Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
Diabetes ; 71(8): 1746-1762, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35167651

ABSTRACT

Dysregulation of extracellular matrix proteins in obese adipose tissue (AT) induces systemic insulin resistance. The metabolic roles of type VI collagen and its cleavage peptide endotrophin in obese AT are well established. However, the mechanisms regulating endotrophin generation remain elusive. Herein, we identified that several endotrophin-containing peptides (pre-endotrophins) were generated from the COL6A3 chain in a stepwise manner for the efficient production of mature endotrophin, partly through the action of hypoxia-induced matrix metalloproteinases (MMPs), including MMP2, MMP9, and MMP16. Hypoxia is an upstream regulator of COL6A3 expression and the proteolytic processing that regulates endotrophin generation. Hypoxia-inducible factor 1α (HIF1α) and the hypoxia-associated suppression of microRNA-29 (miR-29) cooperatively control the levels of COL6A3 and MMPs, which are responsible for endotrophin generation in hypoxic ATs. Adipocyte-specific Hif1α knock-out (APN-HIF1αKO) mice fed a chronic high-fat diet exhibited the significant amelioration of both local fibro-inflammation in AT and systemic insulin resistance compared with their control littermates, partly through the inhibition of endotrophin generation. Strikingly, adenovirus-mediated miR-29 overexpression in the ATs of APN-HIF1αKO mice in obesity significantly decreased endotrophin levels, suggesting that miR-29, combined with HIF1α inhibition in AT, could be a promising therapeutic strategy for treating obesity and related metabolic diseases.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Insulin Resistance , MicroRNAs , Adipose Tissue/metabolism , Animals , Collagen Type VI/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Inflammation/genetics , Inflammation/metabolism , Insulin Resistance/genetics , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Obesity/genetics , Obesity/metabolism
2.
FASEB J ; 35(10): e21915, 2021 10.
Article in English | MEDLINE | ID: mdl-34496088

ABSTRACT

During development, erythroid cells are generated by two waves of hematopoiesis. In zebrafish, primitive erythropoiesis takes place in the intermediate cell mass region, and definitive erythropoiesis arises from the aorta-gonad mesonephros. TALE-homeoproteins Meis1 and Pbx1 function upstream of GATA1 to specify the erythroid lineage. Embryos lacking Meis1 or Pbx1 have weak gata1 expression and fail to produce primitive erythrocytes. Nevertheless, the underlying mechanism of how Meis1 and Pbx1 mediate gata1 transcription in erythrocytes remains unclear. Here we show that Hif1α acts downstream of Meis1 to mediate gata1 expression in zebrafish embryos. Inhibition of Meis1 expression resulted in suppression of hif1a expression and abrogated primitive erythropoiesis, while injection with in vitro-synthesized hif1α mRNA rescued gata1 transcription in Meis1 morphants and recovered their erythropoiesis. Ablation of Hif1α expression either by morpholino knockdown or Crispr-Cas9 knockout suppressed gata1 transcription and abrogated primitive erythropoiesis. Results of chromatin immunoprecipitation assays showed that Hif1α associates with hypoxia-response elements located in the 3'-flanking region of gata1 during development, suggesting that Hif1α regulates gata1 expression in vivo. Together, our results indicate that Meis1, Hif1α, and GATA1 indeed comprise a hierarchical regulatory network in which Hif1α acts downstream of Meis1 to activate gata1 transcription through direct interactions with its cis-acting elements in primitive erythrocytes.


Subject(s)
Erythroid Cells/metabolism , Erythropoiesis , GATA1 Transcription Factor/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Chromatin Immunoprecipitation , Erythrocytes/cytology , Erythrocytes/metabolism , Erythroid Cells/cytology , Erythropoiesis/genetics , GATA1 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Myeloid Ecotropic Viral Integration Site 1 Protein/deficiency , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Pre-B-Cell Leukemia Transcription Factor 1/deficiency , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Transcription, Genetic , Zebrafish/blood , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
3.
Am J Respir Cell Mol Biol ; 65(4): 390-402, 2021 10.
Article in English | MEDLINE | ID: mdl-34003729

ABSTRACT

Obstructive sleep apnea is associated with insulin resistance, lipid dysregulation, and hepatic steatosis and fibrosis in nonalcoholic fatty liver disease (NAFLD). We have previously shown that hepatocyte HIF-1 (hypoxia-inducible factor-1) mediates the development of liver fibrosis in a mouse model of NAFLD. We hypothesized that intermittent hypoxia (IH) modeling obstructive sleep apnea would worsen hepatic steatosis and fibrosis in murine NAFLD, via HIF-1. Mice with hepatocyte-specific deletion of Hif1a (Hif1a-/-hep) and wild-type (Hif1aF/F) controls were fed a high trans-fat diet to induce NAFLD with steatohepatitis. Half from each group were exposed to IH, and the other half were exposed to intermittent air. A glucose tolerance test was performed just prior to the end of the experiment. Mitochondrial efficiency was assessed in fresh liver tissue at the time of death. The hepatic malondialdehyde concentration and proinflammatory cytokine levels were assessed, and genes of collagen and fatty acid metabolism were examined. Hif1a-/-hep mice gained less weight than wild-type Hif1a mice (-2.3 g, P = 0.029). There was also a genotype-independent effect of IH on body weight, with less weight gain in mice exposed to IH (P = 0.003). Fasting glucose, homeostatic model assessment for insulin resistance, and glucose tolerance test results were all improved in Hif1a-/-hep mice. Liver collagen was increased in mice exposed to IH (P = 0.033) and was reduced in Hif1a-/-hep mice (P < 0.001), without any significant exposure/genotype interaction being demonstrated. Liver TNF-α and IL-1ß were significantly increased in mice exposed to IH and were decreased in Hif1a-/-hep mice. We conclude that HIF-1 signaling worsens the metabolic profile and hastens NAFLD progression and that IH may worsen liver fibrosis. These effects are plausibly mediated by hepatic inflammatory stress.


Subject(s)
Hepatocytes/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia/complications , Liver Cirrhosis/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Hepatocytes/pathology , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin Resistance/physiology , Lipid Metabolism/immunology , Liver/metabolism , Liver Cirrhosis/complications , Liver Cirrhosis/pathology , Mice
4.
Clin Sci (Lond) ; 135(10): 1273-1288, 2021 05 28.
Article in English | MEDLINE | ID: mdl-33997886

ABSTRACT

Cisplatin (Cis) can cause chronic kidney disease (CKD) and promote renal fibrosis, but the underlying mechanism is not fully understood. Hypoxia inducible factor-1α (HIF-1α) can promote renal fibrosis in some kidney diseases, but its role in Cis-induced CKD is still unknown. Notch-1 is a recognized molecule that promotes renal fibrosis under pathological circumstances, and evidence shows that HIF-1α and Notch-1 are closely related to each other. In the present study, mice with HIF-1α gene knockout in proximal tubular cells (PTCs) (PT-HIF-1α-KO) were generated and treated with Cis to induce CKD. A human proximal tubular cell line (HK-2) and primary mouse PTCs were used for in vitro studies. The results showed that HIF-1α was increased in the kidneys of Cis-treated wild-type mice, accompanied by elevated Notch-1, Notch-1 intracellular domain (N1ICD), Hes-1 and renal fibrosis. However, these alterations were partially reversed in PT-HIF-1α-KO mice. Similar results were observed in HK-2 cells and primary mouse PTCs. In addition, treating the cells with Cis induced a marked interaction of HIF-1α and N1ICD. Further inhibiting Notch-1 significantly reduced cellular fibrogenesis but did not affect HIF-1α expression. The data suggested that HIF-1α could promote renal fibrosis in Cis-induced CKD by activating Notch-1 both transcriptionally and post-transcriptionally and that HIF-1α may serve as a potential therapeutic target for Cis-induced CKD.


Subject(s)
Fibrosis/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Animals , Epithelial Cells/metabolism , Fibrosis/etiology , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Kidney/metabolism , Kidney Tubules, Proximal/pathology
5.
Circulation ; 143(23): 2254-2272, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33663226

ABSTRACT

BACKGROUND: Cyanotic congenital heart disease (CCHD) is a complex pathophysiological condition involving systemic chronic hypoxia (CH). Some patients with CCHD are unoperated for various reasons and remain chronically hypoxic throughout their lives, which heightens the risk of heart failure as they age. Hypoxia activates cellular metabolic adaptation to balance energy demands by accumulating hypoxia-inducible factor 1-α (HIF-1α). This study aims to determine the effect of CH on cardiac metabolism and function in patients with CCHD and its association with age. The role of HIF-1α in this process was investigated, and potential therapeutic targets were explored. METHODS: Patients with CCHD (n=25) were evaluated for cardiac metabolism and function with positron emission tomography/computed tomography and magnetic resonance imaging. Heart tissue samples were subjected to metabolomic and protein analyses. CH rodent models were generated to enable continuous observation of changes in cardiac metabolism and function. The role of HIF-1α in cardiac metabolic adaptation to CH was investigated with genetically modified animals and isotope-labeled metabolomic pathway tracing studies. RESULTS: Prepubertal patients with CCHD had glucose-dominant cardiac metabolism and normal cardiac function. In comparison, among patients who had entered puberty, the levels of myocardial glucose uptake and glycolytic intermediates were significantly decreased, but fatty acids were significantly increased, along with decreased left ventricular ejection fraction. These clinical phenotypes were replicated in CH rodent models. In patients with CCHD and animals exposed to CH, myocardial HIF-1α was upregulated before puberty but was significantly downregulated during puberty. In cardiomyocyte-specific Hif-1α-knockout mice, CH failed to initiate the switch of myocardial substrates from fatty acids to glucose, thereby inhibiting ATP production and impairing cardiac function. Increased insulin resistance during puberty suppressed myocardial HIF-1α and was responsible for cardiac metabolic maladaptation in animals exposed to CH. Pioglitazone significantly reduced myocardial insulin resistance, restored glucose metabolism, and improved cardiac function in pubertal CH animals. CONCLUSIONS: In patients with CCHD, maladaptation of cardiac metabolism occurred during puberty, along with impaired cardiac function. HIF-1α was identified as the key regulator of cardiac metabolic adaptation in animals exposed to CH, and pubertal insulin resistance could suppress its expression. Pioglitazone administration during puberty might help improve cardiac function in patients with CCHD.


Subject(s)
Heart Defects, Congenital/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myocardium/metabolism , Animals , Disease Models, Animal , Fatty Acids/metabolism , Glucose/metabolism , Glycolysis/drug effects , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Insulin Resistance , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Pioglitazone/pharmacology , Positron Emission Tomography Computed Tomography , Puberty , Up-Regulation , Ventricular Function, Left/drug effects
6.
Genes Dev ; 35(3-4): 250-260, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33446567

ABSTRACT

Reactive oxygen species (ROS) produced by NADPH1 oxidase 1 (NOX1) are thought to drive spermatogonial stem cell (SSC) self-renewal through feed-forward production of ROS by the ROS-BCL6B-NOX1 pathway. Here we report the critical role of oxygen on ROS-induced self-renewal. Cultured SSCs proliferated poorly and lacked BCL6B expression under hypoxia despite increase in mitochondria-derived ROS. Due to lack of ROS amplification under hypoxia, NOX1-derived ROS were significantly reduced, and Nox1-deficient SSCs proliferated poorly under hypoxia but normally under normoxia. NOX1-derived ROS also influenced hypoxic response in vivo because Nox1-deficient undifferentiated spermatogonia showed significantly reduced expression of HIF1A, a master transcription factor for hypoxic response. Hypoxia-induced poor proliferation occurred despite activation of MYC and suppression of CDKN1A by HIF1A, whose deficiency exacerbated self-renewal efficiency. Impaired proliferation of Nox1- or Hif1a-deficient SSCs under hypoxia was rescued by Cdkn1a depletion. Consistent with these observations, Cdkn1a-deficient SSCs proliferated actively only under hypoxia but not under normoxia. On the other hand, chemical suppression of mitochondria-derived ROS or Top1mt mitochondria-specific topoisomerase deficiency did not influence SSC fate, suggesting that NOX1-derived ROS play a more important role in SSCs than mitochondria-derived ROS. These results underscore the importance of ROS origin and oxygen tension on SSC self-renewal.


Subject(s)
Adult Germline Stem Cells/cytology , Cell Hypoxia/physiology , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Division/genetics , Cell Proliferation/genetics , Cells, Cultured , DNA Topoisomerases, Type I/genetics , Gene Expression Regulation, Developmental , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Mice , Mice, Knockout , Mitochondria/physiology , NADPH Oxidase 1/metabolism
7.
Cell Mol Life Sci ; 78(1): 195-206, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32088728

ABSTRACT

Hypoxia-inducible factors (HIFs) mediate metabolic reprogramming in response to hypoxia. However, the role of HIFs in branched-chain amino acid (BCAA) metabolism remains unknown. Here we show that hypoxia upregulates mRNA and protein levels of the BCAA transporter LAT1 and the BCAA metabolic enzyme BCAT1, but not their paralogs LAT2-4 and BCAT2, in human glioblastoma (GBM) cell lines as well as primary GBM cells. Hypoxia-induced LAT1 protein upregulation is mediated by both HIF-1 and HIF-2 in GBM cells. Although both HIF-1α and HIF-2α directly bind to the hypoxia response element at the first intron of the human BCAT1 gene, HIF-1α is exclusively responsible for hypoxia-induced BCAT1 expression in GBM cells. Knockout of HIF-1α and HIF-2α significantly reduces glutamate labeling from BCAAs in GBM cells under hypoxia, which provides functional evidence for HIF-mediated reprogramming of BCAA metabolism. Genetic or pharmacological inhibition of BCAT1 inhibits GBM cell growth under hypoxia. Together, these findings uncover a previously unrecognized HIF-dependent metabolic pathway that increases GBM cell growth under conditions of hypoxic stress.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , CRISPR-Cas Systems/genetics , Cell Hypoxia , Cell Proliferation , Cells, Cultured , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Glioblastoma/metabolism , Glioblastoma/pathology , Glutamic Acid/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Protein Binding , Transaminases/antagonists & inhibitors , Transaminases/genetics , Transaminases/metabolism
8.
Gastroenterology ; 159(5): 1882-1897.e5, 2020 11.
Article in English | MEDLINE | ID: mdl-32768595

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinomas (PDACs) are hypovascular, resulting in the up-regulation of hypoxia inducible factor 1 alpha (HIF1A), which promotes the survival of cells under low-oxygen conditions. We studied the roles of HIF1A in the development of pancreatic tumors in mice. METHODS: We performed studies with KrasLSL-G12D/+;Trp53LSL-R172H/+;Pdx1-Cre (KPC) mice, KPC mice with labeled pancreatic epithelial cells (EKPC), and EKPC mice with pancreas-specific depletion of HIF1A. Pancreatic and other tissues were collected and analyzed by histology and immunohistochemistry. Cancer cells were cultured from PDACs from mice and analyzed in cell migration and invasion assays and by immunoblots, real-time polymerase chain reaction, and liquid chromatography-mass spectrometry. We performed studies with the human pancreatic cancer cell lines PATU-8988T, BxPC-3, PANC-1, and MiaPACA-2, which have no or low metastatic activity, and PATU-8988S, AsPC-1, SUIT-2 and Capan-1, which have high metastatic activity. Expression of genes was knocked down in primary cancer cells and pancreatic cancer cell lines by using small hairpin RNAs; cells were injected intravenously into immune-competent and NOD/SCID mice, and lung metastases were quantified. We compared levels of messenger RNAs in pancreatic tumors and normal pancreas in The Cancer Genome Atlas. RESULTS: EKPC mice with pancreas-specific deletion of HIF1A developed more advanced pancreatic neoplasias and PDACs with more invasion and metastasis, and had significantly shorter survival times, than EKPC mice. Pancreatic cancer cells from these tumors had higher invasive and metastatic activity in culture than cells from tumors of EKPC mice. HIF1A-knockout pancreatic cancer cells had increased expression of protein phosphatase 1 regulatory inhibitor subunit 1B (PPP1R1B). There was an inverse correlation between levels of HIF1A and PPP1R1B in human PDAC tumors; higher expression of PPP1R1B correlated with shorter survival times of patients. Metastatic human pancreatic cancer cell lines had increased levels of PPP1R1B and lower levels of HIF1A compared with nonmetastatic cancer cell lines; knockdown of PPP1R1B significantly reduced the ability of pancreatic cancer cells to form lung metastases in mice. PPP1R1B promoted degradation of p53 by stabilizing phosphorylation of MDM2 at Ser166. CONCLUSIONS: HIF1A can act a tumor suppressor by preventing the expression of PPP1R1B and subsequent degradation of the p53 protein in pancreatic cancer cells. Loss of HIF1A from pancreatic cancer cells increases their invasive and metastatic activity.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cell Movement , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/metabolism , Pancreatic Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/secondary , Cell Line, Tumor , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Male , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proteolysis , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism , Tumor Hypoxia , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics , Up-Regulation
9.
J Clin Invest ; 130(10): 5063-5073, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32809974

ABSTRACT

Hypoxia can be defined as a relative deficiency in the amount of oxygen reaching the tissues. Hypoxia-inducible factors (HIFs) are critical regulators of the mammalian response to hypoxia. In normal circumstances, HIF-1α protein turnover is rapid, and hyperglycemia further destabilizes the protein. In addition to their role in diabetes pathogenesis, HIFs are implicated in development of the microvascular and macrovascular complications of diabetes. Improving glucose control in people with diabetes increases HIF-1α protein and has wide-ranging benefits, some of which are at least partially mediated by HIF-1α. Nevertheless, most strategies to improve diabetes or its complications via regulation of HIF-1α have not currently proven to be clinically useful. The intersection of HIF biology with diabetes is a complex area in which many further questions remain, especially regarding the well-conducted studies clearly describing discrepant effects of different methods of increasing HIF-1α, even within the same tissues. This Review presents a brief overview of HIFs; discusses the range of evidence implicating HIFs in ß cell dysfunction, diabetes pathogenesis, and diabetes complications; and examines the differing outcomes of HIF-targeting approaches in these conditions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Diabetes Mellitus/physiopathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Diabetes Complications/etiology , Diabetes Complications/physiopathology , Diabetes Mellitus/etiology , Glucose/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Models, Biological , Tissue Distribution
10.
Sci Rep ; 10(1): 3906, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32127571

ABSTRACT

Hypoxia-inducible factor 1 (HIF1) is a heterodimeric transcription factor, consisting of a constitutively expressed ß-subunit (HIF1B) and a regulated α-subunit (HIF1A). In the present study, we analyzed the HIF1 driven transcriptional activity in bovine granulosa cells (GC). Treatment of GC with FSH (follicle stimulating hormone) and IGF1 (insulin-like growth factor 1) resulted in the upregulation of HIF1A mRNA expression under normoxia. Immunohistochemistry of bovine ovarian sections showed distinct staining of HIF1A in the GC layer of different staged ovarian follicles. Suppression of HIF1 using echinomycin and gene knockdown procedures revealed that HIF1 transcriptionally regulates the genes associated with steroidogenesis (STAR, HSD3B and CYP19A1) and proliferation (CCND2 and PCNA) of GC. Further, our data suggest that CYP19A1, the key gene of estradiol production, is one of the plausible downstream targets of HIF1 in bovine GC as shown by gene expression, radioimmunoassay, and chromatin precipitation analysis. Based on these results, we propose that HIF1 driven transcriptional activity plays a crucial role in GC functionality, especially steroidogenesis and proliferation in developing bovine ovarian follicles.


Subject(s)
Granulosa Cells/cytology , Granulosa Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Steroids/biosynthesis , Transcription, Genetic , Animals , Base Sequence , Cattle , Cell Proliferation/drug effects , Echinomycin/pharmacology , Female , Gene Knockdown Techniques , Granulosa Cells/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics
11.
Emerg Microbes Infect ; 9(1): 691-706, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32208814

ABSTRACT

Infection of influenza A virus (IAV) can trigger exaggerated pulmonary inflammation and induce acute lung injury (ALI). Limiting IAV replication and alleviation of pulmonary inflammation are two important therapeutic strategies for influenza virus infection. Recent studies have shown that hypoxia inducible factor-1α (HIF-1α) is an essential factor for the development and repair of ALI; however, the role and the underlying mechanisms of HIF-1α in IAV-induced ALI remain elusive. Here, we demonstrated that lung epithelial cell-specific Hif1α knockout mice infected with IAV developed more lung IAV replication and severe lung inflammation, which led to increased mortality compared to IAV-infected control mice. Moreover, knockdown of HIF1A in A549 cells (human alveolar type II epithelial cell line) promoted IAV replication in vitro. Mechanistically, knockdown of HIF1A reduced glycolysis by regulating transcription of glycolysis-related enzymes, which subsequently activated the AMPKα-ULK1 signalling pathway. Interestingly, AMPKα-ULK1 signalling promoted autophagy and augmented IAV replication. Taken together, deficiency of HIF-1α in lung epithelial cells reduces glycolysis and enhances AMPKα-ULK1-mediated autophagy, which finally facilitates IAV replication. These findings have deepened our understanding of the role of HIF-1α in regulating IAV replication and provided us novel therapeutic targets for combating influenza infection.


Subject(s)
Alveolar Epithelial Cells/physiology , Alveolar Epithelial Cells/virology , Autophagy , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Influenza A virus/physiology , Lung/virology , Orthomyxoviridae Infections/virology , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy-Related Protein-1 Homolog/metabolism , Cell Line , Cytokines/biosynthesis , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred C57BL , Orthomyxoviridae Infections/physiopathology , Pneumonia, Viral/physiopathology , Pneumonia, Viral/virology , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Up-Regulation , Virus Replication
12.
Arterioscler Thromb Vasc Biol ; 40(3): 583-596, 2020 03.
Article in English | MEDLINE | ID: mdl-31996026

ABSTRACT

OBJECTIVE: Inflammatory activation changes the mitochondrial function of macrophages from oxidative phosphorylation to reactive oxygen species production, which may promote necrotic core formation in atherosclerotic lesions. In hypoxic and cancer cells, HIF-1α (hypoxia-inducible factor) promotes oxygen-independent energy production by microRNAs. Therefore, we studied the role of HIF-1α in the regulation of macrophage energy metabolism in the context of atherosclerosis. Approach and Results: Myeloid cell-specific deletion of Hif1a reduced atherosclerosis and necrotic core formation by limiting macrophage necroptosis in apolipoprotein E-deficient mice. In inflammatory bone marrow-derived macrophages, deletion of Hif1a increased oxidative phosphorylation, ATP levels, and the expression of genes encoding mitochondrial proteins and reduced reactive oxygen species production and necroptosis. microRNA expression profiling showed that HIF-1α upregulates miR-210 and downregulates miR-383 levels in lesional macrophages and inflammatory bone marrow-derived macrophages. In contrast to miR-210, which inhibited oxidative phosphorylation and enhanced mitochondrial reactive oxygen species production, miR-383 increased ATP levels and inhibited necroptosis. The effect of miR-210 was due to targeting 2,4-dienoyl-CoA reductase, which is essential in the ß oxidation of unsaturated fatty acids. miR-383 affected the DNA damage repair pathway in bone marrow-derived macrophages by targeting poly(ADP-ribose)-glycohydrolase (Parg), which reduced energy consumption and increased cell survival. Blocking the targeting of Parg by miR-383 prevented the protective effect of Hif1a deletion in macrophages on atherosclerosis and necrotic core formation in mice. CONCLUSIONS: Our findings unveil a new mechanism by which activation of HIF-1α in inflammatory macrophages increases necroptosis through microRNA-mediated ATP depletion, thus increasing atherosclerosis by necrotic core formation.


Subject(s)
Aorta/metabolism , Atherosclerosis/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , Necroptosis , Adenosine Triphosphate/metabolism , Animals , Aorta/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Disease Models, Animal , Energy Metabolism , Gene Expression Regulation , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammation/genetics , Inflammation/pathology , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , MicroRNAs/genetics , Mitochondria/metabolism , Mitochondria/pathology , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
13.
Artif Cells Nanomed Biotechnol ; 47(1): 3657-3663, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31478766

ABSTRACT

Background: Myocardial ischemia is the main reason for ischemic heart disease. Baicalin is a plant-derived flavonoid with cardio-protective activity. Herein, we tested the influences of baicalin on cardiomyocytes H9c2 apoptosis aroused by hypoxia stimulation. Methods: Firstly, H9c2 cells were subjected to hypoxia and/or baicalin exposure. Cell viability and apoptosis, along with hypoxia-inducible factor 1α (HIF1α) and Bcl-2/adenovirus E1B 19-KDa interacting protein 3 (BNIP3) expressions were tested respectively. Then, si-HIF1α was transfected into H9c2 cells to probe whether up-regulation of HIF1α attended to the influences of baicalin on hypoxia-stimulated H9c2 cells. Finally, the regulatory effect of nuclear factor E2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway on HIF1α expression was analyzed. Results: Hypoxia exposure aroused H9c2 cell viability reduction and apoptosis. Baicalin mitigated H9c2 cell viability reduction and apoptosis aroused by hypoxia. Moreover, HIF1α/BNIP3 pathway was further activated by baicalin in hypoxia-exposed H9c2 cells. Silencing HIF1α lowered the functions of baicalin on hypoxia-exposed H9c2 cells. Besides, baicalin enhanced hypoxia-caused activation of Nrf2/HO-1 pathway. Activation of Nrf2/HO-1 pathway was associated with the up-regulation of HIF1α and protective functions of baicalin on hypoxia-exposed H9c2 cells. Conclusion: Baicalin relieved cardiomyocytes H9c2 apoptosis aroused by hypoxia might be achieved through activating Nrf2/HO-1-mediated HIF1α/BNIP3 pathway. Highlights Baicalin mitigates H9c2 cell viability loss and apoptosis aroused by hypoxia; Baicalin activates HIF1a/BNIP3 pathway in hypoxia-exposed H9c2 cells; Silencing HIF1α weakens the influences of baicalin on hypoxia-exposed H9c2 cells; Baicalin promotes Nrf2/HO-1 pathway in hypoxia-exposed H9c2 cells; Promotion of Nrf2/HO-1 pathway is related to the up-regulation of HIF1α.


Subject(s)
Apoptosis/drug effects , Flavonoids/pharmacology , Heme Oxygenase-1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Cell Hypoxia/drug effects , Cell Line , Cell Survival/drug effects , Gene Silencing , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Signal Transduction/drug effects
14.
J Neurovirol ; 25(4): 496-507, 2019 08.
Article in English | MEDLINE | ID: mdl-31025265

ABSTRACT

Microglia are resident brain macrophages with key roles in development and brain homeostasis. Cytomegalovirus (CMV) readily infects microglia cells, even as a possible primary target of infection in development. Effects of CMV infection on a cellular level in microglia are still unclear; therefore, the aim of this research was to assess the immunometabolic changes of BV-2 microglia cells following the murine cytomegalovirus (MCMV) infection. In light of that aim, we established an in vitro model of ramified BV-2 microglia (BV-2∅FCS, inducible nitric oxide synthase (iNOSlow), arginase-1 (Arg-1high), mannose receptor CD206high, and hypoxia-inducible factor 1α (HIF-1αlow)) to better replicate the in vivo conditions by removing FCS from the cultivation media, while the cells cultivated in 10% FCS DMEM displayed an ameboid morphology (BV-2FCS high, iNOShigh, Arg-1low, CD206low, and HIF-1αhigh). Experiments were performed using both ramified and ameboid microglia, and both of them were permissive to productive viral infection. Our results indicate that MCMV significantly alters the immunometabolic phenotypic properties of BV-2 microglia cells through the manipulation of iNOS and Arg-1 expression patterns, along with an induction of a glycolytic shift in the infected cell cultures.


Subject(s)
Arginase/immunology , Herpesviridae Infections/immunology , Host-Pathogen Interactions/immunology , Microglia/virology , Muromegalovirus/genetics , Nitric Oxide Synthase Type II/immunology , Animals , Arginase/genetics , Cell Line , Culture Media, Serum-Free/pharmacology , Embryo, Mammalian , Fibroblasts/immunology , Fibroblasts/virology , Gene Expression Regulation , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Host-Pathogen Interactions/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Mannose Receptor , Mannose-Binding Lectins/deficiency , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/immunology , Mice , Mice, Inbred BALB C , Microglia/immunology , Models, Biological , Muromegalovirus/growth & development , Muromegalovirus/metabolism , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/genetics , Primary Cell Culture , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Signal Transduction
15.
Circulation ; 139(24): 2778-2792, 2019 06 11.
Article in English | MEDLINE | ID: mdl-30922078

ABSTRACT

BACKGROUND: Enhancers are genomic regulatory elements conferring spatiotemporal and signal-dependent control of gene expression. Recent evidence suggests that enhancers can generate noncoding enhancer RNAs, but their (patho)biological functions remain largely elusive. METHODS: We performed chromatin immunoprecipitation-coupled sequencing of histone marks combined with RNA sequencing of left ventricular biopsies from experimental and genetic mouse models of human cardiac hypertrophy to identify transcripts revealing enhancer localization, conservation with the human genome, and hypoxia-inducible factor 1α dependence. The most promising candidate, hypoxia-inducible enhancer RNA ( HERNA)1, was further examined by investigating its capacity to modulate neighboring coding gene expression by binding to their gene promoters by using chromatin isolation by RNA purification and λN-BoxB tethering-based reporter assays. The role of HERNA1 and its neighboring genes for pathological stress-induced growth and contractile dysfunction, and the therapeutic potential of HERNA1 inhibition was studied in gapmer-mediated loss-of-function studies in vitro using human induced pluripotent stem cell-derived cardiomyocytes and various in vivo models of human pathological cardiac hypertrophy. RESULTS: HERNA1 is robustly induced on pathological stress. Production of HERNA1 is initiated by direct hypoxia-inducible factor 1α binding to a hypoxia-response element in the histoneH3-lysine27acetylation marks-enriched promoter of the enhancer and confers hypoxia responsiveness to nearby genes including synaptotagmin XVII, a member of the family of membrane-trafficking and Ca2+-sensing proteins and SMG1, encoding a phosphatidylinositol 3-kinase-related kinase. Consequently, a substrate of SMG1, ATP-dependent RNA helicase upframeshift 1, is hyperphoshorylated in a HERNA1- and SMG1-dependent manner. In vitro and in vivo inactivation of SMG1 and SYT17 revealed overlapping and distinct roles in modulating cardiac hypertrophy. Finally, in vivo administration of antisense oligonucleotides targeting HERNA1 protected mice from stress-induced pathological hypertrophy. The inhibition of HERNA1 postdisease development reversed left ventricular growth and dysfunction, resulting in increased overall survival. CONCLUSIONS: HERNA1 is a novel heart-specific noncoding RNA with key regulatory functions in modulating the growth, metabolic, and contractile gene program in disease, and reveals a molecular target amenable to therapeutic exploitation.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/prevention & control , Cardiomyopathy, Hypertrophic/prevention & control , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myocytes, Cardiac/metabolism , Oligonucleotides, Antisense/administration & dosage , RNA, Untranslated/metabolism , Animals , Binding Sites , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Case-Control Studies , Disease Models, Animal , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Promoter Regions, Genetic , RNA, Untranslated/genetics , Signal Transduction , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
16.
Front Immunol ; 10: 161, 2019.
Article in English | MEDLINE | ID: mdl-30804946

ABSTRACT

Abortions are the most important reason for unintentional childlessness. During pregnancy, maternal immune cells are in close contact to cells of the semi-allogeneic fetus. Dysregulation of the maternal immune system leading to defective adaptation to pregnancy often plays a role in pathogenesis of abortions. Myeloid-derived suppressor cells (MDSC) are myeloid cells that suppress functions of other immune cells, especially T-cells, thereby negatively affecting diseases such as cancer, sepsis or trauma. They seem, however, also necessary for maintenance of maternal-fetal tolerance. Mechanisms regulating MDSC expansion and function during pregnancy are only incompletely understood. In tumor environment, hypoxia is crucial for MDSC accumulation and activation. Hypoxia is also important for early placenta and embryo development. Effects of hypoxia are mediated through hypoxia-inducible factor 1α (HIF-1α). In the present study we aimed to examine the role of HIF-1α in myeloid cells for MDSC accumulation and MDSC function during pregnancy and for pregnancy outcome. We therefore used a mouse model with targeted deletion of HIF-1α in myeloid cells (myeloid HIF-KO) and analyzed blood, spleens and uteri of pregnant mice at gestational day E 10.5 in comparison to non-pregnant animals and wildtype (WT) animals. Further we analyzed pregnancy success by determining rates of failed implantation and abortion in WT and myeloid HIF-KO animals. We found that myeloid HIF-KO in mice led to an abrogated MDSC accumulation in the pregnant uterus and to impaired suppressive activity of MDSC. While expression of chemokine receptors and integrins on MDSC was not affected by HIF-1α, myeloid HIF-KO led to increased apoptosis rates of MDSC in the uterus. Myeloid-HIF-KO resulted in increased proportions of non-pregnant animals after positive vaginal plug and increased abortion rates, suggesting that activation of HIF-1α dependent pathways in MDSC are important for maintenance of pregnancy.


Subject(s)
Abortion, Spontaneous/etiology , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Abortion, Induced , Animals , Apoptosis , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Female , Mice , Myeloid Cells/immunology , Myeloid Cells/metabolism , Pregnancy
17.
QJM ; 111(10): 707-714, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30016480

ABSTRACT

BACKGROUND: Deficiency of hypoxia-induced factor-1α (HIF-1α) in macrophages reduced lipopolysaccharide (LPS)-induced mortality; however, whether HIF-1α expression in myeloid cells would contribute to the development of Escherichia coli (E. coli) or LPS-induced acute lung injury (ALI) is less investigated. AIM: To test whether deletion of Hif1α in myeloid cells affects E. coli or LPS-induced ALI and to elicit the underlying mechanisms. DESIGN: Laboratory study. METHODS: We intratracheally challenged Hif1αfl/fl and Hif1αfl/flLysMCre mice with E. coli or LPS to analyze lung and spleen inflammatory responses. Flow cytometry was used to analyze the changes of α7 nAChR+CD11b+ cells in the lung and spleen. Double knockout of Chrna7 and Itgam mice were used to examine expression of HIF-1α during E. coli lung infection. Vagotomy was performed to demonstrate the role of vagus nerve in mediating protective effects of deletion of Hif1α in myeloid cells on LPS-induced ALI. RESULTS: Deletion of Hif1α in myeloid cells could reduce lung edema, inflammatory cell infiltration, and lung and BAL inflammatory cytokines in E. coli-induced ALI. Flow cytometric analysis revealed that α7 nAChR+CD11b+ cells in the lung and spleen were markedly increased in E. coli-challenged Hif1αfl/flLysMCre mice compared with E. coli-challenged Hif1αfl/fl mice. Double knockout of Chrna7 and Itgam increased HIF-1α expression in lung and spleen cells during lung E. coli infection. Vagotomy abolished the protective effect of deletion of Hif1α in myeloid cells on LPS-induced ALI. CONCLUSION: Deletion of Hif1α in myeloid cells could protect mice from lung injury depending on α7 nAChR+CD11b+ cells and innervation of vagal circuits.


Subject(s)
Acute Lung Injury/prevention & control , Escherichia coli , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Lipopolysaccharides , Myeloid Cells/metabolism , Acute Lung Injury/metabolism , Animals , Cytokines/metabolism , Flow Cytometry , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Knockout , Pulmonary Edema , Spleen/metabolism , Spleen/pathology
18.
J Clin Invest ; 128(7): 3186-3197, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29911998

ABSTRACT

Although it has been reported that hypoxia inducible factor 2 α (Hif2a), a major transcriptional factor inducible by low oxygen tension, is expressed in the mouse uterus during embryo implantation, its role in pregnancy outcomes remains unclear. This study aimed to clarify functions of uterine HIF using transgenic mouse models. Mice with deletion of Hif2a in the whole uterus (Hif2a-uKO mice) showed infertility due to implantation failure. Supplementation with progesterone (P4) and leukemia inhibitory factor (LIF) restored decidual growth arrest and aberrant position of implantation sites in Hif2a-uKO mice, respectively, but did not rescue pregnancy failure. Histological analyses in Hif2a-uKO mice revealed persistence of the intact luminal epithelium, which blocked direct contact between stroma and embryo, inactivation of PI3K-AKT pathway (embryonic survival signal), and failed embryo invasion. Mice with stromal deletion of Hif2a (Hif2a-sKO mice) showed infertility with impaired embryo invasion and those with epithelial deletion of Hif2a (Hif2a-eKO mice) showed normal fertility, suggesting the importance of stromal HIF2α in embryo invasion. This was reflected in reduced expression of membrane type 2 metalloproteinase (MT2-MMP), lysyl oxidase (LOX), VEGF, and adrenomedullin (ADM) in Hif2a-uKO stroma at the attachment site, suggesting that stromal HIF2α regulates these mediators to support blastocyst invasion. These findings provide new insight that stromal HIF2α allows trophoblast invasion through detachment of the luminal epithelium and activation of an embryonic survival signal.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Embryo Implantation/physiology , Uterus/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Blastocyst/physiology , Decidua/drug effects , Decidua/physiology , Disease Models, Animal , Epithelium/physiology , Female , Fertility/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Infertility, Female/etiology , Infertility, Female/pathology , Infertility, Female/physiopathology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Mice, Transgenic , Pregnancy , Progesterone/administration & dosage , Signal Transduction
19.
Free Radic Res ; 52(11-12): 1348-1358, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29764240

ABSTRACT

Iron or oxygen regulates the stability of hypoxia inducible factor-1α (HIF-1α). We investigated whether ferrous glycinate would affect HIF-1α accumulation, aerobic glycolysis and mitochondrial energy metabolism in human A549 lung cancer cells. Incubation of A549 cells with ferrous glycinate decreased the protein levels of HIF-1α, which was abrogated by proteosome inhibitor, or prolyl hydroxylase inhibitor. The addition of ferrous glycinate decreased protein levels of glucose transporter-1, hexokinase-2, and lactate dehydrogenase A, and decreased pyruvate dehydrogenase kinase-1 (PDK-1) and pyruvate dehydrogenase (PDH) phosphorylation in A549 cells. Ferrous glycinate also increased the expression of the mitochondrial transcription factor A (TFAM), and the mitochondrial protein, cytochrome c oxidase (COX-IV). Silencing of HIF-1α expression mimicked the effects of ferrous glycinate on PDK-1, PDH, TFAM and COX-IV in A549 cells. Ferrous glycinate increased mitochondrial membrane potential and ATP production in A549 cells. These results suggest that ferrous glycinate may reverse Warburg effect through down regulating HIF-1α in A549 cells.


Subject(s)
Energy Metabolism/drug effects , Ferrous Compounds/pharmacology , Glycine/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , A549 Cells , Dose-Response Relationship, Drug , Ferrous Compounds/chemistry , Glycine/analogs & derivatives , Glycine/chemistry , Humans , Membrane Potential, Mitochondrial/drug effects , Structure-Activity Relationship , Tumor Cells, Cultured
20.
Keio J Med ; 67(1): 1-9, 2018 Mar 23.
Article in English | MEDLINE | ID: mdl-28592747

ABSTRACT

The hypoxia response is a fundamental phenomenon mainly regulated by hypoxia-inducible factors (HIFs). For more than a decade, we have investigated and revealed the roles of the hypoxia response in the development, physiology, and pathophysiology of the retina by generating and utilizing cell-type-specific conditional knockout mice. To investigate the functions of genes related to the hypoxia response in cells composing the retina, we generated various mouse lines that lack HIFs and/or related genes specifically in retinal neurons, astrocytes, myeloid cells, or retinal pigment epithelium cells. We found that these genes in the different types of retinal cells contribute in various ways to the homeostasis of ocular vascular and visual function. We hypothesized that the activation of HIFs is likely involved in the development and progress of retinal diseases, and we subsequently confirmed the pathological roles of HIFs in animal models of neovascular and atrophic ocular diseases. Currently, anti-vascular endothelial growth factor (anti-VEGF) therapy is a first-line treatment widely used for neovascular retinal diseases. However, alternative or additional targets are now required because several recent large-scale clinical trials and animal studies, including our own research, have indicated that VEGF antagonism may induce retinal vascular and neuronal degeneration. We have identified and confirmed a microRNA as a candidate for an alternative target against neovascular retinal diseases, and we are now working to establish a novel HIF inhibitor for clinical use based on the disease mechanism that we identified.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia/drug therapy , MicroRNAs/genetics , Neovascularization, Pathologic/prevention & control , Vascular Endothelial Growth Factor A/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Angiogenesis Inhibitors/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Developmental , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Mice , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Retinal Pigment Epithelium/blood supply , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/deficiency , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...