Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
FEBS J ; 288(18): 5446-5458, 2021 09.
Article in English | MEDLINE | ID: mdl-33713542

ABSTRACT

Cellular injury caused by stimuli plays an important role in the progression of various diseases including acute and chronic kidney diseases. The dynamic transcriptional regulation responding to stimuli underlies the important mechanism of injury. In this study, we investigated the regulatory elements and their dynamic activities in kidney tubular epithelial cells. We captured the chromatin accessibility and gene expression with ATAC-seq and RNA sequencing under a variety of extracellular stimuli including H2 O2 , TGF-ß1, and FG4592 which is an agonist of hypoxia-inducible factor. Our results revealed both condition-specific and condition-shared transcription regulation. Interestingly, the shared regulation program revealed that the key transcription factor HNF1B-mediated cellular reprogramming leads to a common change among the stimuli. We found the HNF1B regulatory network was significantly disrupted in various kidney diseases.


Subject(s)
Acute Kidney Injury/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Kidney/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Renal Insufficiency, Chronic/genetics , Acute Kidney Injury/pathology , Chromatin/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation/genetics , Humans , Hydrogen Peroxide/pharmacology , Hypoxia-Inducible Factor 1/agonists , Hypoxia-Inducible Factor 1/genetics , Kidney/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Renal Insufficiency, Chronic/pathology , Transcription Factors/genetics , Transforming Growth Factor beta1/genetics
2.
Drug Test Anal ; 12(11-12): 1614-1619, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32809277

ABSTRACT

Early in 2020, racehorse doping cases revolved around the hypoxia-inducible factor (HIF) activator IOX-2. While the composition of IOX-2 has also been known and monitored in human doping controls for several years, the testing capability of routine sports drug testing methods was revisited for this newly surfaced doping agent. IOX-2 and the analytically well-established HIF activator roxadustat (FG-4592) share identical precursor/product ion pairs, enabling their co-detection in existing initial testing procedures in routine doping controls for the intact unconjugated analytes. In addition, hydroxylated IOX-2 and the corresponding glucuronic acid conjugates were identified as major metabolites in a microdose elimination study, contributing to enhanced initial testing and confirmation procedures.


Subject(s)
Doping in Sports/prevention & control , Hypoxia-Inducible Factor 1/agonists , Hypoxia-Inducible Factor 1/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/urine , Isoquinolines/urine , Substance Abuse Detection/standards , Doping in Sports/methods , Glycine/administration & dosage , Glycine/analogs & derivatives , Glycine/urine , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/administration & dosage , Isoquinolines/administration & dosage , Male , Middle Aged , Pilot Projects , Retrospective Studies , Substance Abuse Detection/methods
3.
Artif Organs ; 42(3): 280-289, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29266279

ABSTRACT

The protective role of hypoxia-inducible factor-1 (HIF-1) against liver ischemia-reperfusion injury has been well proved. However its role in liver donation and preservation from donation after cardiac death (DCD) is still unknown. The objective of this study was to test the hypothesis that pharmaceutical stabilization of HIF-1 in DCD donors would result in a better graft liver condition. Male SD rats (6 animals per group) were randomly given the synthetic prolyl hydroxylase domain inhibitor FG-4592 (Selleck, 6 mg/kg of body weight) or its vehicle (dimethylsulfoxide). Six hours later, cardiac arrest was induced by bilateral pneumothorax. Rat livers were retrieved 30 min after cardiac arrest, and subsequently cold stored in University of Wisconsin solution for 24 h. They were reperfused for 60 min with Krebs-Henseleit bicarbonate buffer in an isolated perfused liver model, after which the perfusate and liver tissues were investigated. Pretreatment with FG-4592 in DCD donors significantly improved graft function with increased bile production and synthesis of adenosine triphosphate, decreased perfusate liver enzyme release, histology injury scores and oxidative stress-induced cell injury and apoptosis after reperfusion with the isolated perfused liver model. The beneficial effects of FG-4592 is attributed in part to the accumulation of HIF-1 and ultimately increased PDK1 production. Pretreatment with FG-4592 in DCD donors resulted in activation of the HIF-1 pathway and subsequently protected liver grafts from warm ischemia and cold-stored injury. These data suggest that the pharmacological HIF-1 induction may provide a clinically applicable therapeutic intervention to prevent injury to DCD allografts.


Subject(s)
Hypoxia-Inducible Factor 1/agonists , Liver Transplantation/methods , Liver/drug effects , Liver/physiology , Organ Preservation/methods , Perfusion/methods , Adenosine/metabolism , Allopurinol/metabolism , Animals , Death , Glutathione/metabolism , Graft Survival/drug effects , Hypoxia-Inducible Factor 1/metabolism , Insulin/metabolism , Male , Organ Preservation Solutions/metabolism , Raffinose/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar
4.
Mol Med Rep ; 15(4): 1847-1852, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28259984

ABSTRACT

Mimosine, which is a natural plant amino acid present in the Leucaena genus, is able to induce hypoxia­inducible factors (HIFs). Previous evidence has indicated that HIF regulates angiogenesis­osteogenesis coupling in bone metabolism, and it has previously been reported that mimosine inhibits prostaglandin (PG)F2α­induced osteoprotegerin (OPG) synthesis without affecting interleukin­6 (IL­6) production in osteoblast­like MC3T3­E1 cells. In addition, PGE1 has been demonstrated to induce OPG synthesis via activation of p38 mitogen­activated protein (MAP) kinase and stress­activated protein kinase/c­Jun N­terminal kinase (SAPK/JNK) in these cells, and PGE1 stimulates IL­6 production via the activation of protein kinase A. In the present study, the effects of mimosine on the PGE1­stimulated synthesis of OPG and IL­6 were investigated in osteoblast­like MC3T3­E1 cells. The concentrations of OPG and IL­6 were measured using relevant ELISA kits. OPG mRNA was measured by semi­quantitative reverse transcription polymerase chain reaction. The phosphorylation of p38 MAP kinase and SAPK/JNK was analyzed by western blotting. Mimosine significantly reduced PGE1­induced release of OPG and OPG mRNA expression levels without affecting the release of IL­6. In addition, deferoxamine, which is also a normoxic HIF inducer, significantly inhibited PGE1­induced OPG release and OPG mRNA expression levels; however, it had little effect on IL­6 release. Furthermore, mimosine and deferoxamine failed to affect PGE1­stimulated phosphorylation of p38 MAP kinase or SAPK/JNK. These results strongly suggest that normoxic HIF inducers attenuate PGE1­stimulated OPG synthesis without affecting IL­6 production in osteoblasts.


Subject(s)
Alprostadil/metabolism , Deferoxamine/pharmacology , Hypoxia-Inducible Factor 1/agonists , Mimosine/pharmacology , Osteoblasts/drug effects , Osteoprotegerin/metabolism , Animals , Cell Line , Fabaceae/chemistry , Hypoxia-Inducible Factor 1/metabolism , Interleukin-6/metabolism , MAP Kinase Kinase 4/metabolism , Mice , Mimosine/chemistry , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoprotegerin/genetics , Phosphorylation/drug effects , Protein Biosynthesis , RNA, Messenger/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
5.
J Antibiot (Tokyo) ; 69(10): 747-753, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26956789

ABSTRACT

In the course of our screening for activators of hypoxia-inducible factor (HIF), A-503451 A and virantmycin were isolated from the cultured broth of an actinomycete strain, Streptomyces sp. SANK 60101. From the same culture, the non-active homologs A-503451 B and D were also isolated. A-503451 A and virantmycin activated HIF-dependent reporter gene expression with EC50 values of 8 and 17 ng ml-1, respectively. They are highly potent activators of HIF and thus may be therapeutically useful for erythropoiesis and neural cell protection.


Subject(s)
Fermentation , Hypoxia-Inducible Factor 1/agonists , Quinolines/chemistry , Streptomyces/metabolism , Genes, Reporter , Hep G2 Cells , Humans , Molecular Structure , Streptomyces/genetics
6.
Inflamm Bowel Dis ; 21(2): 267-75, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25545377

ABSTRACT

BACKGROUND: Pharmacological induction of hypoxia-inducible factor (HIF), a global transcriptional regulator of the hypoxic response, by prolyl hydroxylase inhibitors (PHDi) is protective in murine models of colitis, and epithelial cells are critical for the observed therapeutic efficacy. Because systemic HIF activation may lead to potentially negative off-target effects, we hypothesized that targeting epithelial HIF through oral delivery of PHDi would be sufficient to protect against colitis in a mouse model. METHODS: Using a chemically induced trinitrobenzene sulfonic acid murine model of colitis, we compared the efficacy of oral and intraperitoneal (i.p.) delivery of the PHDi; AKB-4924 in preventing colitis, as measured by endoscopy, histology, barrier integrity, and immune profiling. Furthermore, we measured potential off-target effects, examining HIF and HIF target genes in the heart and kidney, as well as erythropoietin and hematocrit levels. RESULTS: Oral administration of AKB-4924 exhibited mucosal protection comparable i.p. dosing. Oral delivery of PHDi led to reduced colonic epithelial HIF stabilization compared with i.p. delivery, but this was still sufficient to induce transcription of downstream HIF targets. Furthermore, oral delivery of PHDi led to reduced stabilization of HIF and activation of HIF targets in extraintestinal organs. CONCLUSIONS: Oral delivery of PHDi therapies to this intestinal mucosa protects against colitis in animal models and represents a potential therapeutic strategy for inflammatory bowel disease, which also precludes unwanted extraintestinal effects.


Subject(s)
Colitis/drug therapy , Disease Models, Animal , Mucous Membrane/drug effects , Piperazines/administration & dosage , Prolyl-Hydroxylase Inhibitors/administration & dosage , Pyridones/administration & dosage , Wound Healing/drug effects , Administration, Oral , Animals , Colitis/chemically induced , Colitis/pathology , Female , Hypoxia-Inducible Factor 1/agonists , Hypoxia-Inducible Factor 1/metabolism , Immunoenzyme Techniques , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Mice , Mice, Inbred BALB C , Mucous Membrane/metabolism , Piperazines/pharmacology , Prolyl-Hydroxylase Inhibitors/pharmacology , Pyridones/pharmacology , Trinitrobenzenesulfonic Acid/toxicity
7.
Mucosal Immunol ; 7(1): 114-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23695513

ABSTRACT

Pharmacological stabilization of hypoxia-inducible factor (HIF) through prolyl hydroxylase (PHD) inhibition limits mucosal damage associated with models of murine colitis. However, little is known about how PHD inhibitors (PHDi) influence systemic immune function during mucosal inflammation or the relative importance of immunological changes to mucosal protection. We hypothesized that PHDi enhances systemic innate immune responses to colitis-associated bacteremia. Mice with colitis induced by trinitrobenzene sulfonic acid were treated with AKB-4924, a new HIF-1 isoform-predominant PHDi, and clinical, immunological, and biochemical endpoints were assessed. Administration of AKB-4924 led to significantly reduced weight loss and disease activity compared with vehicle controls. Treated groups were pyrexic but did not become subsequently hypothermic. PHDi treatment augmented epithelial barrier function and led to an approximately 50-fold reduction in serum endotoxin during colitis. AKB-4924 also decreased cytokines involved in pyrogenesis and hypothermia, significantly reducing serum levels of interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α while increasing IL-10. Treatment offered no protection against colitis in epithelial-specific HIF-1α-deficient mice, strongly implicating epithelial HIF-1α as the tissue target for AKB-4924-mediated protection. Taken together, these results indicate that inhibition of prolyl hydroxylase with AKB-4924 enhances innate immunity and identifies that the epithelium is a central site of inflammatory protection afforded by PHDi in murine colitis.


Subject(s)
Colitis/immunology , Colitis/metabolism , Immunity, Innate/drug effects , Immunity, Mucosal/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Prolyl Hydroxylases/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Animals , Colitis/chemically induced , Colitis/drug therapy , Disease Models, Animal , Endotoxemia/drug therapy , Female , Hypoxia-Inducible Factor 1/agonists , Hypoxia-Inducible Factor 1, alpha Subunit/agonists , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Permeability/drug effects , Piperazines/administration & dosage , Piperazines/pharmacology , Pyridones/administration & dosage , Pyridones/pharmacology , Trinitrobenzenesulfonic Acid/adverse effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
J Mol Med (Berl) ; 90(9): 1079-89, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22371073

ABSTRACT

Hypoxia inducible factor-1 (HIF-1) is a transcription factor that is a major regulator of energy homeostasis and cellular adaptation to low oxygen stress. HIF-1 is also activated in response to bacterial pathogens and supports the innate immune response of both phagocytes and keratinocytes. In this work, we show that a new pharmacological compound AKB-4924 increases HIF-1 levels and enhances the antibacterial activity of phagocytes and keratinocytes against both methicillin-sensitive and methicillin-resistant strains of Staphylococcus aureus in vitro. AKB-4924 is also effective in stimulating the killing capacity of keratinocytes against the important opportunistic skin pathogens Pseudomonas aeruginosa and Acinetobacter baumanii. The effect of AKB-4924 is mediated through the activity of host cells, as the compound exerts no direct antimicrobial activity. Administered locally as a single agent, AKB-4924 limits S. aureus proliferation and lesion formation in a mouse skin abscess model. This approach to pharmacologically boost the innate immune response via HIF-1 stabilization may serve as a useful adjunctive treatment for antibiotic-resistant bacterial infections.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Hypoxia-Inducible Factor 1/immunology , Immunity, Innate/drug effects , Piperazines/therapeutic use , Pyridones/therapeutic use , Skin Diseases, Bacterial/prevention & control , Skin/microbiology , Animals , Cell Line , Female , Humans , Hypoxia-Inducible Factor 1/agonists , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/microbiology , Mice , Phagocytes/drug effects , Phagocytes/immunology , Phagocytes/microbiology , Piperazines/pharmacology , Pyridones/pharmacology , Skin/drug effects , Skin/immunology , Skin Diseases, Bacterial/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/drug effects , Staphylococcus aureus/immunology
10.
Antioxid Redox Signal ; 12(4): 459-80, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19737089

ABSTRACT

Abstract Ischemic stroke is a major cause of death worldwide, and current therapeutic options are very limited. Preconditioning with an ischemic or hypoxic insult is beneficial in experimental models of ischemic stroke. Ischemia/hypoxia results in activation of numerous transcription factors, including hypoxia inducible factor (HIF), which is a master regulator of oxygen homeostasis. HIF activation induces a diverse range of target genes, encompassing a wide variety of cellular processes; including angiogenesis, energy metabolism, cell survival, radical production/scavenging, iron metabolism, stem cell homing, and differentiation. Inhibition of HIF prolyl hydroxylase domain (PHD) enzymes results in activation of HIF and is likely to mimic, at least in part, the effects of hypoxia preconditioning. A caveat is that not all consequences of HIF activation will be beneficial and some could even be deleterious. Nevertheless, PHD inhibitors may be therapeutically useful in the treatment of stroke. Prototype PHD inhibitors have shown promising results in preclinical models.


Subject(s)
Brain Ischemia/drug therapy , Cytoprotection , Enzyme Inhibitors/pharmacology , Hypoxia-Inducible Factor 1/agonists , Neuroprotective Agents/pharmacology , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Stroke/drug therapy , Animals , Blood-Brain Barrier/enzymology , Brain/blood supply , Brain Ischemia/physiopathology , Calcium Channel Blockers/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Free Radical Scavengers/pharmacology , Humans , Ischemic Preconditioning , Mice , Oxidative Stress/drug effects , Rats , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Stroke/enzymology , Stroke/pathology
11.
J Biol Chem ; 284(44): 30350-9, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19726677

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1) is a key transcription factor regulating cellular responses to hypoxia and is composed of alpha and beta subunits. During normoxia, factor inhibiting HIF-1 (FIH-1) inhibits the activity of HIF-1 by preventing HIF-1alpha binding to p300/CBP via modification of the Asn(803) residue. However, it is not known whether FIH-1 activity can be regulated in an oxygen-independent manner. In this study, we survey possible binding proteins to FIH-1 and identify Mint3/APBA3, which has been reported to bind Alzheimer beta-amyloid precursor protein. Purified Mint3 binds FIH-1 and inhibits the ability of FIH-1 to modify HIF-1alpha in vitro. In a reporter assay, the activity of HIF-1alpha is suppressed because of endogenous FIH-1 in HEK293 cells, and expression of Mint3 antagonizes this suppression. Macrophages are known to depend on glycolysis for ATP production because of elevated HIF-1 activity. FIH-1 activity is suppressed in macrophages by Mint3 so as to maintain HIF-1 activity. FIH-1 forms a complex with Mint3, and these two factors co-localize within the perinuclear region. Knockdown of Mint3 expression in macrophages leads to redistribution of FIH-1 to the cytoplasm and decreases glycolysis and ATP production. Thus, Mint3 regulates the FIH-1-HIF-1 pathway, which controls ATP production in macrophages and therefore represents a potential new therapeutic target to regulate macrophage-mediated inflammation.


Subject(s)
Carrier Proteins/physiology , Macrophages/metabolism , Repressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Adenosine Triphosphate/biosynthesis , Glycolysis , Humans , Hypoxia-Inducible Factor 1/agonists , Hypoxia-Inducible Factor 1/metabolism , Mixed Function Oxygenases , Protein Binding , Protein Transport , Repressor Proteins/antagonists & inhibitors
13.
Biochem Biophys Res Commun ; 338(1): 627-38, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16153592

ABSTRACT

The heterodimeric transcription factor HIF (hypoxia-inducible factor), consisting of a labile alpha-subunit and a stable beta-subunit, is a master regulator of genes involved in acute or chronic adaptation to low oxygen. Studies performed over the past 5 years revealed that HIFalpha-subunits are enzymatically hydroxylated in an oxygen-dependent manner. Hydroxylation of either of two conserved prolyl residues targets HIFalpha for destruction by a ubiquitin ligase containing the von Hippel-Lindau tumor suppressor protein whereas hydroxylation on a C-terminal asparagine affects HIF transactivation function. Pharmacological manipulation of HIF activity might be beneficial in diseases characterized by abnormal tissue oxygenation including myocardial infarction, cerebrovascular disease, and cancer.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Oxygen/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/physiology , Animals , Humans , Hydroxylation , Hypoxia-Inducible Factor 1/agonists , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Oxygen/chemistry , Von Hippel-Lindau Tumor Suppressor Protein/chemistry , Von Hippel-Lindau Tumor Suppressor Protein/genetics , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL