Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Cell Mol Life Sci ; 81(1): 72, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38300329

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1) is the key transcriptional mediator of the cellular response to hypoxia and is also involved in cancer progression. Regulation of its oxygen-sensitive HIF-1α subunit involves post-translational modifications that control its stability, subcellular localization, and activity. We have previously reported that phosphorylation of the HIF-1α C-terminal domain by ERK1/2 promotes HIF-1α nuclear accumulation and stimulates HIF-1 activity while lack of this modification triggers HIF-1α nuclear export and its association with mitochondria. On the other hand, modification of the N-terminal domain of HIF-1α by CK1δ impairs HIF-1 activity by obstructing the formation of a HIF-1α/ARNT heterodimer. Investigation of these two antagonistic events by expressing double phospho-site mutants in HIF1A-/- cells under hypoxia revealed independent and additive phosphorylation effects that can create a gradient of HIF-1α subcellular localization and transcriptional activity. Furthermore, modification by CK1δ caused mitochondrial release of the non-nuclear HIF-1α form and binding to microtubules via its N-terminal domain. In agreement, endogenous HIF-1α could be shown to co-localize with mitotic spindle microtubules and interact with tubulin, both of which were inhibited by CK1δ silencing or inhibition. Moreover, CK1δ expression was necessary for equal partitioning of mother cell-produced HIF-1α to the daughter cell nuclei at the end of mitosis. Overall, our results suggest that phosphorylation by CK1δ stimulates the association of non-nuclear HIF-1α with microtubules, which may serve as a means to establish a symmetric distribution of HIF-1α during cell division under low oxygen conditions.


Subject(s)
MAP Kinase Signaling System , Protein Kinases , Humans , Mitosis , Microtubules , Hypoxia , Oxygen
2.
Cell Mol Life Sci ; 81(1): 58, 2024 Jan 27.
Article in English | MEDLINE | ID: mdl-38279024

ABSTRACT

Reduced oxygen availability (hypoxia) triggers adaptive cellular responses via hypoxia-inducible factor (HIF)-dependent transcriptional activation. Adaptation to hypoxia also involves transcription-independent processes like post-translational modifications; however, these mechanisms are poorly characterized. Investigating the involvement of protein SUMOylation in response to hypoxia, we discovered that hypoxia strongly decreases the SUMOylation of Exosome subunit 10 (EXOSC10), the catalytic subunit of the RNA exosome, in an HIF-independent manner. EXOSC10 is a multifunctional exoribonuclease enriched in the nucleolus that mediates the processing and degradation of various RNA species. We demonstrate that the ubiquitin-specific protease 36 (USP36) SUMOylates EXOSC10 and we reveal SUMO1/sentrin-specific peptidase 3 (SENP3) as the enzyme-mediating deSUMOylation of EXOSC10. Under hypoxia, EXOSC10 dissociates from USP36 and translocates from the nucleolus to the nucleoplasm concomitant with its deSUMOylation. Loss of EXOSC10 SUMOylation does not detectably affect rRNA maturation but affects the mRNA transcriptome by modulating the expression levels of hypoxia-related genes. Our data suggest that dynamic modulation of EXOSC10 SUMOylation and localization under hypoxia regulates the RNA degradation machinery to facilitate cellular adaptation to low oxygen conditions.


Subject(s)
Exosomes , Transcriptome , Humans , Exosomes/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Transcriptional Activation , Oxygen/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Sumoylation , Exoribonucleases/genetics , Exoribonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/genetics , Exosome Multienzyme Ribonuclease Complex/metabolism , Cysteine Endopeptidases/metabolism , Ubiquitin Thiolesterase/metabolism
3.
J Cell Sci ; 132(7)2019 04 08.
Article in English | MEDLINE | ID: mdl-30962349

ABSTRACT

Hypoxia-inducible factor 2 (HIF-2) is a principal component of the cellular response to oxygen deprivation (hypoxia). Its inducible subunit, HIF-2α (also known as EPAS1), is controlled by oxygen-dependent as well as oxygen-independent mechanisms, such as phosphorylation. We show here that HIF-2α is phosphorylated under hypoxia (1% O2) by extracellular signal-regulated protein kinases 1 and 2 (ERK1/2; also known as MAPK3 and MAPK1, respectively) at serine residue 672, as identified by in vitro phosphorylation assays. Mutation of this site to an alanine residue or inhibition of the ERK1/2 pathway decreases HIF-2 transcriptional activity and causes HIF-2α to mislocalize to the cytoplasm without changing its protein expression levels. Localization, reporter gene and immunoprecipitation experiments further show that HIF-2α associates with the exportin chromosomal maintenance 1 (CRM1, also known as XPO1) in a phosphorylation-sensitive manner and identify two critical leucine residues as part of an atypical CRM1-dependent nuclear export signal (NES) neighboring serine 672. Inhibition of CRM1 or mutation of these residues restores nuclear accumulation and activity of HIF-2α lacking the ERK1/2-mediated modification. In summary, we reveal a novel regulatory mechanism of HIF-2, involving ERK1/2-dependent phosphorylation of HIF-2α, which controls its nucleocytoplasmic shuttling and the HIF-2 transcriptional activity.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Nucleus/metabolism , Karyopherins/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Active Transport, Cell Nucleus , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Nucleus/genetics , HeLa Cells , Humans , Karyopherins/genetics , Mitogen-Activated Protein Kinase 3/genetics , Mutation , Phosphorylation , Receptors, Cytoplasmic and Nuclear/genetics , Serine/metabolism , Exportin 1 Protein
4.
Mol Cell Proteomics ; 18(6): 1197-1209, 2019 06.
Article in English | MEDLINE | ID: mdl-30926672

ABSTRACT

Hypoxia occurs in pathological conditions, such as cancer, as a result of the imbalance between oxygen supply and consumption by proliferating cells. HIFs are critical molecular mediators of the physiological response to hypoxia but also regulate multiple steps of carcinogenesis including tumor progression and metastasis. Recent data support that sumoylation, the covalent attachment of the Small Ubiquitin-related MOdifier (SUMO) to proteins, is involved in the activation of the hypoxic response and the ensuing signaling cascade. To gain insights into differences of the SUMO1 and SUMO2/3 proteome of HeLa cells under normoxia and cells grown for 48 h under hypoxic conditions, we employed endogenous SUMO-immunoprecipitation in combination with quantitative mass spectrometry (SILAC). The group of proteins whose abundance was increased both in the total proteome and in the SUMO IPs from hypoxic conditions was enriched in enzymes linked to the hypoxic response. In contrast, proteins whose SUMOylation status changed without concomitant change in abundance were predominantly transcriptions factors or transcription regulators. Particularly interesting was transcription factor TFAP2A (Activating enhancer binding Protein 2 alpha), whose sumoylation decreased on hypoxia. TFAP2A is known to interact with HIF-1 and we provide evidence that deSUMOylation of TFAP2A enhances the transcriptional activity of HIF-1 under hypoxic conditions. Overall, these results support the notion that SUMO-regulated signaling pathways contribute at many distinct levels to the cellular response to low oxygen.


Subject(s)
Gene Expression Regulation/drug effects , Oxygen/pharmacology , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription, Genetic/drug effects , Amino Acid Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lysine/metabolism , Protein Binding/drug effects , Substrate Specificity/drug effects , Sumoylation/drug effects , Transcription Factor AP-2/chemistry , Transcription Factor AP-2/metabolism
5.
Cell Mol Life Sci ; 76(4): 809-825, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30535970

ABSTRACT

Hypoxia is frequently encountered in the microenvironment of solid tumors. Hypoxia-inducible factors (HIFs), the main effectors of cell response to hypoxia, promote cancer cell survival and progression. HIF-1α, the oxygen-regulated subunit of HIF-1, is often correlated with oncogenesis and represents an attractive therapeutic target. We have previously reported that activation HIF-1α by ERK involves modification of two serine residues and masking of a nuclear export signal (NES), all inside a 43-amino acid domain termed ERK Targeted Domain (ETD). Overexpression of ETD variants including wild-type, phospho-mimetic (SE) or NES-less (IA) mutant forms caused HIF-1 inactivation in two hepatocarcinoma cell lines, while a phospho-deficient (SA) form was ineffective and acted as a sequence-specific negative control. To deliver these ETD forms directly into cancer cells, they were fused to the HIV TAT-sequence and produced as cell-permeable peptides. When the TAT-ETD peptides were added to the culture medium of Huh7 cells, they entered the cells and, with the exception of ETD-SA, accumulated inside the nucleus, caused mislocalization of endogenous HIF-1α to the cytoplasm, significant reduction of HIF-1 activity and inhibition of expression of specific HIF-1, but not HIF-2, gene targets under hypoxia. More importantly, transduced nuclear TAT-ETD peptides restricted migration, impaired colony formation and triggered apoptotic cell death of cancer cells grown under hypoxia, while they produced no effects in normoxic cells. These data demonstrate the importance of ERK-mediated activation of HIF-1 for low oxygen adaptation and the applicability of ETD peptide derivatives as sequence-specific HIF-1 and cancer cell growth inhibitors under hypoxia.


Subject(s)
Apoptosis/physiology , Cell-Penetrating Peptides/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Amino Acid Sequence , Apoptosis/drug effects , Apoptosis/genetics , Cell Hypoxia , Cell Line, Tumor , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/pharmacology , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Export Signals/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Sequence Homology, Amino Acid , tat Gene Products, Human Immunodeficiency Virus/genetics
6.
J Cell Sci ; 130(2): 466-479, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27909249

ABSTRACT

Hypoxia inducible factor-1 (HIF-1) is the main transcriptional activator of the cellular response to hypoxia and an important target of anticancer therapy. Phosphorylation by ERK1 and/or ERK2 (MAPK3 and MAPK1, respectively; hereafter ERK) stimulates the transcriptional activity of HIF-1α by inhibiting its CRM1 (XPO1)-dependent nuclear export. Here, we demonstrate that phosphorylation by ERK also regulates the association of HIF-1α with a so-far-unknown interaction partner identified as mortalin (also known as GRP75 and HSPA9), which mediates non-genomic involvement of HIF-1α in apoptosis. Mortalin binds specifically to HIF-1α that lacks modification by ERK, and the HIF-1α-mortalin complex is localized outside the nucleus. Under hypoxia, mortalin mediates targeting of unmodified HIF-1α to the outer mitochondrial membrane, as well as association with VDAC1 and hexokinase II, which promotes production of a C-terminally truncated active form of VDAC1, denoted VDAC1-ΔC, and protection from apoptosis when ERK is inactivated. Under normoxia, transcriptionally inactive forms of unmodified HIF-1α or its C-terminal domain alone are also targeted to mitochondria, stimulate production of VDAC1-ΔC and increase resistance to etoposide- or doxorubicin-induced apoptosis. These findings reveal an ERK-controlled, unconventional and anti-apoptotic function of HIF-1α that might serve as an early protective mechanism upon oxygen limitation and promote cancer cell resistance to chemotherapy.


Subject(s)
Apoptosis , Extracellular Signal-Regulated MAP Kinases/metabolism , HSP70 Heat-Shock Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondria/metabolism , Cell Hypoxia , Enzyme Activation , Green Fluorescent Proteins/metabolism , HeLa Cells , Hexokinase/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Mitochondrial Membranes/metabolism , Protein Domains , Protein Transport , Voltage-Dependent Anion Channel 1/metabolism
7.
Arch Biochem Biophys ; 664: 174-182, 2019 03 30.
Article in English | MEDLINE | ID: mdl-30776328

ABSTRACT

Hypoxia-inducible factors (HIF) are master regulators of the response to hypoxia. Although several kinases are known to modify their oxygen sensitive HIF-α subunits or affect indirectly their function, little is known about the role of phosphatases in HIF control. To address this issue, a library containing siRNAs for the 25 known catalytic subunits of human phosphatases was used to screen for their effect on HIF transcriptional activity in HeLa cells. Serine-threonine phosphatase PPP3CA (calcineurin A, isoform a) was identified as the strongest candidate for a negative regulator of HIF activity. Indeed, independent silencing of PPP3CA expression stimulated HIF transcriptional activity under hypoxia, without increasing the protein levels of HIF-1α or HIF-2α. Overexpression of a constitutively active PPP3CA form, but not its catalytically inactive counterpart, inhibited HIF activity and expression of HIF target genes but did not affect HIF-1α or HIF-2α expression. These results were phenocopied by treatment with the ionophore ionomycin, that activates endogenous PPP3CA. The effect of ionomycin was mediated by PPP3CA as it was largely abolished by PPP3CA silencing. Furthermore, ionomycin enhanced the down-regulation of HIF activity by wild-type PPP3CA overexpression. Overall, our results suggest the involvement of PPP3CA in fine-tuning the HIF-dependent transcriptional response to hypoxia.


Subject(s)
Calcineurin/metabolism , Down-Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Transcription, Genetic , Calcineurin/genetics , Enzyme Activation , Gene Silencing , HeLa Cells , Humans , Ionomycin/pharmacology
8.
J Cell Sci ; 129(22): 4213-4226, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27686097

ABSTRACT

Hypoxia inducible factor 2 (HIF-2) is a transcriptional activator implicated in the cellular response to hypoxia. Regulation of its inducible subunit, HIF-2α (also known as EPAS1), involves post-translational modifications. Here, we demonstrate that casein kinase 1δ (CK1δ; also known as CSNK1D) phosphorylates HIF-2α at Ser383 and Thr528 in vitro We found that disruption of these phosphorylation sites, and silencing or chemical inhibition of CK1δ, reduced the expression of HIF-2 target genes and the secretion of erythropoietin (EPO) in two hepatic cancer cell lines, Huh7 and HepG2, without affecting the levels of HIF-2α protein expression. Furthermore, when CK1δ-dependent phosphorylation of HIF-2α was inhibited, we observed substantial cytoplasmic mislocalization of HIF-2α, which was reversed upon the addition of the nuclear protein export inhibitor leptomycin B. Taken together, these data suggest that CK1δ enhances EPO secretion from liver cancer cells under hypoxia by modifying HIF-2α and promoting its nuclear accumulation. This modification represents a new mechanism of HIF-2 regulation that might allow HIF isoforms to undertake differing functions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Casein Kinase Idelta/metabolism , Erythropoietin/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Active Transport, Cell Nucleus , Amino Acid Sequence , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Hypoxia , Cell Nucleus/metabolism , Gene Silencing , HeLa Cells , Hep G2 Cells , Humans , Karyopherins/metabolism , Liver Neoplasms/genetics , Mutation/genetics , Phosphorylation , Protein Binding , Protein Stability , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription, Genetic , Upstream Stimulatory Factors/metabolism , Exportin 1 Protein
9.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(9): 1142-1152, 2018 09.
Article in English | MEDLINE | ID: mdl-29908837

ABSTRACT

Hypoxia inducible factor-1 (HIF-1) supports survival of normal cells under low oxygen concentration and cancer cells in the hypoxic tumor microenvironment. This involves metabolic reprogramming via upregulation of glycolysis, downregulation of oxidative phosphorylation and, less well documented, effects on lipid metabolism. To investigate the latter, we examined expression of relevant enzymes in cancer cells grown under hypoxia. We show that expression of acylglycerol-3-phosphate acyltransferase 2 (AGPAT2), also known as lysophosphatidic acid acyltransferase ß (LPAATß), was upregulated under hypoxia and this was impaired by siRNA-mediated knockdown of HIF-1α. Moreover, a sequence of the AGPAT2 gene promoter region, containing 6 putative Hypoxia Response Elements (HREs), activated transcription of a reporter gene under hypoxic conditions or in normoxic cells over-expressing HIF-1α. Chromatin immunoprecipitation experiments confirmed binding of HIF-1α to one of these HREs, mutation of which abolished hypoxic activation of the AGPAT2 promoter. Knockdown of AGPAT2 by siRNA reduced lipid droplet accumulation and cell viability under hypoxia and increased cancer cell sensitivity to the chemotherapeutic etoposide. In conclusion, our findings demonstrate that AGPAT2, which is mutated in patients with congenital generalized lipodystrophy and over-expressed in different types of cancer, is a direct transcriptional target of HIF-1, suggesting that upregulation of lipid storage by HIF-1 plays an important role in adaptation and survival of cancer cells under low oxygen conditions.


Subject(s)
Acyltransferases/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Hepatocytes/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lipid Metabolism/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Acyltransferases/antagonists & inhibitors , Acyltransferases/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Etoposide/pharmacology , Glycerophospholipids/biosynthesis , HEK293 Cells , HeLa Cells , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lipid Droplets/drug effects , Lipid Droplets/metabolism , Lipid Metabolism/drug effects , Mutation , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Response Elements , Signal Transduction , Transcriptional Activation , Triglycerides/biosynthesis
10.
Cell Mol Life Sci ; 71(21): 4243-58, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24718935

ABSTRACT

Hepatitis C virus (HCV) infection is associated with hepatic iron overload and elevated serum iron that correlate to poor antiviral responses. Hepcidin (HAMP), a 25-aa cysteine-rich liver-specific peptide, controls iron homeostasis. Its expression is up-regulated in inflammation and iron excess. HCV-mediated hepcidin regulation remains controversial. Chronic HCV patients possess relatively low hepcidin levels; however, elevated HAMP mRNA has been reported in HCV core transgenic mice and HCV replicon-expressing cells. We investigated the effect of HCV core protein on HAMP gene expression and delineated the complex interplay of molecular mechanisms involved. HCV core protein up-regulated HAMP promoter activity, mRNA, and secreted protein levels. Enhanced promoter activity was abolished by co-transfections of core with HAMP promoter constructs containing mutated/deleted BMP and STAT binding sites. Dominant negative constructs, pharmacological inhibitors, and silencing experiments against STAT3 and SMAD4 confirmed the participation of both pathways in HAMP gene regulation by core protein. STAT3 and SMAD4 expression levels were found increased in the presence of HCV core, which orchestrated SMAD4 translocation into the nucleus and STAT3 phosphorylation. To further understand the mechanisms governing the core effect, the role of the JAK/STAT-activating kinase CK2 was investigated. A CK2-dominant negative construct, a CK2-specific inhibitor, and RNAi interference abrogated the core-induced increase on HAMP promoter activity, mRNA, and protein levels, while CK2 acted in synergy with core to significantly enhance HAMP gene expression. Therefore, HCV core up-regulates HAMP gene transcription via a complex signaling network that requires both SMAD/BMP and STAT3 pathways and CK2 involvement.


Subject(s)
Casein Kinase II/metabolism , Gene Expression Regulation, Enzymologic , Hepacivirus/metabolism , Hepcidins/metabolism , Viral Core Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Gene Expression Regulation, Viral , Gene Silencing , Hep G2 Cells , Homeostasis , Humans , Iron/metabolism , Promoter Regions, Genetic , RNA Interference , STAT3 Transcription Factor/metabolism , Signal Transduction , Smad4 Protein/metabolism , Up-Regulation
11.
J BUON ; 20(3): 680-9, 2015.
Article in English | MEDLINE | ID: mdl-26214618

ABSTRACT

Colorectal cancer (CRC) is the third most common cancer worldwide and despite the abundance of molecular pathways and markers continually being reported, the mortality rates remain high. Hypoxia inducible factor 1alpha (HIF-1α) plays a major role in the response of tumors to hypoxia, and contributes to tumor aggressiveness, invasiveness and resistance to radiotherapy and chemotherapy. Targeting HIF-1α is an attractive strategy, with the potential for disrupting multiple pathways crucial for tumor growth. In the current study, HIF-1α immunohistochemical expression in CRC is reviewed along with the relation to clinical outcome and prognosis. In addition, the significant correlation of HIF-1α to vascular endothelial growth factor (VEGF) expression is reported, as well as the possible role of HIF-1α in predicting the therapeutic response to anti-EGFR therapies. Herein, an overview of the HIF-1α expression in CRC is presented. This review delineates the crucial role that HIF-1α plays in carcinogenesis, tumor angiogenesis and cancer progression. The evaluation of HIF-1α in patient biopsies could be useful as a prognostic and/or predictive biomarker in personalized cancer treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Biomarkers, Tumor/genetics , Cell Hypoxia , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Molecular Targeted Therapy , Neovascularization, Pathologic , Prognosis , Signal Transduction , Tumor Microenvironment
12.
Biochim Biophys Acta ; 1833(6): 1378-87, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23458834

ABSTRACT

Hypoxia inducible factor-1 (HIF-1), a dimeric transcription factor of the bHLH-PAS family, is comprised of HIF-1α, which is inducible by hypoxia and ARNT or HIF-1ß, which is constitutively expressed. HIF-1 is involved in cellular homeostasis under hypoxia, in development and in several diseases affected by oxygen availability, particularly cancer. Since its expression is positively correlated with poor outcome prognosis for cancer patients, HIF-1 is a target for pharmaceutical therapy. We have previously shown that male germ cell Rac GTPase activating protein (MgcRacGAP), a regulator of Rho proteins which are principally involved in cytoskeletal organization, binds to HIF-1α and inhibits its transcriptional activity. In this work, we have explored the mechanism of the MgcRacGAP-mediated HIF-1 inactivation. We show that the Myo domain of MgcRacGAP, which is both necessary and sufficient for HIF-1 repression, binds to the PAS-B domain of HIF-1α. Furthermore MgcRacGAP competes with ARNT for binding to the HIF-1α PAS-B domain, as shown by in vitro binding pull down assays. In mammalian cells, ARNT overexpression can overcome the MgcRacGAP-mediated inhibition and MgcRacGAP binding to HIF-1α in vivo inhibits its dimerization with ARNT. We additionally present results indicating that MgcRacGAP binding to HIF-1α is specific, since it does not affect the transcriptional activity of HIF-2, a close evolutionary relative of HIF-1 also involved in hypoxia regulation and cancer. Our results reveal a new mechanism for HIF-1 transcriptional activity regulation, suggest a novel hypoxia-cytoskeleton link and provide new tools for selective HIF-1 inhibition.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , GTPase-Activating Proteins/metabolism , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia , Transcription, Genetic , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Blotting, Western , Cells, Cultured , Cytoskeleton , GTPase-Activating Proteins/genetics , Humans , Immunoprecipitation , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Transcriptional Activation
13.
J Cell Sci ; 125(Pt 14): 3485-93, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22467849

ABSTRACT

Adaptation to hypoxia involves hypoxia-inducible transcription factors (HIFs) and requires reprogramming of cellular metabolism that is essential during both physiological and pathological processes. In contrast to the established role of HIF-1 in glucose metabolism, the involvement of HIFs and the molecular mechanisms concerning the effects of hypoxia on lipid metabolism are poorly characterized. Here, we report that exposure of human cells to hypoxia causes accumulation of triglycerides and lipid droplets. This is accompanied by induction of lipin 1, a phosphatidate phosphatase isoform that catalyzes the penultimate step in triglyceride biosynthesis, whereas lipin 2 remains unaffected. Hypoxic upregulation of lipin 1 expression involves predominantly HIF-1, which binds to a single distal hypoxia-responsive element in the lipin 1 gene promoter and causes its activation under low oxygen conditions. Accumulation of hypoxic triglycerides or lipid droplets can be blocked by siRNA-mediated silencing of lipin 1 expression or kaempferol-mediated inhibition of HIF-1. We conclude that direct control of lipin 1 transcription by HIF-1 is an important regulatory feature of lipid metabolism and its adaptation to hypoxia.


Subject(s)
Cell Hypoxia/physiology , Hypoxia-Inducible Factor 1/metabolism , Phosphatidate Phosphatase/biosynthesis , Triglycerides/metabolism , Cell Hypoxia/genetics , Cell Line, Tumor , HeLa Cells , Humans , Hypoxia-Inducible Factor 1/genetics , Phosphatidate Phosphatase/genetics , Promoter Regions, Genetic , Triglycerides/biosynthesis , Triglycerides/genetics , Up-Regulation
14.
J Cell Physiol ; 228(8): 1745-53, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23359428

ABSTRACT

Airway smooth muscle cells (ASMCs) participate in tissue remodeling characteristic of airway inflammatory diseases like asthma. Inflammation and hypoxia pathways are often interconnected and the regulatory subunit of the hypoxia inducible factor, HIF-1α, has been recently shown to be induced by cytokines. Here we investigate the effect of individual or combined treatment of ASMCs with the inflammatory mediator TNFα and/or hypoxia on the expression of HIF-1α, HIF-1 targets and inflammation markers. TNFα enhances HIF-1α protein and mRNA levels, under both normoxia and hypoxia. TNFα-mediated induction of HIF-1α gene transcription is repressed by inhibition of the NF-κB pathway. Despite the up-regulation of HIF-1α protein, the transcription of HIF-1 target genes remains low in the presence of TNFα at normoxia and is even reduced at hypoxia. We show that the reduction in HIF-1 transcriptional activity by TNFα is due to inhibition of the interaction of HIF-1α with ARNT and subsequent blocking of its binding to HREs. Comparison between hypoxia and TNFα for their effects on the expression of inflammatory markers shows significant differences: hypoxia up-regulates the expression of IL-6, but not RANTES or ICAM, and reduces the induction of VCAM by TNFα. Finally, ex vivo treatment of rabbit trachea strips with TNFα increases HIF-1α protein levels, but reduces the expression of HIF-1 targets under hypoxia. Overall, TNFα induces HIF-1α mRNA synthesis via an NF-κB dependent pathway but inhibits binding of HIF-1α to ARNT and DNA, while hypoxia and TNFα have distinct effects on ASMC inflammatory gene expression.


Subject(s)
Bronchi/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Trachea/metabolism , Tumor Necrosis Factor-alpha/physiology , Up-Regulation , Animals , Bronchi/cytology , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cells, Cultured , Gene Targeting , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Rabbits , Trachea/cytology , Up-Regulation/genetics
15.
Biochem Biophys Res Commun ; 435(4): 540-5, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23665013

ABSTRACT

Hepcidin, a hepatic hormone, regulates serum iron levels by controlling both intestinal iron absorption and iron release from macrophages. Although transcription of hepcidin is controlled by diverse stimuli, it remains elusive if post-transcriptional steps of its production are also regulated. To address this issue, GFP was fused to the C-terminus of hepcidin and the chimeric hepcidin-GFP protein was expressed in hepatoma Huh7 cells. Expression and secretion of hepcidin-GFP were analyzed by fluorescence microscopy or western blotting and its activity was assessed by in vitro biological assays. Transient over-expression of hepcidin-GFP resulted in production and secretion of premature forms. On the other hand, stable low-level expression led to synthesis and secretion of a properly matured hepcidin-GFP. This form was biologically active since it affected appropriately the levels of IRP2 and ferritin in human THP1 monocytes and targeted ferroportin in mouse J774 macrophages. Treatment of hepcidin-GFP expressing cells with hypoxia (0.1% O2) altered the subcellular distribution of pro-hepcidin-GFP and significantly reduced the secretion of mature hepcidin-GFP. Our hepcidin-GFP expression system allows the investigation of post-transcriptional processing of hepcidin and implicates hypoxia in its secretion control.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Carcinoma, Hepatocellular/metabolism , Chimera/metabolism , Green Fluorescent Proteins/metabolism , Liver Neoplasms/metabolism , Oxygen/metabolism , Animals , Cell Line, Tumor , Chimera/genetics , Green Fluorescent Proteins/genetics , Hepcidins , Humans , Macrophages , Mice , Recombinant Proteins/metabolism
16.
Cytokine ; 62(1): 38-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23517877

ABSTRACT

Angiogenesis has been considered to be an important step in the initiation and progression of chronic diseases such as psoriasis. The hypoxia inducible factor 1 alpha (HIF-1α), a critical hypoxia-induced factor that regulates angiogenesis has been shown previously to be over-expressed in psoriasis skin both at the mRNA and protein level. In this report we confirm HIF-1α and IL-6 over-expression in psoriatic patients using immunoenzymometric assay and found that the expression of HIF-1α is closely correlated with IL-6 expression (r = 0.61 and p = 0.005), suggesting a close interaction of HIF-1α and IL-6 in the psoriasis immuno-microenvironment. Our findings merit further in vitro and in vivo work before solid suggestions can be made for therapeutic interventions that target HIF-1α pathway and/or IL-6.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/blood , Interleukin-6/blood , Psoriasis/blood , Case-Control Studies , Female , Humans , Male , Middle Aged
17.
FEBS J ; 290(15): 3764-3780, 2023 08.
Article in English | MEDLINE | ID: mdl-36825991

ABSTRACT

Hypoxia-inducible factor-1, a heterodimer of alpha (HIF-1α) and beta (HIF-1ß or ARNT) subunits, is a major regulator of the transcriptional response to hypoxia. However, HIF-1α, the oxygen-regulated subunit, also exerts nontranscriptional functions through interaction with proteins other than ARNT. We have previously shown that the subcellular localization and protein interactions of HIF-1α are controlled by ERK-mediated phosphorylation at Ser641/643. When HIF-1α is modified at these sites, it is nuclear, binds to ARNT, interacts with nucleophosmin 1 (NPM1) and activates transcription of hypoxia-target genes. On the contrary, unmodified HIF-1α is bound by chromosomal region maintenance 1 (CRM1), exits the nucleus and, via its association with mortalin, is targeted to the mitochondria to form an antiapoptotic complex. To further characterize the latter function, recombinant fragments of HIF-1α and mortalin were used for in vitro binding assays and immunoprecipitation experiments to map the respective binding sites and show that their interaction is direct and functional. We could also show that embelin, a natural product and known inhibitor of the mortalin-p53 interaction, also disrupts the mortalin-HIF-1α association and, furthermore, removes unmodified HIF-1α from mitochondria. Mitochondrial dissociation of HIF-1α, either by embelin or overexpression of a HIF-1α peptide harbouring the mortalin binding site, under stress conditions leads to mitochondrial localization of the pro-apoptotic protein B-cell lymphoma 2-associated X protein (Bax) and induction of apoptosis. We suggest that when ERK activity is low under hypoxia, binding of HIF-1α to mortalin inhibits mitochondrial recruitment of Bax and protects cells from apoptotic cell death.


Subject(s)
Hypoxia , Mitochondria , Humans , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , Cell Hypoxia/physiology , Mitochondria/metabolism , Hypoxia/metabolism , Apoptosis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
18.
FEBS J ; 290(8): 2165-2179, 2023 04.
Article in English | MEDLINE | ID: mdl-36453802

ABSTRACT

Cancer cells, when exposed to the hypoxic tumour microenvironment, respond by activating hypoxia-inducible factors (HIFs). HIF-1 mediates extensive metabolic re-programming, and expression of HIF-1α, its oxygen-regulated subunit, is associated with poor prognosis in cancer. Here we analyse the role of pyruvate dehydrogenase phosphatase 1 (PDP1) in the regulation of HIF-1 activity. PDP1 is a key hormone-regulated metabolic enzyme that dephosphorylates and activates pyruvate dehydrogenase (PDH), thereby stimulating the conversion of pyruvate into acetyl-CoA. Silencing of PDP1 down-regulated HIF transcriptional activity and the expression of HIF-dependent genes, including that of PDK1, the kinase that phosphorylates and inactivates PDH, opposing the effects of PDP1. Inversely, PDP1 stimulation enhanced HIF activity under hypoxia. Alteration of PDP1 levels or activity did not have an effect on HIF-1α protein levels, nuclear accumulation or interaction with its partners ARNT and NPM1. However, depletion of PDP-1 decreased histone H3 acetylation of HIF-1 target gene promoters and inhibited binding of HIF-1 to the respective hypoxia-response elements (HREs) under hypoxia. Furthermore, the decrease of HIF transcriptional activity upon PDP1 depletion could be reversed by treating the cells with acetate, as an exogenous source of acetyl-CoA, or the histone deacetylase (HDAC) inhibitor trichostatin A. These data suggest that the PDP1/PDH/HIF-1/PDK1 axis is part of a homeostatic loop which, under hypoxia, preserves cellular acetyl-CoA production to a level sufficient to sustain chromatin acetylation and transcription of hypoxia-inducible genes.


Subject(s)
Histones , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase , Humans , Acetyl Coenzyme A/metabolism , Acetylation , Cell Hypoxia/genetics , Histones/genetics , Histones/metabolism , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/genetics , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/metabolism , Hypoxia-Inducible Factor 1
19.
Cells ; 12(5)2023 03 03.
Article in English | MEDLINE | ID: mdl-36899934

ABSTRACT

The Hypoxia Inducible Factor 1 (HIF-1) plays a major role in the cellular response to hypoxia by regulating the expression of many genes involved in adaptive processes that allow cell survival under low oxygen conditions. Adaptation to the hypoxic tumor micro-environment is also critical for cancer cell proliferation and therefore HIF-1 is also considered a valid therapeutical target. Despite the huge progress in understanding regulation of HIF-1 expression and activity by oxygen levels or oncogenic pathways, the way HIF-1 interacts with chromatin and the transcriptional machinery in order to activate its target genes is still a matter of intense investigation. Recent studies have identified several different HIF-1- and chromatin-associated co-regulators that play important roles in the general transcriptional activity of HIF-1, independent of its expression levels, as well as in the selection of binding sites, promoters and target genes, which, however, often depends on cellular context. We review here these co-regulators and examine their effect on the expression of a compilation of well-characterized HIF-1 direct target genes in order to assess the range of their involvement in the transcriptional response to hypoxia. Delineating the mode and the significance of the interaction between HIF-1 and its associated co-regulators may offer new attractive and specific targets for anticancer therapy.


Subject(s)
Hypoxia-Inducible Factor 1 , Neoplasms , Humans , Hypoxia-Inducible Factor 1/metabolism , Hypoxia/genetics , Promoter Regions, Genetic , Neoplasms/genetics , Chromatin , Oxygen , Tumor Microenvironment
20.
J Cell Sci ; 123(Pt 17): 2976-86, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20699359

ABSTRACT

Hypoxia-inducible factor 1 (HIF-1), a transcriptional activator that mediates cellular response to hypoxia and a promising target of anticancer therapy, is essential for adaptation to low oxygen conditions, embryogenesis and tumor progression. HIF-1 is a heterodimer of HIF-1alpha, expression of which is controlled by oxygen levels as well as by various oxygen-independent mechanisms, and HIF-1beta (or ARNT), which is constitutively expressed. In this work, we investigate the phosphorylation of the N-terminal heterodimerization (PAS) domain of HIF-1alpha and identify Ser247 as a major site of in vitro modification by casein kinase 1delta (CK1delta). Mutation of this site to alanine, surprisingly, enhanced the transcriptional activity of HIF-1alpha, a result phenocopied by inhibition or small interfering RNA (siRNA)-mediated silencing of CK1delta under hypoxic conditions. Conversely, overexpression of CK1delta or phosphomimetic mutation of Ser247 to aspartate inhibited HIF-1alpha activity without affecting its stability or nuclear accumulation. Immunoprecipitation and in vitro binding experiments suggest that CK1-dependent phosphorylation of HIF-1alpha at Ser247 impairs its association with ARNT, a notion also supported by modeling the structure of the complex between HIF-1alpha and ARNT PAS-B domains. We suggest that modification of HIF-1alpha by CK1 represents a novel mechanism that controls the activity of HIF-1 during hypoxia by regulating the interaction between its two subunits.


Subject(s)
Casein Kinase Idelta/metabolism , Hypoxia-Inducible Factor 1/metabolism , Amino Acid Sequence , Casein Kinase Idelta/genetics , Cell Hypoxia/physiology , HEK293 Cells , HeLa Cells , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL