hypoxia causes triglyceride accumulation via hif-1-mediated ......2012/03/29  · 107 peroxisome...

9
Journal of Cell Science Hypoxia causes triglyceride accumulation by HIF-1-mediated stimulation of lipin 1 expression Ilias Mylonis 1 , Hiroshi Sembongi 2 , Christina Befani 1 , Panagiotis Liakos 1 , Symeon Siniossoglou 2 and George Simos 1,3, * 1 Laboratory of Biochemistry, Medical School, University of Thessaly, BIOPOLIS, Larissa 41110, Greece 2 Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/Medical Research Council Building, Hills Road, Cambridge, CB2 0XY, UK 3 Institute of Biomedical Research and Technology (BIOMED), 51 Papanastasiou Street, Larissa 41222, Greece *Author for correspondence ([email protected]) Accepted 6 March 2012 Journal of Cell Science 125, 3485–3493 ß 2012. Published by The Company of Biologists Ltd doi: 10.1242/jcs.106682 Summary 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. Key words: Hypoxia, Lipid metabolism, HIF, Lipin, Phosphatidate phosphatase, PAP1 Introduction Exposure of human tissues or cells to reduced oxygen concentration (hypoxia) is encountered both during physiological and pathological processes (Semenza, 2011) and causes dramatic changes in gene expression. Essential to this response are the hypoxia-inducible transcription factors HIF-1 and HIF-2 (EPAS1), recent major anti-cancer targets (Majmundar et al., 2010). Under normal oxygen conditions the regulatory alpha subunit (HIFa) of HIFs is continuously produced and destroyed, in a process involving modification by prolyl hydroxylases (PHDs), VHL- mediated polyubiquitination and subsequent proteasomal degradation (Epstein et al., 2001; Ivan et al., 2001; Kallio et al., 1999; Kamura et al., 2000; Maxwell et al., 1999; Turkish and Sturley, 2007). When oxygen concentration is low, hydroxylation is impaired and HIFa is stabilized, translocates to the nucleus, dimerizes with ARNT (HIFb) to form HIF and binds to hypoxia- responsive elements (HREs) in the promoters/enhancers of its target genes. Tissue-specific and differential control of HIFs also involves several oxygen-independent molecular mechanisms. For HIF-1a these include regulation of its mRNA synthesis by NF-kB (Belaiba et al., 2007; Rius et al., 2008) or the PKR–Stat3 axis (Papadakis et al., 2010), control of its stability by phosphorylation (Flu ¨gel et al., 2007) or protein–protein interactions (Liu et al., 2007), MAPK- dependent regulation of its nuclear transport (Mylonis et al., 2008) and CK1-mediated control of its heterodimerization (Kalousi et al., 2010). One of the major adaptive changes in response to hypoxia is HIF-mediated reprogramming of cellular metabolism. HIF-1 activates genes responsible for switching from oxidative to glycolytic metabolism such as the ones coding for glucose transporters, glycolytic enzymes, lactate dehydrogenase and pyruvate dehydrogenase kinase 1 (Majmundar et al., 2010). Furthermore, hypoxia has been shown to stimulate lipid storage and inhibit lipid catabolism in cultured cardiac myocytes and macrophages (Bostro ¨m et al., 2006; Huss et al., 2001), but the involvement of HIFs in these processes was not investigated. More recent studies using transgenic mice have suggested that artificial or diet induced activation of liver HIF signalling can augment lipid accumulation by modifying hepatocyte lipid metabolism (Kim et al., 2006; Nath et al., 2011; Qu et al., 2011; Rankin et al., 2009). Although these studies have implicated both HIF-1a and HIF-2a in liver fat accumulation and the subsequent maladaptive pathologies, their particular roles have remained largely unclear or controversial and no direct HIF targets have been so far identified among the major lipidogenic enzymes. A key role in lipid biosynthesis is played by lipins, a family of proteins with Mg 2+ -dependent phosphatidate phosphatase (PAP1) activity that catalyze the conversion of phosphatidic acid (PA) to diacylglycerol (DAG) in the penultimate step of triglyceride (TG) synthesis (Donkor et al., 2007; Han et al., 2006). TGs form the core of lipid droplets (LD), the major neutral lipid stores in adipocytes as well as other cell types. Mutations in the lipin 1 gene (Lpin1), which is expressed predominantly in adipose tissue Research Article 3485

Upload: others

Post on 29-Jan-2021

5 views

Category:

Documents


0 download

TRANSCRIPT

  • Journ

    alof

    Cell

    Scie

    nce

    Hypoxia causes triglyceride accumulation byHIF-1-mediated stimulation of lipin 1 expression

    Ilias Mylonis1, Hiroshi Sembongi2, Christina Befani1, Panagiotis Liakos1, Symeon Siniossoglou2 andGeorge Simos1,3,*1Laboratory of Biochemistry, Medical School, University of Thessaly, BIOPOLIS, Larissa 41110, Greece2Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/Medical Research Council Building, Hills Road, Cambridge,CB2 0XY, UK3Institute of Biomedical Research and Technology (BIOMED), 51 Papanastasiou Street, Larissa 41222, Greece

    *Author for correspondence ([email protected])

    Accepted 6 March 2012Journal of Cell Science 125, 3485–3493� 2012. Published by The Company of Biologists Ltddoi: 10.1242/jcs.106682

    SummaryAdaptation to hypoxia involves hypoxia-inducible transcription factors (HIFs) and requires reprogramming of cellular metabolism that isessential 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 byinduction of lipin 1, a phosphatidate phosphatase isoform that catalyzes the penultimate step in triglyceride biosynthesis, whereas lipin 2remains 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 hypoxictriglycerides or lipid droplets can be blocked by siRNA-mediated silencing of lipin 1 expression or kaempferol-mediated inhibition ofHIF-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.

    Key words: Hypoxia, Lipid metabolism, HIF, Lipin, Phosphatidate phosphatase, PAP1

    IntroductionExposure of human tissues or cells to reduced oxygen

    concentration (hypoxia) is encountered both during physiological

    and pathological processes (Semenza, 2011) and causes dramatic

    changes in gene expression. Essential to this response are the

    hypoxia-inducible transcription factors HIF-1 and HIF-2 (EPAS1),

    recent major anti-cancer targets (Majmundar et al., 2010). Under

    normal oxygen conditions the regulatory alpha subunit (HIFa) ofHIFs is continuously produced and destroyed, in a process

    involving modification by prolyl hydroxylases (PHDs), VHL-

    mediated polyubiquitination and subsequent proteasomal

    degradation (Epstein et al., 2001; Ivan et al., 2001; Kallio et al.,

    1999; Kamura et al., 2000; Maxwell et al., 1999; Turkish and

    Sturley, 2007). When oxygen concentration is low, hydroxylation

    is impaired and HIFa is stabilized, translocates to the nucleus,dimerizes with ARNT (HIFb) to form HIF and binds to hypoxia-responsive elements (HREs) in the promoters/enhancers of its

    target genes.

    Tissue-specific and differential control of HIFs also involves

    several oxygen-independent molecular mechanisms. For HIF-1athese include regulation of its mRNA synthesis by NF-kB (Belaibaet al., 2007; Rius et al., 2008) or the PKR–Stat3 axis (Papadakis

    et al., 2010), control of its stability by phosphorylation (Flügel et al.,

    2007) or protein–protein interactions (Liu et al., 2007), MAPK-

    dependent regulation of its nuclear transport (Mylonis et al., 2008)

    and CK1-mediated control of its heterodimerization (Kalousi et al.,

    2010).

    One of the major adaptive changes in response to hypoxia is

    HIF-mediated reprogramming of cellular metabolism. HIF-1

    activates genes responsible for switching from oxidative to

    glycolytic metabolism such as the ones coding for glucose

    transporters, glycolytic enzymes, lactate dehydrogenase and

    pyruvate dehydrogenase kinase 1 (Majmundar et al., 2010).

    Furthermore, hypoxia has been shown to stimulate lipid storage

    and inhibit lipid catabolism in cultured cardiac myocytes and

    macrophages (Boström et al., 2006; Huss et al., 2001), but the

    involvement of HIFs in these processes was not investigated. More

    recent studies using transgenic mice have suggested that artificial

    or diet induced activation of liver HIF signalling can augment lipid

    accumulation by modifying hepatocyte lipid metabolism (Kim

    et al., 2006; Nath et al., 2011; Qu et al., 2011; Rankin et al., 2009).

    Although these studies have implicated both HIF-1a and HIF-2ain liver fat accumulation and the subsequent maladaptive

    pathologies, their particular roles have remained largely unclear

    or controversial and no direct HIF targets have been so far

    identified among the major lipidogenic enzymes.

    A key role in lipid biosynthesis is played by lipins, a family of

    proteins with Mg2+-dependent phosphatidate phosphatase (PAP1)

    activity that catalyze the conversion of phosphatidic acid (PA) to

    diacylglycerol (DAG) in the penultimate step of triglyceride (TG)

    synthesis (Donkor et al., 2007; Han et al., 2006). TGs form the

    core of lipid droplets (LD), the major neutral lipid stores in

    adipocytes as well as other cell types. Mutations in the lipin 1

    gene (Lpin1), which is expressed predominantly in adipose tissue

    Research Article 3485

    mailto:[email protected]

  • Journ

    alof

    Cell

    Scie

    nce

    and skeletal muscle, are the cause of lipodystrophy in the fattyliver dystrophic (fld) mouse and, conversely, overexpression of

    lipin 1 in the adipose tissue of transgenic mice promotes obesity(Péterfy et al., 2001; Phan and Reue, 2005). Moreover, lipin 1 aswell as lipin 2, a liver-enriched isoform, interact with and/or

    modulate the activity of several transcription factors, includingmembers of the peroxisome proliferator-activated receptor(PPAR) family and SREBP that control the expression of genesinvolved in fatty acid and lipid metabolism (Donkor et al., 2009;

    Finck et al., 2006; Koh et al., 2008; Peterson et al., 2011). Apartof its apparently important role in adipogenesis, relatively little isknown about the function of lipin 1 in cells other than adipocytes.

    Various stimuli such as glucocorticoids, insulin or fastingalter lipin 1 expression and/or activity, regulation of which isincompletely understood and involves transcriptional control,

    post-translational modifications and compartmentalization (reviewedby Harris and Finck, 2011).

    We now report that the lipin 1 gene is directly controlled by

    HIF-1 and its up-regulation is linked to TG and LD accumulationin cells of non-adipocyte origin, suggesting that lipin 1 is animportant mediator of the metabolic adaptation to hypoxia at the

    cellular level. Moreover, inhibition of HIF-1 by pharmacologicalor genetic means can suppress the hypoxic induction of lipin 1and TG or LD production.

    ResultsHypoxia induces lipin 1-dependent triglycerideaccumulation

    To investigate the effect of hypoxia on lipid biosynthesis

    we analysed human hepatoblastoma (Huh7) and cervicaladenocarcinoma (HeLaM) cells exposed to normoxia orhypoxia (1% O2). As expected, hypoxia caused a robustexpression of nuclear HIF-1a that could be detected byimmunofluorescence microscopy (Fig. 1A,B, left panels).Furthermore, Nile Red staining revealed that in both cell typeshypoxia caused a significant and time-dependent increase in LD

    accumulation, which was comparable to that caused by oleic acid(OA) treatment used as positive control under normoxia(Fig. 1A,B, middle panels). The induction of neutral lipid

    accumulation by hypoxia was confirmed in Huh7 cells bydetermining the cellular TG content using a biochemical assay(Fig. 1C). Also in this case, TG accumulation under hypoxia wassimilar to the accumulation triggered by oleic acid.

    Analysis of Huh7 cells by western blotting showed that inaddition to HIF-1a, the protein levels of lipin 1, which is consideredto be the minor liver isoform, also increased in response to hypoxia(Fig. 2A). On the contrary, lipin 2, normally the major hepaticisoform, remained unaffected indicating an isoform-specific effectof hypoxia on lipin expression. Similar results were also obtained

    with HeLaM cells treated with hypoxia or dimethyloxalyl glycine(DMOG), a PHD inhibitor that stabilizes HIFa and activates theHIFs (Fig. 2B). Induction of lipin 1 by hypoxia was also confirmed

    using immunofluorescence microscopy in both Huh7 (Fig. 2C) andHeLaM (Fig. 2D) cells. The overexpression of endogenous lipin 1under low oxygen conditions allowed its visualization, which was

    previously not easily attainable in this type of cells. In Huh7 cells,hypoxically induced lipin 1 displayed a predominant cytoplasmic,both diffuse and reticular, localization. Nuclear staining was weak

    but became more evident at later time points (Fig. 2C). Similarimages were obtained in HeLaM cells, in which, however,cytoplasmic spots and nuclei were stained more intensely at early

    and late time points, respectively (Fig. 2D). Therefore, hypoxia

    causes an increase in the cytoplasmic protein levels of lipin 1,

    which, under prolonged treatment, also appears to migrate inside

    the nucleus.

    To test whether the effects of hypoxia on lipin 1 expression and

    lipid droplet accumulation can also be reproduced in normal, non-

    cancer cells we used primary, non-transformed human bronchial

    smooth muscle (hBSM) cells. Analysis of these cells by western

    blotting showed that both HIF-1a and lipin 1 increased in responseto hypoxia, especially after 48 hours of treatment, whereas lipin 2

    expression was not detectable (Fig. 3A). Hypoxic induction was

    confirmed using immunofluorescence microscopy (Fig. 3B), which

    additionally showed that lipin 1 is predominantly cytoplasmic in

    hBSM cells under normoxia but also translocates inside the nucleus

    under hypoxia. Finally, Nile Red staining revealed that 48 hours of

    hypoxic treatment causes significant LD accumulation also in hBSM

    cells (Fig. 3C), suggesting that hypoxia affects lipin 1 expression

    and neutral lipid content in both cancer and non-cancer cells.

    The analysis was continued using Huh7 cells, which, as derived

    by hepatocytes, are more appropriate for studying the relationship

    between lipin 1 induction and lipid accumulation. First, to

    investigate the increase in lipin 1 protein levels, we determined

    Fig. 1. Hypoxia induces neutral lipid accumulation in Huh7 and HeLaM

    cells. (A,B) Fluorescence microscopy analysis. Huh7 (A) or HeLaM (B) cells

    incubated at 1% O2 for 0–48 hours or with 0.4 mM oleic acid (OA) for

    24 hours were processed for immunodetection of HIF-1a, stained with Nile

    Red to visualize lipid droplets and DAPI to show the nuclei and observed by

    fluorescence microscopy. (C) Determination of triglyceride content in Huh7

    cells incubated as in A. Results are mean 6 s.e.m. *P,0.05.

    Journal of Cell Science 125 (14)3486

  • Journ

    alof

    Cell

    Scie

    nce

    its mRNA levels using qPCR. Lipin 1 mRNA significantly

    increased in Huh7 cells subjected to hypoxia for 8 hours, whereas

    lipin 2 mRNA was little affected (Fig. 4A). In the same

    experiment, hypoxia caused robust mRNA expression of VEGF,

    a well-known HIF target. Interestingly, lipin 1 mRNA levels

    decreased after 24 hours of hypoxic treatment (although still

    remaining higher than normoxia) suggesting a transient effect or

    the operation of a negative feedback loop, possibly related to the

    nuclear entry of lipin 1 that was also observed at later time points.

    Since Lpin1 appears to be an early hypoxia-inducible gene, we

    addressed its involvement in hypoxic TG accumulation by siRNA-

    mediated silencing. Knocking down of lipin 1 was effective both

    under normoxia and hypoxia as judged by western blotting

    (Fig. 4B, upper panel). Depletion of lipin 1 under normoxia did not

    significantly affect the TG content of the cells (Fig. 4B, lower

    panel). In contrast, suppression of lipin 1 expression completely

    blocked hypoxia-inducible TG accumulation. To a lesser extent,

    lipin 1 knockdown also diminished TG accumulation triggered by

    OA treatment, in agreement with a general role of lipin 1 in

    response to lipidogenic stimuli in hepatocytes. Therefore, lipin 1 is

    not only transcriptionally controlled by hypoxia but is also

    required for the ensuing overproduction of triglycerides.

    HIF-1 mediates hypoxic up-regulation of lipin 1 and TG

    synthesis

    We then investigated the role of HIFs in the hypoxic induction of

    lipin 1 and TG synthesis. Overexpression of either HIF-1a or

    HIF-2a in Huh7 cells increased the protein levels of lipin 1, withHIF-1a being considerably more effective (Fig. 5A). In contrast,neither HIF-1a nor HIF-2a overexpression affected lipin 2expression. Quantification of the fluorescent signal of Huh7 cells

    overexpressing HIFa and stained with Nile Red showed thatHIFa overexpression also causes LD accumulation (Fig. 5B). Inaccordance to its effect on lipin 1 expression, HIF-1a exerted astronger effect on LD content than HIF-2a. Therefore, HIF-1 and,to lesser extent, HIF-2 activation is sufficient to induce both lipin

    1 and LD production even under normoxic conditions.

    The involvement of HIFs in the hypoxic induction of lipin 1

    was further tested by siRNA-mediated repression of HIF-1a orHIF-2a in Huh7 cells treated with DMOG. Knockdown of HIF-1a greatly reduced lipin 1 expression (Fig. 5C). HIF-2a silencingalso had a negative, but considerably weaker, effect on lipin 1.

    Neither knockdowns affected lipin 2. These data imply that

    hypoxic lipin 1 induction and subsequent TG and LD

    accumulation is predominantly mediated by HIF-1, with HIF-2

    playing a possible minor role. Knocking down HIF-1a using ashRNA plasmid also largely eliminated the capacity of hypoxia

    Fig. 2. Hypoxia induces lipin 1 expression in Huh7 and HeLaM cells.

    (A) Western blotting analysis of Huh7 cells incubated at 1% O2 for 0–

    48 hours. (B) Western blotting analysis of HeLaM cells incubated at 1% O2 or

    with 1 mM DMOG for 0–24 hours. (C) Immunofluorescence microscopy

    analysis. Huh7 cells were incubated at 1% O2 for 0–48 hours, analyzed by

    immunofluorescence microscopy for HIF-1a and lipin 1 and stained with

    DAPI to show the nuclei. (D) Immunofluorescence microscopy analysis.

    HeLaM cells were incubated at 1% O2 for 0–24 hours and analyzed as in C. Fig. 3. Hypoxia induces lipin 1 expression and lipid droplet accumulation

    in hBSM cells. (A) Western blotting analysis of hBSM cells incubated at 1%

    O2 for 0–48 hours. (B) Immunofluorescence microscopy analysis. hBSM

    cells were incubated at 1% O2 for 0–48 hours, analyzed by

    immunofluorescence microscopy for HIF-1a and lipin 1 and stained with

    DAPI to show the nuclei. (C) Fluorescence microscopy analysis. hBSMC

    cells were incubated at 1% O2 for 0–48 hours, processed for immunodetection

    of HIF-1a, stained with Nile Red to visualize lipid droplets and DAPI to show

    the nuclei and observed by fluorescence microscopy.

    HIF-1 regulates lipin 1 3487

  • Journ

    alof

    Cell

    Scie

    nce

    to induce lipin 1 (Fig. 5D) and, moreover, caused a modest but

    significant reduction in hypoxic TG accumulation (Fig. 5E),

    while it did not affect TG accumulation caused by OA treatment.

    Oxygen tension and hydroxylation regulate HIF-1a proteinstability. However, the ability of HIF-1a to enter the nucleus andactivate transcription is additionally controlled by phosphorylation.

    We have previously shown that CK1d targets HIF-1a and inhibitsits activity by impairing its binding to ARNT (Kalousi et al., 2010),

    while phosphorylation by p44/42 MAPK stimulates HIF-1aactivity by blocking its nuclear export (Mylonis et al., 2008). To

    correlate lipin 1 expression to HIF-1a activity rather than just itsprotein levels, we used kinase inhibitors. Treatment of Huh7 cells

    with the CK1d inhibitor IC261, which stimulates HIF-1a activity(Kalousi et al., 2010), potentiated hypoxic induction of lipin 1

    without significantly affecting lipin 2 or HIF-1a protein levels(Fig. 5F). Inversely, treatment of Huh7 cells with the flavonoid

    kaempferol, which suppresses HIF-1a activity by impairing itsnuclear accumulation through MAPK inhibition (Mylonis et al.,

    2010), diminished lipin 1 expression under hypoxia (Fig. 6A,B).

    The same treatment also significantly reduced the amount of LDs in

    Huh7 cells grown under hypoxia (Fig. 6B,C). Therefore, both gain-

    of-function and loss-of-function experiments strongly suggest that

    hypoxic induction of lipin 1 and neutral lipid accumulation involves

    predominantly HIF-1.

    The human Lpin1 promoter contains a conserved

    functional HRE

    A direct role of HIF-1 in Lpin1 regulation requires the

    contribution of an HRE. Examination of the 3.5 kb promoter

    region of Lpin1 revealed the presence of 8 potential HRE sites

    conforming to the consensus XCGTG (X5A or G or C). To test

    Fig. 4. Hypoxic triglyceride accumulation is lipin 1 dependent. (A) Lipin

    1 is transcriptionally upregulated by hypoxia. Determination of lipin 1, lipin 2

    and VEGF mRNA levels in Huh7 cells incubated at 1% O2 for 0–24 hours.

    (B) Lipin 1 is required for TG accumulation under hypoxia. Top panel: Huh7

    cells transfected with lipin 1 or control siRNAs and incubated at 20% or 1%

    O2 for 48 hours were analyzed by western blotting. Bottom panel:

    Determination of triglyceride content in cells treated as above or with 0.4 mM

    oleic acid (OA) for the same period. Values in charts represent the mean 6

    s.e.m. of two independent experiments performed in triplicate (*P,0.05;

    ***P,0.001).

    Fig. 5. HIF-1 mediates hypoxic upregulation of lipin 1 and TG synthesis.

    (A) HIF overexpression induces lipin 1. Western blot analysis of Huh7 cells

    overexpressing GFP, GFP-HIF-1a or GFP-HIF-2a. (B) HIF overexpression

    induces LD accumulation. Huh7 cells overexpressing GFP, GFP-HIF-1a or

    GFP-HIF-2a were stained with Nile Red and their fluorescent signal was

    monitored by microscopy and quantified using ImageJ software. Values

    represent the mean area of Nile Red staining of 50 GFP-negative (2) or GFP-

    positive (+) cells (6 s.e.m.) from two independent experiments (*P,0.05).

    (C) HIFs are required for induction of lipin 1 by DMOG. Western blot

    analysis of Huh7 cells transfected with siRNA against HIF-1a or HIF-2a and

    treated with 1 mM DMOG for 8 hours. (D) HIF-1 is required for hypoxic

    induction of lipin 1. Western blot analysis of Huh7 cells transfected with

    pSUPERshHIF1 or a control plasmid and incubated under 1% or 20% O2 for 8

    hours. (E) HIF-1 is involved in the hypoxic induction of TG accumulation.

    Determination of triglyceride content in Huh7 cells transfected with

    pSUPERshHIF1 or a control plasmid and incubated for 48 hours under 20%

    or 1% O2 or treated with 0.4 mM oleic acid (OA) for 48 hours. Data represent

    the mean (6 s.e.m.) of two independent experiments performed in

    quadruplicate (**P,0.01). (F) Activation of HIF-1 by CK-1d inhibition

    enhances hypoxic induction of lipin 1. Western blot analysis of Huh7 cells

    incubated for 8 hours under 20% or 1% O2 with or without 2 mM IC261.

    Journal of Cell Science 125 (14)3488

  • Journ

    alof

    Cell

    Scie

    nce

    the functionality of these potential HREs, we constructed

    luciferase reporter plasmids containing the whole region

    (3298 bp; HRE1–8) or 59 truncated forms containing 7(HRE2–8), 4 (HRE5–8), 2 (HRE7–8) or 1 (HRE8) proximal

    potential HREs (Fig. 7A, upper panel) and introduced them in

    Huh7 cells. The full-length region (HRE1–8) exhibited the highest

    promoter activity under normoxia and, more importantly, it was

    the only one responding to and activated by hypoxia (Fig. 7A,

    upper panel), suggesting that only the most distal HRE (HRE1) is

    functional. This was also positively shown by using two 39-truncated forms of the Lpin1 promoter region containing only

    HRE1 or four distal HRE sites (HRE1–4): both of these promoter

    fragments could also be activated by hypoxia (Fig. 7A, lower

    panel). HRE1 is conserved (Fig. 7B) as it is perfectly matched by

    a potential distal HRE in the mouse Lpin1 promoter region and is

    found close to the only other known distal regulatory elements,

    the SRE and NF-Y sites (Ishimoto et al., 2009). Finally, mutation

    of HRE1 (Fig. 7B), in the context of both full-length and

    truncated Lpin1 promoter regions (producing mHRE1–8, mHRE1and mHRE1–4), led to complete loss of responsiveness to hypoxia

    (Fig. 7C). Therefore, transcriptional activation of Lpin1 under

    hypoxia is mediated by a single distal HRE site in its promoter.

    HIF-1 directly interacts with the Lpin1 promoter

    Interaction of HIFs with HRE1 in the Lpin1 promoter was tested by

    introducing wild-type or mutant reporter constructs HRE1–8, HRE1and HRE2–8 in Huh7 cells overexpressing HIF-1a or HIF-2a. HIF-1

    Fig. 6. Inhibition of HIF-1 by kaempferol impairs hypoxic induction of

    lipin 1 expression and LD accumulation. (A) Treatment with kaempferol

    inhibits hypoxic induction of lipin 1. Western blot analysis of Huh7 cells

    incubated for 8 hours under 20% or 1% O2 with or without or 10 mMkaempferol (Kae). (B,C) Treatment with kaempferol inhibits hypoxic LD

    accumulation. Huh7 cells incubated for 24 hours under 1% O2 with or without

    50 mM kaempferol (Kae) were processed for immunodetection of HIF-1a,stained with Nile Red and monitored with fluorescence microscopy (B). Nile

    Red staining was quantified using ImageJ software and the results represent

    the mean area of Nile Red staining of 50 cells grown in the absence (2) or

    presence (+) of kaempferol (6 s.e.m.) (*P,0.05).Fig. 7. The human Lpin1 promoter contains a conserved functional distal

    HRE. (A) A single distal HRE is both necessary and sufficient for the

    hypoxic induction of the Lpin1 promoter. Luciferase activity in Huh7 cells

    transfected with pGL3 constructs of the human Lpin1 promoter or its

    fragments and incubated under 20% or 1% O2 for 16 hours. Values are

    expressed in relation to the values obtained for the empty pGL3 vector and

    represent the mean (6 s.e.m.) of three independent experiments performed in

    triplicate. The potential HREs in the promoter region are schematically

    depicted as inverted triangles. (B) The distal HRE in Lpin1 promoter is

    conserved. Top panel: Schematic representation of the human and mouse

    Lpin1 promoter regions (23300 to 21) depicting potential HRE sites

    (inverted triangles), conserved SRE and NF-Y elements (grey polygon and

    circle, respectively) and the distal functional HRE (boxed). Bottom panel:

    Sequence alignment of the region of the human and mouse Lpin1 promoters

    surrounding HRE1. Asterisks indicate conserved nucleotides. The sequence of

    the HRE1 and its mutated form are depicted in boxes. (C) Mutation of the

    distal HRE inhibits hypoxic induction of the Lpin1 promoter. Luciferase

    activity in Huh7 cells transfected with pGL3 constructs containing wild-type

    or mutant fragments of the human Lpin1 promoter and incubated for 16 hours

    under normoxia or hypoxia. Values are expressed as percentage activity in

    relation to normoxia and represent the mean (6 s.e.m.) of three independent

    experiments performed in triplicate. The potential HREs are schematically

    depicted as inverted triangles and a grey X indicates the mutation.

    HIF-1 regulates lipin 1 3489

  • Journ

    alof

    Cell

    Scie

    nce

    could activate transcription from wild-type full-length or single

    HRE1 promoter constructs but not from constructs lacking HRE1(HRE2–8) or containing its mutant form (mHRE1–8 and mHRE1;

    Fig. 8A). HIF-2 also activated the wild-type full-length promoter,

    albeit weaker than HIF-1, but failed to activate transcription of the

    single distal HRE construct (HRE1) suggesting that its role in Lpin1

    activation is minor and involves additional regulatory elements

    aside of HRE1.

    Involvement of endogenous HIF-1 in Lpin1 regulation was

    tested in cells transformed with the full-length Lpin1 promoter

    construct and overexpressing the wild-type or SA mutant form of

    the HIF-1a MTD under hypoxia. Overexpression of wild-typeMTD (MAPK-target domain), a 42 amino acid long fragment of

    HIF-1a containing its MAPK modification sites, specificallyinhibits HIF-1a by blocking its phosphorylation and triggering itsnuclear export, while the mutant SA form of MTD lacks the

    MAPK sites and is inactive (Mylonis et al., 2008). In cells

    expressing wild-type MTD the hypoxic activation of Lpin1

    promoter was virtually abolished but persisted in those cells

    expressing the mutant SA MTD form (Fig. 8B), providing further

    evidence that HIF-1 is the major transcriptional activator of

    Lpin1 gene under hypoxia.

    Direct binding of HIF-1a to HRE1 was tested by chromatinimmunoprecipitation using Huh7 cells cultured under hypoxia or

    normoxia. The Lpin1 promoter region containing HRE1 was

    greatly enriched in anti-HIF-1a immunoprecipitates of DNA-protein complexes isolated from hypoxically treated cells in

    comparison to rabbit IgG immunoprecipitates or anti-HIF-1aimmunoprecipitates from normoxic cells (Fig. 8C,D). Therefore,

    HIF-1a not only activates but also physically associates with theLpin1 promoter.

    DiscussionWe have revealed a direct link between HIF-1a and lipin 1 or thecellular response to hypoxia and the metabolic pathway that leads

    to synthesis of triglycerides and formation of lipid droplets

    (Fig. 8E). Although this connection was identified in cultured

    human cells, it can mechanistically explain previous observations

    made with whole transgenic animals. Over-expression of a

    constitutively active form of HIF-1a in mouse liver led to hepaticfat accumulation and liver steatosis (Kim et al., 2006). In

    contrast, similar over-expression of HIF-2a did not result in liversteatosis but rather caused vascular lesions. Furthermore, lipid

    accumulation in mouse hepatocytes caused by alcohol feeding

    Fig. 8. HIF-1 directly interacts with and activates the Lpin1

    promoter. (A) HIF-1 activates the Lpin1 promoter via the distal

    HRE. Luciferase activity in Huh7 cells overexpressing GFP, GFP-

    HIF-1a or GFP-HIF-2a and transfected with pGL3 constructs

    containing wild-type or mutant fragments of the human Lpin1

    promoter as indicated. Values are expressed as a percentage of

    activity in relation to that of the empty GFP vector and represent

    the mean (6 s.e.m.) of three independent experiments performed

    in triplicate. (B) Inhibition of HIF-1 by MTD overexpression

    inhibits hypoxic induction of the Lpin1 promoter. Luciferase

    activity in Huh7 cells co-transfected with pGL3 constructs of the

    human Lpin1 promoter and plasmids expressing wild-type or

    mutant (SA) MTD and incubated for 16 hours under 20% or 1%

    O2. Values are expressed in relation to the values obtained for the

    empty pGL3 vector and represent the mean (6 s.e.m.) of three

    independent experiments performed in triplicate. (C) HIF-1a

    associates with the distal HRE in the Lpin1 promoter. Gel

    electrophoresis of PCR products amplified from anti-HIF-1a or

    rabbit IgG chromatin immunoprecipitates of Huh7 cells, incubated

    for 8 hours under 20% or 1% O2, using primers specific for the

    distal HRE of the Lpin1 promoter (schematically indicated).

    (D) Quantification of results in C by real-time PCR. The data

    represent the mean (6 s.e.m.) of two independent experiments

    performed in triplicate (***P,0.001). (E) Schematic model

    describing a HIF-1- and lipin-1-dependent mechanism of hypoxic

    induction of TG accumulation.

    Journal of Cell Science 125 (14)3490

  • Journ

    alof

    Cell

    Scie

    nce

    was also shown to involve HIF-1a (Nath et al., 2011). On theother hand, in double knockout studies, hepatic steatosis caused

    by liver-specific deficiency of pVHL could be avoided bysimultaneous inactivation of HIF-2a (Qu et al., 2011; Rankinet al., 2009). However, in these animal models HIF-2 was mainlyassociated with suppression of fatty acid oxidation, increased

    lipid storage capacity, serum TG levels and inflammation. In ourstudy with human cells, HIF-2 also seems to be involved in lipin1 regulation and LD accumulation but its role is minor compared

    to HIF-1 and probably indirect since it requires more complexregulatory elements. It, therefore, appears that HIF-1a and HIF-2a may have overlapping or complementary but distinct roles inthe regulation of hepatic lipid metabolism and the developmentof related pathologies such as alcoholic and non-alcoholic fatty-liver disease (AFLD and NAFLD, respectively), with HIF-1predominantly stimulating lipin 1 and triglyceride synthesis

    under hypoxic or other stress conditions. HIF-1 has also beenimplicated in cardiac steatosis by controlling expression ofPPARc, a principal transcriptional activator of lipid anabolism(Krishnan et al., 2009). Lipin 1 also co-activates PPARc (Kohet al., 2008), so the HIF-1–lipin-1 axis may serve to amplify notonly PAP1 activity but also the non-catalytic transcriptional lipin

    1 functions.

    In order for lipin 1 to exert its transcriptional role, it has totranslocate inside the nucleus. Although lipin 1 is predominantlycytoplasmic in our tested cell lines under normoxia, hypoxic

    treatment increases its nuclear pool especially after long-terminduction. A recent report has shown that nuclear localization oflipin 1 is regulated by nutrient- and growth factor-responsive

    kinase mTOR complex 1 (mTORC1)-mediated phosphorylation(Peterson et al., 2011). Inhibition of mTORC1 causes normoxicredistribution of lipin 1 from the cytoplasm to the nucleus, which

    in turn reduces the nuclear protein levels of SREBP-1 (Petersonet al., 2011), a transcription factor regulating many lipogenicgenes, including lipin 1 (Ishimoto et al., 2009). Therefore, our

    data showing nuclear accumulation of lipin 1 and a decrease oflipin 1 mRNA expression at later points of hypoxic treatmentcould be due to inhibition of mTORC1 that is known to occurafter prolonged exposure to hypoxia (Wouters and Koritzinsky,

    2008). The physiological relevance of lipin 1 hypoxic nuclearmigration could thus be justified by the operation of thefollowing negative feedback mechanism. After the initial HIF-

    1-mediated induction of lipin 1 and the subsequent TGaccumulation, inhibition of mTOR by prolonged hypoxiacauses translocation of lipin 1 inside the nucleus, where, by

    suppressing SREBP-1 function, it reduces expression of its ownand other lipogenic genes and blocks further TG de novosynthesis.

    The idea that cells in a stressful situation, such as oxygen

    deprivation, respond by stimulating, even transiently, an anabolicprocess, e.g. lipid biosynthesis, is counterintuitive but not withoutprecedent. Hypoxia also stimulates glycogen synthesis in a HIF-

    1-dependent way (Brahimi-Horn et al., 2011; Fakas et al., 2011;Pescador et al., 2010). Apparently, exposure of cells to mildhypoxia may serve as a way of sensing ensuing ischemia/nutrient

    limitation and, while it is still available, glucose is stored for lateruse as anaerobic source of energy. However, this may not be thecase for lipids since they can only produce energy through

    oxidative phosphorylation, a process drastically inhibited bysevere hypoxia. A more likely explanation for storingtriglycerides in the form of lipid droplets under hypoxia could

    be that, in this way, cells buffer the toxicity caused by free fatty

    acids, which build up because of suppressed respiratory chain

    activity. Increased PAP1 activity and formation of lipid droplets

    can, thus, be a protective response that prevents intracellular

    lipotoxicity (Fakas et al., 2011; Turkish and Sturley, 2007)

    although stored triglycerides could still serve as a source of

    lipotoxic intermediates if the cells remain for long unable to

    handle them metabolically (Neuschwander-Tetri, 2010).

    Irrespective of its cellular role, the direct involvement of HIF-1

    in triglyceride formation may open new avenues for therapeutic

    interventions in lipid disorders leading to NAFLD and

    steatohepatitis. Furthermore, LD accumulation in non-adipose

    tissue (ectopic fat) is a hallmark of the metabolic syndrome and is

    linked to the pathogenesis of insulin resistance and diabetes

    (Unger et al., 2010). The extensive recent pharmacological

    targeting of HIF-1 as means of anti-cancer or anti-ischemic

    treatment, has led to the identification and availability of many

    agents that inhibit or activate HIF-1, respectively (Semenza,

    2011). Our results with HIF-1 inhibitors, such as kaempferol and

    MTD, provide proof-of-principle that such agents may also be

    useful as effective modulators of intracellular lipid metabolism

    and can form the basis of novel molecular therapies targeting

    lipin 1.

    Materials and MethodsPlasmids

    The human Lpin1 promoter region (nucleotides 23298 to 21; Acc. No.:NT_005334.16) was amplified by PCR using HeLaM total DNA as template,primers hL1prmF1 and hL1prmR1 and KOD Hot start DNA polymerase(TOYOBO Life Science, Tokyo, Japan). The PCR fragment was then introducedinto pUC19 vector, sequenced and subcloned into pGL3-basic vector (Promega,Madison, WI, USA) to construct pGL3-HRE1–8. Truncated promoter forms, pGL3-HRE2–8 (22198), pGL3-HRE5–8 (21297), pGL3-HRE7–8 (2602) and pGL3-HRE8 (2445) were constructed by using as template the pUC19-HRE1–8 plasmid,and an appropriate set of primers listed in supplementary material Table S1. Theoccurring PCR fragments were then subcloned into the pGL3-basic vector andsequenced. To construct HRE mutant plasmid pGL3-mHRE1–8, three nucleotidesubstitutions (CCGTG to CAACG) were introduced by PCR using hL1pHRE1mF1and hL1pHRE1mR1 primers listed in supplementary material Table S1. PlasmidspGL3-HRE1 (23298 to 22588), pGL3-HRE1–4 (23298 to 21585) and theirrespective mutant forms, pGL3-mHRE1 and pGL3-mHRE1–4, were constructed bydigesting pGL3-HRE1–8 with BglII, to yield fragment HRE1, or HindIII, to yieldHRE1–4, and subcloning to the empty pGL3-vector. pEGFP-HIF-1a, pCMV-FLAG-MTD and pCMV-FLAG-MTD-SA plasmids were previously described(Mylonis et al., 2008). To construct pEGFP-HIF-2a, full-length HIF-2a cDNA wasobtained from pcDNA-HIF-2a, kindly provided by S. L. McKnight [University ofTexas Southwestern Medical Center (Tian et al., 1997)].

    Cell culture, transfection and reporter gene assays

    Human HeLaM and Huh7 cells were cultured in DMEM (Biosera) containing10% FCS and 100 U/ml penicillin-streptomycin (Gibco BRL). Human bronchialsmooth muscle cells (Lonza, Switzerland) were cultured in DMEM F-12containing 10% FCS and 100 U/ml penicillin-streptomycin. When required,cells were treated for 4–24 hours with 1 mM dimethyloxalyl glycine (DMOG;Alexis Biochemicals), with CK1 inhibitor IC261 (2 mM; Sigma, St Louis, MO) orwith kaempferol (10–50 mM; Sigma). For oleic acid treatment, the medium wassupplemented with 0.4 mM oleic acid (Sigma) bound to 1.5% bovine serumalbumin. For hypoxic treatment, cells were exposed for 4–48 hours to 1% O2, 95%N2 and 5% CO2 in an IN VIVO2 200 hypoxia workstation (RUSKINN LifeSciences, Pencoed, UK). Transient transfections and reporter gene assays wereperformed as described previously (Mylonis et al., 2008).

    Western blot and immunofluorescence

    Antibodies used for western blotting, immunofluorescence and chromatinimmunoprecipitation were: affinity purified rabbit polyclonal antibodies againstHIF-1a (Lyberopoulou et al., 2007), lipin 1 and lipin 2 (Grimsey et al., 2008),rabbit polyclonal antibodies against GFP (Mylonis et al., 2008) or anti-HIF-2a(Abcam, Cambridge, UK) and mouse monoclonal antibodies against actin ortubulin (Millipore, Billerica, MA, USA). Analysis of total cellular proteins byimmunoblotting and immunofluorescence microscopy were carried out as

    HIF-1 regulates lipin 1 3491

  • Journ

    alof

    Cell

    Scie

    nce

    previously described (Grimsey et al., 2008; Lyberopoulou et al., 2007; Mylonis

    et al., 2008; Mylonis et al., 2006). To visualize lipid droplets after usualimmunofluorescence microscopy procedure cells were stained with Nile Red

    (Sigma; 0.1 mg in PBS) for 15 minutes, washed with PBS and mounted on slides(Greenspan et al., 1985).

    Triglyceride determination

    Huh7 cells were lysed in 150 ml PBS with 0.1% Triton X-100 (Sigma). Themixture was then sonicated (five 10 sec pulses at maximum intensity), boiled for5 minutes to facilitate the formation of an even suspension (Al-Anzi and

    Zinn, 2010) and 20 ml aliquots were tested using a biochemical triglyceridedetermination kit (Biosys, Athens, Greece). Triglyceride values were normalized

    to protein measured with Bio-Rad Protein Assay (Bio-Rad, Hercules, CA).

    siRNA- and shRNA-mediated silencing

    Huh7 cells were incubated in serum-free DMEM for 4 hours with siRNA (10 nM)

    against HIF-1a or HIF-2a (Qiagen, Hilden, Germany) or Lpin1 (target sequencesshown in supplementary material Table S1) in the presence of Lipofectamine

    RNAiMAX (Invitrogen, Carlsbad, CA). AllStars siRNA (Qiagen, Hilden,Germany) was used as negative control. shRNA-mediated silencing of HIF-1awas done using pSuper-shHIF-1a, kindly provided by I. Papandreou and N. C.Denko [Stanford University School of Medicine (Papandreou et al., 2006)] and

    Lipofectamine 2000 (Invitrogen).

    RNA extraction and Real-Time PCR

    Total RNA from Huh7 cells was isolated using the Trizol reagent (Invitrogen) andcDNA was synthesized with the High Capacity Reverse Transcription kit (Applied

    Biosystems, Carlsbad, CA). Real-time PCR was performed with SYBR GreenERqPCR SuperMix Universal (Invitrogen) in a MiniOpticon instrument (Bio-Rad).

    The mRNAs encoding lipin 1, lipin 2 and VEGF were amplified using primerslisted in supplementary material Table S1. Each sample was assayed in triplicate

    for both target and internal control. Relative quantitative gene expression wascalculated using the DDCt method and presented as relative units.

    Chromatin immunoprecipitation

    Huh7 cells were treated with 1.1% formaldehyde for 30 minutes, quenched with125 mM glycine for 5 min and collected by centrifugation. Following steps were

    performed as previously described (Braliou et al., 2008) using a polyclonal anti-HIF-1a antibody (Lyberopoulou et al., 2007) or rabbit IgG. The region 22916 to22686 of the hLPIN1 promoter was amplified from immunoprecipitatedchromatin or input samples using primers listed in supplementary material Table

    S1. The 230 bp product was analyzed by agarose gel electrophoresis or quantifiedby real-time PCR. The Ct (threshold cycle) of immunoprecipitated samples (with

    specific anti-HIF-1a antibody or with non-specific IgG) was normalized to the Ctof total input DNA (DCt). Results obtained with specific anti-HIF-1a antibody wasthen normalized to those obtained with non-specific IgG antibodies (DDCt).

    Densitometric and statistical analysis

    Quantification of the surface covered by Nile Red fluorescence was performed

    with ImageJ public domain software for image analysis and expressed as pixels/cell (Abramoff et al., 2004). Statistical differences between two groups of data

    were assessed using the unpaired t-test in the SigmaPlot v. 9.0 software (Systat);P,0.05 was considered to be significant.

    AcknowledgementsWe are grateful to Drs I. Papandreou, N. C. Denko and S. L.McKnight for generously providing plasmids and to Dr E. Paraskeva,Laboratory of Physiology, University of Thessaly, for providinghBMS cells and instructions for their culture.

    FundingThis study was supported by the National Strategic ReferenceFramework/National Action ‘‘Cooperation’’ [grant number 09SYN-12-682] co-funded by the Greek Government and the EuropeanRegional Development Fund (G.S.); A Medical Research CouncilSenior Fellowship [grant number G0701446 to S.S.]; and a EuropeanMolecular Biology Organization short-term fellowship (I.M.).Deposited in PMC for release after 6 months.

    Supplementary material available online at

    http://jcs.biologists.org/lookup/suppl/doi:10.1242/jcs.106682/-/DC1

    ReferencesAbramoff, M. D., Magelhaes, P. J. and Ram, S. J. (2004). Image Processing with

    ImageJ. Biophot. Intern. 11, 36-42.

    Al-Anzi, B. and Zinn, K. (2010). Colorimetric measurement of triglycerides cannotprovide an accurate measure of stored fat content in Drosophila. PLoS ONE 5,e12353.

    Belaiba, R. S., Bonello, S., Zähringer, C., Schmidt, S., Hess, J., Kietzmann, T. andGörlach, A. (2007). Hypoxia up-regulates hypoxia-inducible factor-1alpha transcriptionby involving phosphatidylinositol 3-kinase and nuclear factor kappaB in pulmonaryartery smooth muscle cells. Mol. Biol. Cell 18, 4691-4697.

    Boström, P., Magnusson, B., Svensson, P. A., Wiklund, O., Borén, J., Carlsson,

    L. M., Ståhlman, M., Olofsson, S. O. and Hultén, L. M. (2006). Hypoxia convertshuman macrophages into triglyceride-loaded foam cells. Arterioscler. Thromb. Vasc.Biol. 26, 1871-1876.

    Brahimi-Horn, M. C., Bellot, G. and Pouysségur, J. (2011). Hypoxia and energetictumour metabolism. Curr. Opin. Genet. Dev. 21, 67-72.

    Braliou, G. G., Verga Falzacappa, M. V., Chachami, G., Casanovas, G.,

    Muckenthaler, M. U. and Simos, G. (2008). 2-Oxoglutarate-dependent oxygenasescontrol hepcidin gene expression. J. Hepatol. 48, 801-810.

    Donkor, J., Sariahmetoglu, M., Dewald, J., Brindley, D. N. and Reue, K. (2007).Three mammalian lipins act as phosphatidate phosphatases with distinct tissueexpression patterns. J. Biol. Chem. 282, 3450-3457.

    Donkor, J., Zhang, P., Wong, S., O’Loughlin, L., Dewald, J., Kok, B. P., Brindley,D. N. and Reue, K. (2009). A conserved serine residue is required for thephosphatidate phosphatase activity but not the transcriptional coactivator functions oflipin-1 and lipin-2. J. Biol. Chem. 284, 29968-29978.

    Epstein, A. C., Gleadle, J. M., McNeill, L. A., Hewitson, K. S., O’Rourke, J., Mole,

    D. R., Mukherji, M., Metzen, E., Wilson, M. I., Dhanda, A. et al. (2001). C.elegans EGL-9 and mammalian homologs define a family of dioxygenases thatregulate HIF by prolyl hydroxylation. Cell 107, 43-54.

    Fakas, S., Qiu, Y., Dixon, J. L., Han, G. S., Ruggles, K. V., Garbarino, J., Sturley,S. L. and Carman, G. M. (2011). Phosphatidate phosphatase activity plays key rolein protection against fatty acid-induced toxicity in yeast. J. Biol. Chem. 286, 29074-29085.

    Finck, B. N., Gropler, M. C., Chen, Z., Leone, T. C., Croce, M. A., Harris, T. E.,

    Lawrence, J. C., Jr and Kelly, D. P. (2006). Lipin 1 is an inducible amplifier of thehepatic PGC-1alpha/PPARalpha regulatory pathway. Cell Metab. 4, 199-210.

    Flügel, D., Görlach, A., Michiels, C. and Kietzmann, T. (2007). Glycogen synthasekinase 3 phosphorylates hypoxia-inducible factor 1alpha and mediates its destabilizationin a VHL-independent manner. Mol. Cell. Biol. 27, 3253-3265.

    Greenspan, P., Mayer, E. P. and Fowler, S. D. (1985). Nile red: a selective fluorescentstain for intracellular lipid droplets. J. Cell Biol. 100, 965-973.

    Grimsey, N., Han, G. S., O’Hara, L., Rochford, J. J., Carman, G. M. and

    Siniossoglou, S. (2008). Temporal and spatial regulation of the phosphatidatephosphatases lipin 1 and 2. J. Biol. Chem. 283, 29166-29174.

    Han, G. S., Wu, W. I. and Carman, G. M. (2006). The Saccharomyces cerevisiaeLipin homolog is a Mg2+-dependent phosphatidate phosphatase enzyme. J. Biol.Chem. 281, 9210-9218.

    Harris, T. E. and Finck, B. N. (2011). Dual function lipin proteins and glycerolipidmetabolism. Trends Endocrinol. Metab. 22, 226-233.

    Huss, J. M., Levy, F. H. and Kelly, D. P. (2001). Hypoxia inhibits the peroxisomeproliferator-activated receptor alpha/retinoid X receptor gene regulatory pathway incardiac myocytes: a mechanism for O2-dependent modulation of mitochondrial fattyacid oxidation. J. Biol. Chem. 276, 27605-27612.

    Ishimoto, K., Nakamura, H., Tachibana, K., Yamasaki, D., Ota, A., Hirano, K.,Tanaka, T., Hamakubo, T., Sakai, J., Kodama, T. et al. (2009). Sterol-mediatedregulation of human lipin 1 gene expression in hepatoblastoma cells. J. Biol. Chem.284, 22195-22205.

    Ivan, M., Kondo, K., Yang, H., Kim, W., Valiando, J., Ohh, M., Salic, A., Asara, J. M.,

    Lane, W. S. and Kaelin, W. G., Jr (2001). HIFalpha targeted for VHL-mediateddestruction by proline hydroxylation: implications for O2 sensing. Science 292, 464-468.

    Kallio, P. J., Wilson, W. J., O’Brien, S., Makino, Y. and Poellinger, L. (1999).Regulation of the hypoxia-inducible transcription factor 1alpha by the ubiquitin-proteasome pathway. J. Biol. Chem. 274, 6519-6525.

    Kalousi, A., Mylonis, I., Politou, A. S., Chachami, G., Paraskeva, E. and Simos, G.

    (2010). Casein kinase 1 regulates human hypoxia-inducible factor HIF-1. J. Cell Sci.123, 2976-2986.

    Kamura, T., Sato, S., Iwai, K., Czyzyk-Krzeska, M., Conaway, R. C. and Conaway,

    J. W. (2000). Activation of HIF1alpha ubiquitination by a reconstituted von Hippel-Lindau (VHL) tumor suppressor complex. Proc. Natl. Acad. Sci. USA 97, 10430-10435.

    Kim, W. Y., Safran, M., Buckley, M. R., Ebert, B. L., Glickman, J., Bosenberg, M.,

    Regan, M. and Kaelin, W. G., Jr (2006). Failure to prolyl hydroxylate hypoxia-inducible factor alpha phenocopies VHL inactivation in vivo. EMBO J. 25, 4650-4662.

    Koh, Y. K., Lee, M. Y., Kim, J. W., Kim, M., Moon, J. S., Lee, Y. J., Ahn, Y. H. and

    Kim, K. S. (2008). Lipin1 is a key factor for the maturation and maintenance ofadipocytes in the regulatory network with CCAAT/enhancer-binding protein alpha andperoxisome proliferator-activated receptor gamma 2. J. Biol. Chem. 283, 34896-34906.

    Krishnan, J., Suter, M., Windak, R., Krebs, T., Felley, A., Montessuit, C.,Tokarska-Schlattner, M., Aasum, E., Bogdanova, A., Perriard, E. et al. (2009).Activation of a HIF1alpha-PPARgamma axis underlies the integration of glycolyticand lipid anabolic pathways in pathologic cardiac hypertrophy. Cell Metab. 9, 512-524.

    Journal of Cell Science 125 (14)3492

    http://jcs.biologists.org/lookup/suppl/doi:10.1242/jcs.106682/-/DC1http://dx.doi.org/10.1371%2Fjournal.pone.0012353http://dx.doi.org/10.1371%2Fjournal.pone.0012353http://dx.doi.org/10.1371%2Fjournal.pone.0012353http://dx.doi.org/10.1091%2Fmbc.E07-04-0391http://dx.doi.org/10.1091%2Fmbc.E07-04-0391http://dx.doi.org/10.1091%2Fmbc.E07-04-0391http://dx.doi.org/10.1091%2Fmbc.E07-04-0391http://dx.doi.org/10.1161%2F01.ATV.0000229665.78997.0bhttp://dx.doi.org/10.1161%2F01.ATV.0000229665.78997.0bhttp://dx.doi.org/10.1161%2F01.ATV.0000229665.78997.0bhttp://dx.doi.org/10.1161%2F01.ATV.0000229665.78997.0bhttp://dx.doi.org/10.1016%2Fj.gde.2010.10.006http://dx.doi.org/10.1016%2Fj.gde.2010.10.006http://dx.doi.org/10.1016%2Fj.jhep.2007.12.021http://dx.doi.org/10.1016%2Fj.jhep.2007.12.021http://dx.doi.org/10.1016%2Fj.jhep.2007.12.021http://dx.doi.org/10.1074%2Fjbc.M610745200http://dx.doi.org/10.1074%2Fjbc.M610745200http://dx.doi.org/10.1074%2Fjbc.M610745200http://dx.doi.org/10.1074%2Fjbc.M109.023663http://dx.doi.org/10.1074%2Fjbc.M109.023663http://dx.doi.org/10.1074%2Fjbc.M109.023663http://dx.doi.org/10.1074%2Fjbc.M109.023663http://dx.doi.org/10.1016%2FS0092-8674%2801%2900507-4http://dx.doi.org/10.1016%2FS0092-8674%2801%2900507-4http://dx.doi.org/10.1016%2FS0092-8674%2801%2900507-4http://dx.doi.org/10.1016%2FS0092-8674%2801%2900507-4http://dx.doi.org/10.1074%2Fjbc.M111.258798http://dx.doi.org/10.1074%2Fjbc.M111.258798http://dx.doi.org/10.1074%2Fjbc.M111.258798http://dx.doi.org/10.1074%2Fjbc.M111.258798http://dx.doi.org/10.1016%2Fj.cmet.2006.08.005http://dx.doi.org/10.1016%2Fj.cmet.2006.08.005http://dx.doi.org/10.1016%2Fj.cmet.2006.08.005http://dx.doi.org/10.1128%2FMCB.00015-07http://dx.doi.org/10.1128%2FMCB.00015-07http://dx.doi.org/10.1128%2FMCB.00015-07http://dx.doi.org/10.1083%2Fjcb.100.3.965http://dx.doi.org/10.1083%2Fjcb.100.3.965http://dx.doi.org/10.1074%2Fjbc.M804278200http://dx.doi.org/10.1074%2Fjbc.M804278200http://dx.doi.org/10.1074%2Fjbc.M804278200http://dx.doi.org/10.1074%2Fjbc.M600425200http://dx.doi.org/10.1074%2Fjbc.M600425200http://dx.doi.org/10.1074%2Fjbc.M600425200http://dx.doi.org/10.1016%2Fj.tem.2011.02.006http://dx.doi.org/10.1016%2Fj.tem.2011.02.006http://dx.doi.org/10.1074%2Fjbc.M100277200http://dx.doi.org/10.1074%2Fjbc.M100277200http://dx.doi.org/10.1074%2Fjbc.M100277200http://dx.doi.org/10.1074%2Fjbc.M100277200http://dx.doi.org/10.1074%2Fjbc.M109.028753http://dx.doi.org/10.1074%2Fjbc.M109.028753http://dx.doi.org/10.1074%2Fjbc.M109.028753http://dx.doi.org/10.1074%2Fjbc.M109.028753http://dx.doi.org/10.1126%2Fscience.1059817http://dx.doi.org/10.1126%2Fscience.1059817http://dx.doi.org/10.1126%2Fscience.1059817http://dx.doi.org/10.1074%2Fjbc.274.10.6519http://dx.doi.org/10.1074%2Fjbc.274.10.6519http://dx.doi.org/10.1074%2Fjbc.274.10.6519http://dx.doi.org/10.1242%2Fjcs.068122http://dx.doi.org/10.1242%2Fjcs.068122http://dx.doi.org/10.1242%2Fjcs.068122http://dx.doi.org/10.1073%2Fpnas.190332597http://dx.doi.org/10.1073%2Fpnas.190332597http://dx.doi.org/10.1073%2Fpnas.190332597http://dx.doi.org/10.1073%2Fpnas.190332597http://dx.doi.org/10.1038%2Fsj.emboj.7601300http://dx.doi.org/10.1038%2Fsj.emboj.7601300http://dx.doi.org/10.1038%2Fsj.emboj.7601300http://dx.doi.org/10.1074%2Fjbc.M804007200http://dx.doi.org/10.1074%2Fjbc.M804007200http://dx.doi.org/10.1074%2Fjbc.M804007200http://dx.doi.org/10.1074%2Fjbc.M804007200http://dx.doi.org/10.1016%2Fj.cmet.2009.05.005http://dx.doi.org/10.1016%2Fj.cmet.2009.05.005http://dx.doi.org/10.1016%2Fj.cmet.2009.05.005http://dx.doi.org/10.1016%2Fj.cmet.2009.05.005http://dx.doi.org/10.1016%2Fj.cmet.2009.05.005

  • Journ

    alof

    Cell

    Scie

    nce

    Liu, Y. V., Baek, J. H., Zhang, H., Diez, R., Cole, R. N. and Semenza, G. L. (2007).RACK1 competes with HSP90 for binding to HIF-1alpha and is required for O(2)-independent and HSP90 inhibitor-induced degradation of HIF-1alpha. Mol. Cell 25,207-217.

    Lyberopoulou, A., Venieris, E., Mylonis, I., Chachami, G., Pappas, I., Simos, G.,

    Bonanou, S. and Georgatsou, E. (2007). MgcRacGAP interacts with HIF-1alpha andregulates its transcriptional activity. Cell. Physiol. Biochem. 20, 995-1006.

    Majmundar, A. J., Wong, W. J. and Simon, M. C. (2010). Hypoxia-inducible factorsand the response to hypoxic stress. Mol. Cell 40, 294-309.

    Maxwell, P. H., Wiesener, M. S., Chang, G. W., Clifford, S. C., Vaux, E. C.,

    Cockman, M. E., Wykoff, C. C., Pugh, C. W., Maher, E. R. and Ratcliffe, P. J.(1999). The tumour suppressor protein VHL targets hypoxia-inducible factors foroxygen-dependent proteolysis. Nature 399, 271-275.

    Mylonis, I., Chachami, G., Samiotaki, M., Panayotou, G., Paraskeva, E., Kalousi,

    A., Georgatsou, E., Bonanou, S. and Simos, G. (2006). Identification of MAPKphosphorylation sites and their role in the localization and activity of hypoxia-inducible factor-1alpha. J. Biol. Chem. 281, 33095-33106.

    Mylonis, I., Chachami, G., Paraskeva, E. and Simos, G. (2008). Atypical CRM1-dependent nuclear export signal mediates regulation of hypoxia-inducible factor-1alpha by MAPK. J. Biol. Chem. 283, 27620-27627.

    Mylonis, I., Lakka, A., Tsakalof, A. and Simos, G. (2010). The dietary flavonoidkaempferol effectively inhibits HIF-1 activity and hepatoma cancer cell viabilityunder hypoxic conditions. Biochem. Biophys. Res. Commun. 398, 74-78.

    Nath, B., Levin, I., Csak, T., Petrasek, J., Mueller, C., Kodys, K., Catalano, D.,

    Mandrekar, P. and Szabo, G. (2011). Hepatocyte-specific hypoxia-inducible factor-1a is a determinant of lipid accumulation and liver injury in alcohol-induced steatosisin mice. Hepatology 53, 1526-1537.

    Neuschwander-Tetri, B. A. (2010). Hepatic lipotoxicity and the pathogenesis ofnonalcoholic steatohepatitis: the central role of nontriglyceride fatty acid metabolites.Hepatology 52, 774-788.

    Papadakis, A. I., Paraskeva, E., Peidis, P., Muaddi, H., Li, S., Raptis, L.,

    Pantopoulos, K., Simos, G. and Koromilas, A. E. (2010). eIF2alpha Kinase PKRmodulates the hypoxic response by Stat3-dependent transcriptional suppression ofHIF-1alpha. Cancer Res. 70, 7820-7829.

    Papandreou, I., Cairns, R. A., Fontana, L., Lim, A. L. and Denko, N. C. (2006). HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygenconsumption. Cell Metab. 3, 187-197.

    Pescador, N., Villar, D., Cifuentes, D., Garcia-Rocha, M., Ortiz-Barahona, A.,

    Vazquez, S., Ordoñez, A., Cuevas, Y., Saez-Morales, D., Garcia-Bermejo, M. L.

    et al. (2010). Hypoxia promotes glycogen accumulation through hypoxia inducible

    factor (HIF)-mediated induction of glycogen synthase 1. PLoS ONE 5, e9644.

    Péterfy, M., Phan, J., Xu, P. and Reue, K. (2001). Lipodystrophy in the fld mouse

    results from mutation of a new gene encoding a nuclear protein, lipin. Nat. Genet. 27,

    121-124.

    Peterson, T. R., Sengupta, S. S., Harris, T. E., Carmack, A. E., Kang, S. A.,

    Balderas, E., Guertin, D. A., Madden, K. L., Carpenter, A. E., Finck, B. N. et al.

    (2011). mTOR complex 1 regulates lipin 1 localization to control the SREBP

    pathway. Cell 146, 408-420.

    Phan, J. and Reue, K. (2005). Lipin, a lipodystrophy and obesity gene. Cell Metab. 1,

    73-83.

    Qu, A., Taylor, M., Xue, X., Matsubara, T., Metzger, D., Chambon, P., Gonzalez,

    F. J. and Shah, Y. M. (2011). Hypoxia-inducible transcription factor 2a promotessteatohepatitis through augmenting lipid accumulation, inflammation, and fibrosis.

    Hepatology 54, 472-483.

    Rankin, E. B., Rha, J., Selak, M. A., Unger, T. L., Keith, B., Liu, Q. and Haase,

    V. H. (2009). Hypoxia-inducible factor 2 regulates hepatic lipid metabolism. Mol.

    Cell. Biol. 29, 4527-4538.

    Rius, J., Guma, M., Schachtrup, C., Akassoglou, K., Zinkernagel, A. S., Nizet, V.,

    Johnson, R. S., Haddad, G. G. and Karin, M. (2008). NF-kappaB links innate

    immunity to the hypoxic response through transcriptional regulation of HIF-1alpha.

    Nature 453, 807-811.

    Semenza, G. L. (2011). Oxygen sensing, homeostasis, and disease. N. Engl. J. Med. 365,

    537-547.

    Tian, H., McKnight, S. L. and Russell, D. W. (1997). Endothelial PAS domain protein

    1 (EPAS1), a transcription factor selectively expressed in endothelial cells. Genes

    Dev. 11, 72-82.

    Turkish, A. and Sturley, S. L. (2007). Regulation of triglyceride metabolism. I.

    Eukaryotic neutral lipid synthesis: ‘‘Many ways to skin ACAT or a DGAT’’. Am. J.

    Physiol. Gastrointest. Liver Physiol. 292, G953-G957.

    Unger, R. H., Clark, G. O., Scherer, P. E. and Orci, L. (2010). Lipid homeostasis,

    lipotoxicity and the metabolic syndrome. Biochim. Biophys. Acta 1801, 209-214.

    Wouters, B. G. and Koritzinsky, M. (2008). Hypoxia signalling through mTOR and

    the unfolded protein response in cancer. Nat. Rev. Cancer 8, 851-864.

    HIF-1 regulates lipin 1 3493

    http://dx.doi.org/10.1016%2Fj.molcel.2007.01.001http://dx.doi.org/10.1016%2Fj.molcel.2007.01.001http://dx.doi.org/10.1016%2Fj.molcel.2007.01.001http://dx.doi.org/10.1016%2Fj.molcel.2007.01.001http://dx.doi.org/10.1159%2F000110460http://dx.doi.org/10.1159%2F000110460http://dx.doi.org/10.1159%2F000110460http://dx.doi.org/10.1016%2Fj.molcel.2010.09.022http://dx.doi.org/10.1016%2Fj.molcel.2010.09.022http://dx.doi.org/10.1038%2F20459http://dx.doi.org/10.1038%2F20459http://dx.doi.org/10.1038%2F20459http://dx.doi.org/10.1038%2F20459http://dx.doi.org/10.1074%2Fjbc.M605058200http://dx.doi.org/10.1074%2Fjbc.M605058200http://dx.doi.org/10.1074%2Fjbc.M605058200http://dx.doi.org/10.1074%2Fjbc.M605058200http://dx.doi.org/10.1074%2Fjbc.M803081200http://dx.doi.org/10.1074%2Fjbc.M803081200http://dx.doi.org/10.1074%2Fjbc.M803081200http://dx.doi.org/10.1016%2Fj.bbrc.2010.06.038http://dx.doi.org/10.1016%2Fj.bbrc.2010.06.038http://dx.doi.org/10.1016%2Fj.bbrc.2010.06.038http://dx.doi.org/10.1002%2Fhep.24256http://dx.doi.org/10.1002%2Fhep.24256http://dx.doi.org/10.1002%2Fhep.24256http://dx.doi.org/10.1002%2Fhep.24256http://dx.doi.org/10.1002%2Fhep.23719http://dx.doi.org/10.1002%2Fhep.23719http://dx.doi.org/10.1002%2Fhep.23719http://dx.doi.org/10.1158%2F0008-5472.CAN-10-0215http://dx.doi.org/10.1158%2F0008-5472.CAN-10-0215http://dx.doi.org/10.1158%2F0008-5472.CAN-10-0215http://dx.doi.org/10.1158%2F0008-5472.CAN-10-0215http://dx.doi.org/10.1016%2Fj.cmet.2006.01.012http://dx.doi.org/10.1016%2Fj.cmet.2006.01.012http://dx.doi.org/10.1016%2Fj.cmet.2006.01.012http://dx.doi.org/10.1371%2Fjournal.pone.0009644http://dx.doi.org/10.1371%2Fjournal.pone.0009644http://dx.doi.org/10.1371%2Fjournal.pone.0009644http://dx.doi.org/10.1371%2Fjournal.pone.0009644http://dx.doi.org/10.1038%2F83685http://dx.doi.org/10.1038%2F83685http://dx.doi.org/10.1038%2F83685http://dx.doi.org/10.1016%2Fj.cell.2011.06.034http://dx.doi.org/10.1016%2Fj.cell.2011.06.034http://dx.doi.org/10.1016%2Fj.cell.2011.06.034http://dx.doi.org/10.1016%2Fj.cell.2011.06.034http://dx.doi.org/10.1016%2Fj.cmet.2004.12.002http://dx.doi.org/10.1016%2Fj.cmet.2004.12.002http://dx.doi.org/10.1002%2Fhep.24400http://dx.doi.org/10.1002%2Fhep.24400http://dx.doi.org/10.1002%2Fhep.24400http://dx.doi.org/10.1002%2Fhep.24400http://dx.doi.org/10.1128%2FMCB.00200-09http://dx.doi.org/10.1128%2FMCB.00200-09http://dx.doi.org/10.1128%2FMCB.00200-09http://dx.doi.org/10.1038%2Fnature06905http://dx.doi.org/10.1038%2Fnature06905http://dx.doi.org/10.1038%2Fnature06905http://dx.doi.org/10.1038%2Fnature06905http://dx.doi.org/10.1056%2FNEJMra1011165http://dx.doi.org/10.1056%2FNEJMra1011165http://dx.doi.org/10.1101%2Fgad.11.1.72http://dx.doi.org/10.1101%2Fgad.11.1.72http://dx.doi.org/10.1101%2Fgad.11.1.72http://dx.doi.org/10.1152%2Fajpgi.00509.2006http://dx.doi.org/10.1152%2Fajpgi.00509.2006http://dx.doi.org/10.1152%2Fajpgi.00509.2006http://dx.doi.org/10.1016%2Fj.bbalip.2009.10.006http://dx.doi.org/10.1016%2Fj.bbalip.2009.10.006http://dx.doi.org/10.1038%2Fnrc2501http://dx.doi.org/10.1038%2Fnrc2501

    Fig 1Fig 2Fig 3Fig 4Fig 5Fig 6Fig 7Fig 8Ref 1Ref 2Ref 3Ref 4Ref 5Ref 6Ref 7Ref 8Ref 9Ref 10Ref 11Ref 12Ref 13Ref 14Ref 15Ref 16Ref 17Ref 18Ref 19Ref 20Ref 21Ref 22Ref 23Ref 24Ref 25Ref 26Ref 27Ref 28Ref 29Ref 30Ref 31Ref 32Ref 33Ref 34Ref 35Ref 36Ref 37Ref 38Ref 39Ref 40Ref 41Ref 42Ref 43Ref 44Ref 45Ref 46Ref 47Ref 48

    /ColorImageDict > /JPEG2000ColorACSImageDict > /JPEG2000ColorImageDict > /AntiAliasGrayImages false /CropGrayImages true /GrayImageMinResolution 150 /GrayImageMinResolutionPolicy /OK /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 200 /GrayImageDepth 8 /GrayImageMinDownsampleDepth 2 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /FlateEncode /AutoFilterGrayImages false /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict > /GrayImageDict > /JPEG2000GrayACSImageDict > /JPEG2000GrayImageDict > /AntiAliasMonoImages false /CropMonoImages true /MonoImageMinResolution 1200 /MonoImageMinResolutionPolicy /OK /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 600 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict > /AllowPSXObjects false /CheckCompliance [ /None ] /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly true /PDFXNoTrimBoxError false /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox false /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile (Euroscale Coated v2) /PDFXOutputConditionIdentifier (FOGRA1) /PDFXOutputCondition () /PDFXRegistryName (http://www.color.org) /PDFXTrapped /False

    /CreateJDFFile false /SyntheticBoldness 1.000000 /Description >>> setdistillerparams> setpagedevice