisted of two parts. In part 1, 54 hemodialysis patients were treated with roxadustat thrice-weekly for 6 weeks and compared to control patients treated with epoetin alfa. Part 2 consisted of a 19-week study of 90 patients randomized to 6 dosing groups and active comparator cohort. Intravenous iron was prohibited. Part 1 of the study showed a dose-dependent hemoglobin response and decrease in plasma hepcidin. In part 2 of the study, the average roxadustat dose requirement for maintaining hemoglobin ≥11.0 g/dL was ~1.7 mg/kg and hepcidin levels decreased by ~18%. Mean peak plasma EPO levels in patients receiving a mean roxadustat dose of 1.3 mg/kg were ~130 U/L compared to ~700 U/L in study participants receiving a median dose of 90 U/kg/wk of epoetin alfa.82

The fourth study by Besarab et al. investigated the efficacy of roxadustat treatment in EPO-naïve hemodialysis (n = 48) and peritoneal dialysis patients (n = 12).85 Sixty patients were treated with roxadustat for 12 weeks (mean weekly dose range of 4.0–4.3 mg/kg administered TIW) in conjunction with oral iron, intravenous iron, or without iron therapy; 12 peritoneal dialysis patients received oral iron therapy. The study reported that the hemoglobin response in the iron-treated group was larger than in the noniron group, that hepcidin levels decreased between 40% and 80% and that no correlation was found between hemoglobin responses and C-reactive protein level prior to study begin.

Vadadustat (AKB-6548)

AKB-6548 is Akebia’s lead compound. It is based on a hydroxy-pyridine core and contains a carbonylglycine side chain that is characteristic of other HIF-PHIs as well. AKB-6548/Vadadustat stabilizes HIF-2α to a greater extent than HIF-1α and produces a dose-dependent increase in plasma EPO levels.103 It maintains hemoglobin levels in CKD patients, increases TIBC, and lowers plasma ferritin and hepcidin level without significant changes in baseline plasma EPO levels at the doses studied.100,104 A slight decrease in mean arterial blood pressure was reported in a 28-day dose escalation study.100 Akebia recently published the results of a 20-week, double-blind, randomized, placebo-controlled, phase 2b study in patients with stages 3a to 5 NDD-CKD.86 The primary endpoint of this study was the percentage of patients who achieved or maintained a hemoglobin ≥11.0 g/dL or increased hemoglobin by ≥1.2 g/dL. The intent-to-treat study population consisted of 210 patients, 138 in the vadadustat, and 72 patients in the control group. Vadadustat was orally administered once-daily at a starting dose of 450 mg to ESA-naïve patients with hemoglobin ≤10.5 g/dL, patients who previously received ESA with hemoglobin ≤10.5 g/dL, or patients actively treated with ESA and hemoglobin ≥10.5 to ≤12.0 g/dL. Vadadustat was titrated to either 600 or 150 mg as per dosing algorithm and suspended for hemoglobin ≥13.0 g/dL. Vadadustat raised and maintained hemoglobin (primary endpoint) in 55% of patients compared to 10% in the placebo group. Hemoglobin excursions ≥13.0 g/dL occurred in 4% of patients. With regard to iron parameters, vadadustat decreased hepcidin, and ferritin levels and significantly increased TIBC. TSAT and serum iron were comparable between the vadadustat and the placebo group. VEGF levels were not different between the two groups, changes in total serum cholesterol, LDL-, and HDL-cholesterol or triglycerides were not reported. The overall incidence of adverse events was comparable between groups. Three deaths occurred in the vadadustat arm of the study; two deaths were considered as unrelated to the study medication and one death could not be further evaluated as it occurred at home and no autopsy was performed. Vadadustat was also evaluated in hemodialysis patients previously receiving ESAs (3 cohorts, starting doses of 300 mg QD, 450 mg QD, and 450 mg TIW). This 16-week open-label phase 2 study demonstrated that vadadustat maintained stabile hemoglobin levels in ESRD patients converted from injectable ESAs.88 A correlation between baseline C-reactive protein levels at study entry and response to treatment with vadadustat was not observed. No serious adverse events were assessed as being related to vadadustat and no deaths were reported.89 Because of these successful phase 2 trials, Akebia Therapeutics has recently launched two major phase 3 programs, PRO2TECT in NDD-CKD patients, and INNO2VATE to investigate vadadustat in DD-CKD patients

Molidustat (BAY 85-3934)

Bayer Healthcare compound 85-3934 (Molidustat) is structurally different from other HIF-PHIs in that it does not contain a carbonylglycine side chain. The compound is based on a dihydropyrazolone ring structure and has a moderate preference for PDH2. Two 16-week phase 2 studies have been completed, but are not yet published in peer-reviewed journals. Meeting abstracts indicate that molidustat effectively stimulated erythropoiesis in EPO-naïve NDD-CKD patients (fixed doses of 25, 50, or 75 mg once-daily, 25 or 50 mg BID) and NDD-CKD patients previously treated with ESAs (starting doses of 25, 50, and 75 mg with planned dose range between 15 and 150 mg; darbepoetin as active comparator).94,95 The data on iron metabolism are not yet reported for molidustat. In addition to its effect on erythropoiesis, molidustat displayed antihypertensive and cardioprotective effects in rats that underwent subtotal nephrectomy.93 Adverse events were relatively equally distributed between molidustat and control cohorts.

Potential adverse effects associated with HIF-PHI therapy

General considerations

The types of potential adverse effects associated with HIF-PHI therapy are frequently inferred from the analysis of genetically modified mice, which are characterized by the tissue-specific blockade or activation of HIF signaling. Although animal studies have provided important insights into cell- and context-specific HIF functions, the clinical consequences of systemic HIF activation in patients with CKD may be difficult to predict from these data. Genetic models with permanent HIF activation, e.g., as a result of VHL and PHD function loss, are often characterized by dramatic gene expression and functional changes, limiting their predictive value for clinical studies with short-acting HIF-PHIs that stabilize HIF-α transiently. Long-term pre-clinical studies in animals treated with HIF-PHIs would be more informative in this regard. In fact, one study assessed the effect of HIF-PHI roxadustat on tumor initiation, progression and metastasis in a VEGF-sensitive model of spontaneous breast cancer and did not report adverse outcomes.105 Important information regarding the type and likelihood of adverse effects can also be gained from the study of patients with genetic mutations in the PHD/HIF pathway. For example, patients with Chuvash polycythemia are homozygous for a specific VHL mutation (R200W) that predisposes to the development of polycythemia and vascular complications such as cerebral vascular events and peripheral thromboembolism, but not the classic VHL-associated tumors, such as CNS hemangioblastomas, renal cysts, and clear cell carcinomas or pheochromocytomas.106108

Adverse effects due to HIF activation

Although HIF-PHIs have been well tolerated in short-term clinical trials, there are several safety concerns relating to potential nonerythropoietic effects of repeated HIF activation on tissue homeostasis as HIF-1 and HIF-2 regulate a broad spectrum of cellular functions.109 The extent of nonerythropoietic HIF effects produced by HIF-PHI administration are likely to be determined by the degree and duration of HIF activation, cell types involved, and the pharmacokinetic characteristics of HIF-PHIs.

Prolonged activation of the HIF system has profound effects on metabolic and cardiovascular homeostasis.109 For example, HIF-2 has been implicated in the pathogenesis of pulmonary hypertension,109114 which raises concerns whether HIF-PHI therapy may adversely affect pulmonary artery pressure regulation in the CKD/ESRD patient population, which is characterized by a higher prevalence of pulmonary hypertension.115 With regard to systemic blood pressure regulation, HIF-PHI therapy seems to be associated with a trend toward decreasing rather than increasing blood pressure.93,100

HIF is also a potent stimulator of angiogenesis as vascular endothelial growth factor (VEGF) and other proangiogenic factors are bona fide transcriptional targets of HIF. Increases in tissue VEGF levels may have pro-oncogenic effects and could exacerbate pathologic conditions that are characterized by vascular proliferation, such as diabetic retinopathy. Notwithstanding these concerns, increased plasma VEGF concentrations were not found in several phase 2 clinical trials.81,86 However, it is unclear whether plasma VEGF levels alone can be used to assess these potential pro-angiogenic risks. The absence of elevated plasma VEGF levels may be due to relatively low levels of cellular HIF activation, which are sufficient to increase erythropoietic effects but not sufficient to increased VEGF expression. Daprodustat for example produces increases in plasma VEGF concentrations at doses ≥50 mg (up to 5 mg was used in the clinical studies published by Holdstock et al.).91,116

HIF has been shown to regulate glucose, fatty acid, cholesterol, and mitochondrial metabolism.109,117 Some of the metabolic consequences of HIF activation are also seen in patients treated with HIF-PHIs. A decrease in serum cholesterol levels was reported in NDD-CKD and hemodialysis patients for daprodustat and roxadustat.81,82,84 Furthermore, a trend for increased glucose was noted for daprodustat at high doses (50 and 100 mg).90 To what degree intermittent HIF activation affects glucose homeostasis and fatty acid metabolism in diabetics and nondiabetics is not known and will need to be determined in long-term clinical trials. Preclinical studies in animal models indicate that the PHD/HIF axis plays an important role in pancreatic β-cell function.118 This notion is consistent with genome-wide mRNA expression analysis, which showed that the HIF-1β subunit was expressed at very low levels in pancreatic islets isolated from patients with type II diabetes.119

Safety concerns also exist with regard to HIF’s role in the regulation of tumor cell growth and metastasis, as HIF signaling in malignant cells has been associated with tumor initiation and progression.120

Due to the short duration of phase 2 trials, the effects of HIF-PHI therapy on CKD progression and matrix turnover are not yet known. The role of HIF in CKD progression is controversial and long-term clinical studies are needed to address this issue.121,122 A recent study in animal models investigated the role of hypoxia and HIF-1 signaling in the development of vascular calcifications. The study suggested that HIF-PHIs might enhance phosphate-induced vascular smooth muscle cell calcification in CKD or ESRD patients with poorly controlled mineral bone metabolism.123 A molecular link also exists between HIF and FGF23 signaling, which may negatively impact cardiovascular risk in HIF-PHI-treated CKD patients.124,125 Another potential adverse effect that may be observed in CKD patients on HIF-PHI therapy is hepatic toxicity. FG2216, the sister compound of roxadustat was taken out of clinical trials due to one case of fatal hepatic necrosis, although the death of the patient was not attributed to the study medication. To date, HIF-PHIs have been well tolerated and significant liver toxicity has not been reported in published peer-reviewed studies.

Adverse effects not related to HIF activation

In addition to HIF-mediated adverse effects, HIF-PHIs have the potential to interfere with cell signaling pathways that involve other non-HIF proline hydroxylation targets, non-HIF dioxygenases, or nonspecific effects that are not related to HIF signaling or inhibition of 2OG-dioxygenases. However, meeting reports suggest that HIF-PHIs daprodustat, roxadustat, vadadustat, and molidustat are relatively specific for HIF-PHDs and have little effect on factor inhibiting HIF (FIH) and dioxygenases involved in epigenetic gene regulation. FIH is an asparaginyl hydroxylase that modulates co-factor recruitment to the HIF transcriptional complex.126128

In summary, notwithstanding the very promising performance of HIF-PHI therapy in short-term phase 2 clinical trials, more comprehensive physiologic studies and large long-term clinical trials are needed to better understand the functional and metabolic consequences of systemic HIF-PHI administration before HIF-activating compounds can be safely given to patients with CKD or ESRD for extended periods of time.

CONCLUDING REMARKS

Here, I have provided a brief and concise overview of oxygen-dependent mechanisms that regulate erythropoiesis and iron metabolism in the context of renal anemia. The discovery of the PHD dioxygenases as key regulators of HIF signaling and erythropoiesis has led to the development of novel therapeutic agents that harness hypoxia responses for therapeutic gain. These HIF-activating compounds are currently in phase 3 clinical development for the treatment of renal anemia and have the potential to significantly impact the clinical practice of Nephrology.

Acknowledgments

VHH holds the Krick-Brooks chair in Nephrology at Vanderbilt University and is supported by NIH grants R01-DK101791 and R01-DK081646, and a Department of Veterans Affairs Merit Award 1I01BX002348. The author thanks Andres A. Urrutia and Pinelopi P. Kapitsinou for the critical reading of the manuscript. The author apologizes to those colleagues whose work could not be cited due to space limitations.

Disclosure of grants or other funding: The author is supported by grants from the National Institutes of Health and the Department of Veterans Affairs.

Footnotes

Conflict of Interest: VHH serves on the scientific advisory board of Akebia Therapeutics, Inc., a company that develops PHD inhibitors for the treatment of anemia.

References

  • 1.Jourdanet D. Influence de la Pression de L’air sur la vie de L’homme. Paris: G Masson; 1875. [Google Scholar]
  • 2.Bert P. La Pression Barométrique Recherches de Physiologie Exp erimentale. Paris: G Masson; 1878. [Google Scholar]
  • 3.Bert P. Sur la richesse en hemoglobine du sang des animaux vivant sur les hauts lieux. C R Acad Sci Paris. 1882;94:805–807. [Google Scholar]
  • 4.Miyake T, Kung CK, Goldwasser E. Purification of human erythropoietin. J Biol Chem. 1977;252:5558–5564. [PubMed] [Google Scholar]
  • 5.Jacobs K, Shoemaker C, Rudersdorf R, et al. Isolation and characterization of genomic and cDNA clones of human erythropoietin. Nature. 1985;313:806–810. doi: 10.1038/313806a0. [DOI] [PubMed] [Google Scholar]
  • 6.Lin FK, Suggs S, Lin CH, et al. Cloning and expression of the human erythropoietin gene. Proc Natl Acad Sci USA. 1985;82:7580–7584. doi: 10.1073/pnas.82.22.7580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Semenza GL, Wang GL. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol. 1992;12:5447–5454. doi: 10.1128/mcb.12.12.5447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Wang GL, Semenza GL. Purification and characterization of hypoxia-inducible factor 1. J Biol Chem. 1995;270:1230–1237. doi: 10.1074/jbc.270.3.1230. [DOI] [PubMed] [Google Scholar]
  • 9.Wang GL, Jiang BH, Rue EA, Semenza GL. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci USA. 1995;92:5510–5514. doi: 10.1073/pnas.92.12.5510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Forsythe JA, Jiang BH, Iyer NV, et al. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol. 1996;16:4604–4613. doi: 10.1128/mcb.16.9.4604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Maxwell PH, Pugh CW, Ratcliffe PJ. Inducible operation of the erythropoietin 3′ enhancer in multiple cell lines: Evidence for a widespread oxygen-sensing mechanism. Proc Natl Acad Sci USA. 1993;90:2423–2427. doi: 10.1073/pnas.90.6.2423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Jaakkola P, Mole DR, Tian YM, et al. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science. 2001;292:468–472. doi: 10.1126/science.1059796. [DOI] [PubMed] [Google Scholar]
  • 13.Ivan M, Kondo K, Yang H, et al. HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: Implications for O2 sensing. Science. 2001;292:464–468. doi: 10.1126/science.1059817. [DOI] [PubMed] [Google Scholar]
  • 14.Epstein AC, Gleadle JM, McNeill LA, et al. C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell. 2001;107:43–54. doi: 10.1016/s0092-8674(01)00507-4. [DOI] [PubMed] [Google Scholar]
  • 15.Kewley RJ, Whitelaw ML, Chapman-Smith A. The mammalian basic helix-loop-helix/PAS family of transcriptional regulators. Int J Biochem Cell Biol. 2004;36:189–204. doi: 10.1016/s1357-2725(03)00211-5. [DOI] [PubMed] [Google Scholar]
  • 16.Semenza GL. Regulation of mammalian O2 homeostasis by hypoxia-inducible factor 1. Annu Rev Cell Dev Biol. 1999;15:551–578. doi: 10.1146/annurev.cellbio.15.1.551. [DOI] [PubMed] [Google Scholar]
  • 17.Wenger RH, Stiehl DP, Camenisch G. Integration of oxygen signaling at the consensus HRE. Sci STKE. 2005;2005:re12. doi: 10.1126/stke.3062005re12. [DOI] [PubMed] [Google Scholar]
  • 18.Semenza GL. HIF-1 and mechanisms of hypoxia sensing. Curr Opin Cell Biol. 2001;13:167–171. doi: 10.1016/s0955-0674(00)00194-0. [DOI] [PubMed] [Google Scholar]
  • 19.Schodel J, Oikonomopoulos S, Ragoussis J, Pugh CW, Ratcliffe PJ, Mole DR. High-resolution genome-wide mapping of HIF-binding sites by ChIP-seq. Blood. 2011;117:e207–e217. doi: 10.1182/blood-2010-10-314427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hu CJ, Wang LY, Chodosh LA, Keith B, Simon MC. Differential roles of hypoxia-inducible factor 1alpha (HIF-1alpha) and HIF-2alpha in hypoxic gene regulation. Mol Cell Biol. 2003;23:9361–9374. doi: 10.1128/MCB.23.24.9361-9374.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Morita M, Ohneda O, Yamashita T, et al. HLF/HIF-2alpha is a key factor in retinopathy of prematurity in association with erythropoietin. Embo J. 2003;22:1134–1146. doi: 10.1093/emboj/cdg117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Scortegagna M, Ding K, Zhang Q, et al. HIF-2alpha regulates murine hematopoietic development in an erythropoietin-dependent manner. Blood. 2005;105:3133–3140. doi: 10.1182/blood-2004-05-1695. [DOI] [PubMed] [Google Scholar]
  • 23.Gruber M, Hu CJ, Johnson RS, Brown EJ, Keith B, Simon MC. Acute postnatal ablation of Hif-2alpha results in anemia. Proc Natl Acad Sci USA. 2007;104:2301–2306. doi: 10.1073/pnas.0608382104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Rankin EB, Biju MP, Liu Q, et al. Hypoxia-inducible factor-2 (HIF-2) regulates hepatic erythropoietin in vivo. J Clin Invest. 2007;117:1068–1077. doi: 10.1172/JCI30117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kapitsinou PP, Liu Q, Unger TL, et al. Hepatic HIF-2 regulates erythropoietic responses to hypoxia in renal anemia. Blood. 2010;116:3039–3048. doi: 10.1182/blood-2010-02-270322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mastrogiannaki M, Matak P, Keith B, Simon MC, Vaulont S, Peyssonnaux C. HIF-2alpha, but not HIF-1alpha, promotes iron absorption in mice. J Clin Invest. 2009;119:1159–1166. doi: 10.1172/JCI38499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bernhardt WM, Wiesener MS, Scigalla P, et al. Inhibition of prolyl hydroxylases increases erythropoietin production in ESRD. J Am Soc Nephrol. 2010;21:2151–2156. doi: 10.1681/ASN.2010010116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Maxwell PH, Wiesener MS, Chang GW, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis [see comments] Nature. 1999;399:271–275. doi: 10.1038/20459. [DOI] [PubMed] [Google Scholar]
  • 29.Maynard MA, Qi H, Chung J, et al. Multiple splice variants of the human HIF-3alpha locus are targets of the VHL E3 ubiquitin ligase complex. J Biol Chem. 2003;278:1032–1040. doi: 10.1074/jbc.M208681200. [DOI] [PubMed] [Google Scholar]
  • 30.Kaelin WG. Von Hippel-Lindau disease. Annu Rev Pathol. 2007;2:145–173. doi: 10.1146/annurev.pathol.2.010506.092049. [DOI] [PubMed] [Google Scholar]
  • 31.Lee FS, Percy MJ. The HIF pathway and erythrocytosis. Annu Rev Pathol. 2011;6:165–192. doi: 10.1146/annurev-pathol-011110-130321. [DOI] [PubMed] [Google Scholar]
  • 32.Kaelin WG, Jr, Ratcliffe PJ. Oxygen sensing by metazoans: The central role of the HIF hydroxylase pathway. Mol Cell. 2008;30:393–402. doi: 10.1016/j.molcel.2008.04.009. [DOI] [PubMed] [Google Scholar]
  • 33.Loenarz C, Schofield CJ. Expanding chemical biology of 2-oxoglutarate oxygenases. Nat Chem Biol. 2008;4:152–156. doi: 10.1038/nchembio0308-152. [DOI] [PubMed] [Google Scholar]
  • 34.Pollard PJ, Briere JJ, Alam NA, et al. Accumulation of Krebs cycle intermediates and over-expression of HIF1alpha in tumours which result from germline FH and SDH mutations. Hum Mol Genet. 2005;14:2231–2239. doi: 10.1093/hmg/ddi227. [DOI] [PubMed] [Google Scholar]
  • 35.Isaacs JS, Jung YJ, Mole DR, et al. HIF overexpression correlates with biallelic loss of fumarate hydratase in renal cancer: Novel role of fumarate in regulation of HIF stability. Cancer Cell. 2005;8:143–153. doi: 10.1016/j.ccr.2005.06.017. [DOI] [PubMed] [Google Scholar]
  • 36.Koury MJ, Haase VH. Anaemia in kidney disease: harnessing hypoxia responses for therapy. Nat Rev Neph-rol. 2015;11:394–410. doi: 10.1038/nrneph.2015.82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Obara N, Suzuki N, Kim K, Nagasawa T, Imagawa S, Yamamoto M. Repression via the GATA box is essential for tissue-specific erythropoietin gene expression. Blood. 2008;111:5223–5232. doi: 10.1182/blood-2007-10-115857. [DOI] [PubMed] [Google Scholar]
  • 38.Kobayashi H, Liu Q, Binns TC, et al. Distinct subpopulations of FOXD1 stroma-derived cells regulate renal erythropoietin. J Clin Invest. 2016;126:1926–1938. doi: 10.1172/JCI83551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Haase VH. Regulation of erythropoiesis by hypoxia-inducible factors. Blood Rev. 2013;27:41–53. doi: 10.1016/j.blre.2012.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Urrutia AA, Afzal A, Nelson J, Davidoff O, Gross KW, Haase VH. Prolyl-4-hydroxylase 2 and 3 coregulate murine erythropoietin in brain pericytes. Blood. 2016;128:2550–2560. doi: 10.1182/blood-2016-05-713545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Rankin EB, Rha J, Selak MA, et al. Hypoxia-inducible factor 2 regulates hepatic lipid metabolism. Mol Cell Biol. 2009;29:4527–4538. doi: 10.1128/MCB.00200-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Minamishima YA, Kaelin WG., Jr Reactivation of hepatic EPO synthesis in mice after PHD loss. Science. 2010;329:407. doi: 10.1126/science.1192811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lonnberg M, Garle M, Lonnberg L, Birgegard G. Patients with anaemia can shift from kidney to liver production of erythropoietin as shown by glycoform analysis. J Pharm Biomed Anal. 2013;81–82:187–192. doi: 10.1016/j.jpba.2013.04.009. [DOI] [PubMed] [Google Scholar]
  • 44.Shah YM, Matsubara T, Ito S, Yim SH, Gonzalez FJ. Intestinal hypoxia-inducible transcription factors are essential for iron absorption following iron deficiency. Cell Metab. 2009;9:152–164. doi: 10.1016/j.cmet.2008.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mastrogiannaki M, Matak P, Delga S, Deschemin JC, Vaulont S, Peyssonnaux C. Deletion of HIF-2alpha in the enterocytes decreases the severity of tissue iron loading in hepcidin knockout mice. Blood. 2011;119:587–590. doi: 10.1182/blood-2011-09-380337. [DOI] [PubMed] [Google Scholar]
  • 46.Rolfs A, Kvietikova I, Gassmann M, Wenger RH. Oxygen-regulated transferrin expression is mediated by hypoxia-inducible factor-1. J Biol Chem. 1997;272:20055–20062. doi: 10.1074/jbc.272.32.20055. [DOI] [PubMed] [Google Scholar]
  • 47.Lok CN, Ponka P. Identification of a hypoxia response element in the transferrin receptor gene. J Biol Chem. 1999;274:24147–24152. doi: 10.1074/jbc.274.34.24147. [DOI] [PubMed] [Google Scholar]
  • 48.Tacchini L, Bianchi L, Bernelli-Zazzera A, Cairo G. Transferrin receptor induction by hypoxia. HIF-1-mediated transcriptional activation and cell-specific post-transcriptional regulation. J Biol Chem. 1999;274:24142–24146. doi: 10.1074/jbc.274.34.24142. [DOI] [PubMed] [Google Scholar]
  • 49.Mukhopadhyay CK, Mazumder B, Fox PL. Role of hypoxia-inducible factor-1 in transcriptional activation of ceruloplasmin by iron deficiency. J Biol Chem. 2000;275:21048–21054. doi: 10.1074/jbc.M000636200. [DOI] [PubMed] [Google Scholar]
  • 50.Lee PJ, Jiang BH, Chin BY, et al. Hypoxia-inducible factor-1 mediates transcriptional activation of the heme oxygenase-1 gene in response to hypoxia. J Biol Chem. 1997;272:5375–5381. [PubMed] [Google Scholar]
  • 51.Taylor M, Qu A, Anderson ER, et al. Hypoxia-inducible factor-2alpha mediates the adaptive increase of intestinal ferroportin during iron deficiency in mice. Gastroenterology. 2011;140:2044–2055. doi: 10.1053/j.gastro.2011.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ganz T. Molecular control of iron transport. J Am Soc Nephrol. 2007;18:394–400. doi: 10.1681/ASN.2006070802. [DOI] [PubMed] [Google Scholar]
  • 53.Zaritsky J, Young B, Wang HJ, et al. Hepcidin–a potential novel biomarker for iron status in chronic kidney disease. Clin J Am Soc Nephrol. 2009;4:1051–1056. doi: 10.2215/CJN.05931108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Nicolas G, Chauvet C, Viatte L, et al. The gene encoding the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation. J Clin Invest. 2002;110:1037–1044. doi: 10.1172/JCI15686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Gordeuk VR, Miasnikova GY, Sergueeva AI, et al. Chuvash polycythemia VHLR200W mutation is associated with down-regulation of hepcidin expression. Blood. 2011;118:5278–5282. doi: 10.1182/blood-2011-03-345512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Peyssonnaux C, Zinkernagel AS, Schuepbach RA, et al. Regulation of iron homeostasis by the hypoxia-inducible transcription factors (HIFs) J Clin Invest. 2007;117:1926–1932. doi: 10.1172/JCI31370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Mastrogiannaki M, Matak P, Mathieu JR, et al. Hepatic hypoxia-inducible factor-2 down-regulates hepcidin expression in mice through an erythropoietin-mediated increase in erythropoiesis. Haematologica. 2012;97:827–834. doi: 10.3324/haematol.2011.056119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Liu Q, Davidoff O, Niss K, Haase VH. Hypoxia-inducible factor regulates hepcidin via erythropoietin-induced erythropoiesis. J Clin Invest. 2012;122:4635–4644. doi: 10.1172/JCI63924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Sanchez M, Galy B, Muckenthaler MU, Hentze MW. Iron-regulatory proteins limit hypoxia-inducible factor-2alpha expression in iron deficiency. Nat Struct Mol Biol. 2007;14:420–426. doi: 10.1038/nsmb1222. [DOI] [PubMed] [Google Scholar]
  • 60.Ghosh MC, Zhang DL, Jeong SY, et al. Deletion of iron regulatory protein 1 causes polycythemia and pulmonary hypertension in mice through translational derepression of HIF2alpha. Cell Metab. 2013;17:271–281. doi: 10.1016/j.cmet.2012.12.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Anderson SA, Nizzi CP, Chang YI, et al. The IRP1-HIF-2alpha axis coordinates iron and oxygen sensing with erythropoiesis and iron absorption. Cell Metab. 2013;17:282–290. doi: 10.1016/j.cmet.2013.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wilkinson N, Pantopoulos K. IRP1 regulates erythropoiesis and systemic iron homeostasis by controlling HIF2alpha mRNA translation. Blood. 2013;122:1658–1668. doi: 10.1182/blood-2013-03-492454. [DOI] [PubMed] [Google Scholar]
  • 63.Chin K, Yu X, Beleslin-Cokic B, et al. Production and processing of erythropoietin receptor transcripts in brain. Brain Res Mol Brain Res. 2000;81:29–42. doi: 10.1016/s0169-328x(00)00157-1. [DOI] [PubMed] [Google Scholar]
  • 64.Manalo DJ, Rowan A, Lavoie T, et al. Transcriptional regulation of vascular endothelial cell responses to hypoxia by HIF-1. Blood. 2005;105:659–669. doi: 10.1182/blood-2004-07-2958. [DOI] [PubMed] [Google Scholar]
  • 65.Yoon D, Pastore YD, Divoky V, et al. HIF-1alpha -deficiency results in dysregulated EPO signaling and iron homeostasis in mouse development. J Biol Chem. 2006;281:25703–25711. doi: 10.1074/jbc.M602329200. [DOI] [PubMed] [Google Scholar]
  • 66.Liu YL, Ang SO, Weigent DA, Prchal JT, Bloomer JR. Regulation of ferrochelatase gene expression by hypoxia. Life Sci. 2004;75:2035–2043. doi: 10.1016/j.lfs.2004.03.027. [DOI] [PubMed] [Google Scholar]
  • 67.Hofer T, Wenger RH, Kramer MF, Ferreira GC, Gassmann M. Hypoxic up-regulation of erythroid 5-aminolevulinate synthase. Blood. 2003;101:348–350. doi: 10.1182/blood-2002-03-0773. [DOI] [PubMed] [Google Scholar]
  • 68.Adelman DM, Maltepe E, Simon MC. Multilineage embryonic hematopoiesis requires hypoxic ARNT activity. Genes Dev. 1999;13:2478–2483. doi: 10.1101/gad.13.19.2478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Yamashita T, Ohneda O, Sakiyama A, Iwata F, Ohneda K, Fujii-Kuriyama Y. The microenvironment for erythropoiesis is regulated by HIF-2alpha through VCAM-1 in endothelial cells. Blood. 2008;112:1482–1492. doi: 10.1182/blood-2007-11-122648. [DOI] [PubMed] [Google Scholar]
  • 70.Fishbane S, Nissenson AR. Anemia management in chronic kidney disease. Kidney Int. 2010;117:S3–S9. doi: 10.1038/ki.2010.188. [DOI] [PubMed] [Google Scholar]
  • 71.Jelkmann W. Erythropoietin after a century of research: younger than ever. Eur J Haematol. 2007;78:183–205. doi: 10.1111/j.1600-0609.2007.00818.x. [DOI] [PubMed] [Google Scholar]
  • 72.Jelkmann W. The ESA scenario gets complex: from biosimilar epoetins to activin traps. Nephrol Dial Transplant. 2015;30:553–559. doi: 10.1093/ndt/gfu089. [DOI] [PubMed] [Google Scholar]
  • 73.Besarab A, Bolton WK, Browne JK, et al. The effects of normal as compared with low hematocrit values in patients with cardiac disease who are receiving hemodialysis and epoetin. N Engl J Med. 1998;339:584–590. doi: 10.1056/NEJM199808273390903. [DOI] [PubMed] [Google Scholar]
  • 74.Singh AK, Szczech L, Tang KL, et al. Correction of anemia with epoetin alfa in chronic kidney disease. N Engl J Med. 2006;355:2085–2098. doi: 10.1056/NEJMoa065485. [DOI] [PubMed] [Google Scholar]
  • 75.Pfeffer MA, Burdmann EA, Chen CY, et al. A trial of darbepoetin alfa in type 2 diabetes and chronic kidney disease. N Engl J Med. 2009;361:2019–2032. doi: 10.1056/NEJMoa0907845. [DOI] [PubMed] [Google Scholar]
  • 76.Vaziri ND, Zhou XJ. Potential mechanisms of adverse outcomes in trials of anemia correction with erythropoietin in chronic kidney disease. Nephrol Dial Trans-plant. 2009;24:1082–1088. doi: 10.1093/ndt/gfn601. [DOI] [PubMed] [Google Scholar]
  • 77.McCullough PA, Barnhart HX, Inrig JK, et al. Cardiovascular toxicity of epoetin-alfa in patients with chronic kidney disease. Am J Nephrol. 2013;37:549–558. doi: 10.1159/000351175. [DOI] [PubMed] [Google Scholar]
  • 78.Besarab A, Frinak S, Yee J. What is so bad about a hemoglobin level of 12 to 13 g/dL for chronic kidney disease patients anyway? Adv Chronic Kidney Dis. 2009;16:131–142. doi: 10.1053/j.ackd.2008.12.007. [DOI] [PubMed] [Google Scholar]
  • 79.Solomon SD, Uno H, Lewis EF, et al. Erythropoietic response and outcomes in kidney disease and type 2 diabetes. N Engl J Med. 2010;363:1146–1155. doi: 10.1056/NEJMoa1005109. [DOI] [PubMed] [Google Scholar]
  • 80.Szczech LA, Barnhart HX, Inrig JK, et al. Secondary analysis of the CHOIR trial epoetin-alpha dose and achieved hemoglobin outcomes. Kidney Int. 2008;74:791–798. doi: 10.1038/ki.2008.295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Holdstock L, Meadowcroft AM, Maier R, et al. Four-week studies of oral hypoxia-inducible factor-prolyl hydroxylase inhibitor GSK1278863 for treatment of anemia. J Am Soc Nephrol. 2016;27:1234–1244. doi: 10.1681/ASN.2014111139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Provenzano R, Besarab A, Wright S, et al. Roxadustat (FG-4592) versus epoetin alfa for anemia in patients receiving maintenance hemodialysis: A phase 2, randomized, 6- to 19-week, open-Label, active-comparator, dose-ranging, safety and exploratory efficacy study. Am J Kidney Dis. 2016;67:912–924. doi: 10.1053/j.ajkd.2015.12.020. [DOI] [PubMed] [Google Scholar]
  • 83.Besarab A, Provenzano R, Hertel J, et al. Randomized placebo-controlled dose-ranging and pharmacodynamics study of roxadustat (FG-4592) to treat anemia in nondialysis-dependent chronic kidney disease (NDD-CKD) patients. Nephrol Dial Transplant. 2015;30:1665–1673. doi: 10.1093/ndt/gfv302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Provenzano R, Besarab A, Sun CH, et al. Oral hypoxia-inducible factor prolyl hydroxylase inhibitor roxadustat (FG-4592) for the treatment of anemia in patients with CKD. Clin J Am Soc Nephrol. 2016;11:982–991. doi: 10.2215/CJN.06890615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Besarab A, Chernyavskaya E, Motylev I, et al. Roxadustat (FG-4592): Correction of anemia in incident dialysis patients. J Am Soc Nephrol. 2016;27:1225–1233. doi: 10.1681/ASN.2015030241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Pergola PE, Spinowitz BS, Hartman CS, Maroni BJ, Haase VH. Vadadustat, a novel oral HIF stabilizer, provides effective anemia treatment in nondialysis-dependent chronic kidney disease. Kidney Int. 2016;90:1115–1122. doi: 10.1016/j.kint.2016.07.019. [DOI] [PubMed] [Google Scholar]
  • 87.Souma T, Nezu M, Nakano D, et al. Erythropoietin synthesis in renal myofibroblasts is restored by activation of hypoxia signaling. J Am Soc Nephrol. 2016;27:428–438. doi: 10.1681/ASN.2014121184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Haase VH, Hartman CS, Maroni B, Farzaneh-Far R, McCullough PA. Vadadustat—A novel, oral treatment for anemia of CKD—Maintains stable hemoglobin levels in dialysis patients converting from Erythropoiesis-Stimulating Agent (ESA). National Kidney Foundation Spring Clinical Meeting; Boston, MA. 2016. Abstract 171. [Google Scholar]
  • 89.Haase VH, Khawaja Z, Chan J, et al. Vadadustat maintains Hemoglobin (Hb) levels in Dialysis-Dependent Chronic Kidney Disease (DD-CKD) patients independent of systemic inflammation or prior dose of Erythropoiesis-Stimulating Agent (ESA) J Am Soc Nephrol. 2016;27:318A. [Google Scholar]
  • 90.Brigandi RA, Johnson B, Oei C, et al. A novel hypoxia-inducible factor-prolyl hydroxylase inhibitor (GSK1278863) for anemia in CKD: A 28-day, phase 2A randomized trial. Am J Kidney Dis. 2016;67:861–871. doi: 10.1053/j.ajkd.2015.11.021. [DOI] [PubMed] [Google Scholar]
  • 91.Hara K, Takahashi N, Wakamatsu A, Caltabiano S. Pharmacokinetics, pharmacodynamics and safety of single, oral doses of GSK1278863, a novel HIF-prolyl hydroxylase inhibitor, in healthy Japanese and Caucasian subjects. Drug Metab Pharmacokinet. 2015;30:410–418. doi: 10.1016/j.dmpk.2015.08.004. [DOI] [PubMed] [Google Scholar]
  • 92.Akizawa T, Tsubakihara Y, Nangaku M, et al. Effects of Daprodustat, a novel hypoxia-inducible factor prolyl hydroxylase inhibitor on anemia management in Japanese hemodialysis subjects. Am J Nephrol. 2017;45:127–135. doi: 10.1159/000454818. [DOI] [PubMed] [Google Scholar]
  • 93.Flamme I, Oehme F, Ellinghaus P, Jeske M, Keldenich J, Thuss U. Mimicking hypoxia to treat anemia: HIF-stabilizer BAY 85-3934 (Molidustat) stimulates erythropoietin production without hypertensive effects. PLoS One. 2014;9:e111838. doi: 10.1371/journal.pone.0111838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.MacDougall IC, Akizawa T, Berns J, Lentini S, Bernhardt T. Molidustat increases hemoglobin in erythropoiesis stimulating agents (ESA)—Naive patients with chronic kidney diseases not on dialysis (CKD-ND) Nephrol Dial Transplant. 2016;31(Suppl 1):i16. [Google Scholar]
  • 95.MacDougall IC, Akizawa T, Berns J, Bernhardt T, Krüger T. Safety and efficacy of molidustat in erythropoiesis stimulating agenst (ESA) pre-treated anemic patients with chronic kidney disease not on dialysis (CKD-ND) Nephrol Dial Transplant. 2016;31(Suppl 1):i193. [Google Scholar]
  • 96.Akizawa T, Hanaki K, Arai M. JTZ-591, an oral novel HIF-PHD inhibitor, elevates hemoglobin in Japanese anemic patients with chronic kidney disease not on dialysis. Nephrol Dial Transplant. 2015;30(Suppl 3):iii8–iii10. [Google Scholar]
  • 97.Patel H, Joharapurkar AA, Pandya VB, et al. Influence of acute and chronic kidney failure in rats on the disposition and pharmacokinetics of ZYAN1, a novel prolyl hydroxylase inhibitor, for the treatment of chronic kidney disease-induced anemia. Xenobiotica. 2017;19:1–8. doi: 10.1080/00498254.2016.1278287. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 98.Jain MR, Joharapurkar AA, Pandya V, et al. Pharmacological characterization of ZYAN1, a novel prolyl hydroxylase inhibitor for the treatment of anemia. Drug Res (Stuttg) 2016;66:107–112. doi: 10.1055/s-0035-1554630. [DOI] [PubMed] [Google Scholar]
  • 99.Barrett TD, Palomino HL, Brondstetter TI, et al. Prolyl hydroxylase inhibition corrects functional iron deficiency and inflammation-induced anaemia in rats. Br J Pharmacol. 2015;172:4078–4088. doi: 10.1111/bph.13188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Hartman C, Smith MT, Flinn C, et al. AKB-6548, a new hypoxia-inducible factor prolyl hydroxylase inhibitor increases hemoglobin while decreasing ferritin in a 28-day, phase 2a dose escalation study in stage 3 and 4 chronic kidney disease patients with anemia. J Am Soc Nephrol. 2011;22:435A. [Google Scholar]
  • 101.Brigandi RA, Ariazi JL, Duffy KJ, Luo L, Adams DF, Erickson-Miller CL. The prolyl hydroxylase inhibitor, GSK1278863A, induced EPO in vitro and efficient erythropoiesis leading to increased hemoglobin in vivo. J Am Soc Nephrol. 2010;21:722A. [Google Scholar]
  • 102.Astellas Pharma Inc. News release: the FDA accepts the complete response for clinical holds of FG-2216*/FG-4592 for the treatment of anemia. astellas.-com [online] 2008 http://www.astellas.com/en/corporate/news/pdf/080402_eg.pdf.
  • 103.Shalwitz R, Hartman C, Flinn C, Shalwitz I, Logan DK. AKB-6548, a novel hypoxia-inducible factor prolyl hydroxylase inhibitor reduces hepcidin and ferritin while it increases reticulocyte production and total iron binding capacity in healthy adults. J Am Soc Nephrol. 2011;22:45A. [Google Scholar]
  • 104.Shalwitz R, Hartman C, Flinn C, et al. AKB-6548: A new hypoxia-inducible factor prolyl hydroxylase inhibitor, increases hemoglobin in chronic kidney disease patients without increasing basal erythropoietin levels. J Am Soc Nephrol. 2012;23:56A. [Google Scholar]
  • 105.Seeley TW, Sternlicht MD, Klaus SJ, Neff TB, Liu DY. Induction of erythropoiesis by hypoxia-inducible factor prolyl hydroxylase inhibitors without promotion of tumor initiation, progression, or metastasis in a VEGF-sensitive model of spontaneous breast cancer. Hypoxia. 2017;5:1–9. doi: 10.2147/HP.S130526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Gordeuk VR, Sergueeva AI, Miasnikova GY, et al. Congenital disorder of oxygen sensing: Association of the homozygous Chuvash polycythemia VHL mutation with thrombosis and vascular abnormalities but not tumors. Blood. 2004;103:3924–3932. doi: 10.1182/blood-2003-07-2535. [DOI] [PubMed] [Google Scholar]
  • 107.Gordeuk VR, Prchal JT. Vascular complications in Chuvash polycythemia. Semin Thromb Hemost. 2006;32:289–294. doi: 10.1055/s-2006-939441. [DOI] [PubMed] [Google Scholar]
  • 108.Formenti F, Beer PA, Croft QP, et al. Cardiopulmonary function in two human disorders of the hypoxia-inducible factor (HIF) pathway: Von Hippel-Lindau disease and HIF-2{alpha} gain-of-function mutation. FASEB J. 2011;25:2001–2011. doi: 10.1096/fj.10-177378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Semenza GL. Oxygen sensing, hypoxia-inducible factors, and disease pathophysiology. Annu Rev Pathol. 2014;9:47–71. doi: 10.1146/annurev-pathol-012513-104720. [DOI] [PubMed] [Google Scholar]
  • 110.Hickey MM, Richardson T, Wang T, et al. The von Hippel-Lindau Chuvash mutation promotes pulmonary hypertension and fibrosis in mice. J Clin Invest. 2010;120:827–839. doi: 10.1172/JCI36362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Kapitsinou PP, Rajendran G, Astleford L, et al. The endothelial prolyl-4-hydroxylase domain 2/hypoxia-inducible factor 2 axis regulates pulmonary artery pressure in mice. Mol Cell Biol. 2016;36:1584–1594. doi: 10.1128/MCB.01055-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Cowburn AS, Crosby A, Macias D, et al. HIF2alpha-arginase axis is essential for the development of pulmonary hypertension. Proc Natl Acad Sci USA. 2016;113:8801–8806. doi: 10.1073/pnas.1602978113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Bryant AJ, Carrick RP, McConaha ME, et al. Endothelial HIF signaling regulates pulmonary fibrosis-associated pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2016;310:L249–L262. doi: 10.1152/ajplung.00258.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Dai Z, Li M, Wharton J, Zhu MM, Zhao YY. Prolyl-4 hydroxylase 2 (PHD2) deficiency in endothelial cells and hematopoietic cells induces obliterative vascular remodeling and severe pulmonary arterial hypertension in mice and humans through hypoxia-inducible factor-2alpha. Circulation. 2016;133:2447–2458. doi: 10.1161/CIRCULATIONAHA.116.021494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Bolignano D, Rastelli S, Agarwal R, et al. Pulmonary hypertension in CKD. Am J Kidney Dis. 2013;61:612–622. doi: 10.1053/j.ajkd.2012.07.029. [DOI] [PubMed] [Google Scholar]
  • 116.Brigandi RA, Russ SF, Al-Banna M, Zhang J, Erickson-Miller CL, Peng B. Prolylhydroxylase inhibitor modulation of erythropoietin in a randomized placebo controlled trial. J Am Soc Nephrol. 2010;21:390A. [Google Scholar]
  • 117.Haase VH. Renal cancer: oxygen meets metabolism. Exp Cell Res. 2012;318:1057–1067. doi: 10.1016/j.yexcr.2012.02.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Girgis CM, Cheng K, Scott CH, Gunton JE. Novel links between HIFs, type 2 diabetes, and metabolic syndrome. Trends Endocrinol Metab. 2012;23:372–380. doi: 10.1016/j.tem.2012.05.003. [DOI] [PubMed] [Google Scholar]
  • 119.Gunton JE, Kulkarni RN, Yim S, et al. Loss of ARNT/HIF1beta mediates altered gene expression and pancreatic-islet dysfunction in human type 2 diabetes. Cell. 2005;122:337–349. doi: 10.1016/j.cell.2005.05.027. [DOI] [PubMed] [Google Scholar]
  • 120.Keith B, Johnson RS, Simon MC. HIF1alpha and HIF2alpha: Sibling rivalry in hypoxic tumour growth and progression. Nat Rev Cancer. 2012;12:9–22. doi: 10.1038/nrc3183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Haase VH. Mechanisms of hypoxia responses in renal tissue. J Am Soc Nephrol. 2013;24:537–541. doi: 10.1681/ASN.2012080855. [DOI] [PubMed] [Google Scholar]
  • 122.Nangaku M, Rosenberger C, Heyman SN, Eckardt KU. Regulation of hypoxia-inducible factor in kidney disease. Clin Exp Pharmacol Physiol. 2013;40:148–157. doi: 10.1111/1440-1681.12005. [DOI] [PubMed] [Google Scholar]
  • 123.Mokas S, Lariviere R, Lamalice L, et al. Hypoxia-inducible factor-1 plays a role in phosphate-induced vascular smooth muscle cell calcification. Kidney Int. 2016;90:598–609. doi: 10.1016/j.kint.2016.05.020. [DOI] [PubMed] [Google Scholar]
  • 124.Francis C, David V. Inflammation regulates fibroblast growth factor 23 production. Curr Opin Nephrol Hypertens. 2016;25:325–332. doi: 10.1097/MNH.0000000000000232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Zhang Q, Doucet M, Tomlinson RE, et al. The hypoxia-inducible factor-1alpha activates ectopic production of fibroblast growth factor 23 in tumor-induced osteomalacia. Bone Res. 2016;4:16011. doi: 10.1038/boneres.2016.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Mahon PC, Hirota K, Semenza GL. FIH-1: A novel protein that interacts with HIF-1alpha and VHL to mediate repression of HIF-1 transcriptional activity. Genes Dev. 2001;15:2675–2686. doi: 10.1101/gad.924501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Lando D, Peet DJ, Gorman JJ, Whelan DA, Whitelaw ML, Bruick RK. FIH-1 is an asparaginyl hydroxylase enzyme that regulates the transcriptional activity of hypoxia-inducible factor. Genes Dev. 2002;16:1466–1471. doi: 10.1101/gad.991402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.McNeill LA, Hewitson KS, Claridge TD, Seibel JF, Horsfall LE, Schofield CJ. Hypoxia-inducible factor asparaginyl hydroxylase (FIH-1) catalyses hydroxylation at the beta-carbon of asparagine-803. Biochem J. 2002;367:571–575. doi: 10.1042/BJ20021162. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES