Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

16
of March 13, 2018. This information is current as as a New Hypoxia-Inducible Gene Identification Of CC-Chemokine Ligand 20 Novel Immune-Related Genes and Primary Human Monocytes: Modulation of Hypoxia Modifies the Transcriptome of Luigi Varesio and Anfosso, Ulrich Pfeffer, Paolo Fardin, Florinda Battaglia Maria Carla Bosco, Maura Puppo, Clara Santangelo, Luca http://www.jimmunol.org/content/177/3/1941 doi: 10.4049/jimmunol.177.3.1941 2006; 177:1941-1955; ; J Immunol References http://www.jimmunol.org/content/177/3/1941.full#ref-list-1 , 33 of which you can access for free at: cites 81 articles This article average * 4 weeks from acceptance to publication Fast Publication! Every submission reviewed by practicing scientists No Triage! from submission to initial decision Rapid Reviews! 30 days* Submit online. ? The JI Why Subscription http://jimmunol.org/subscription is online at: The Journal of Immunology Information about subscribing to Permissions http://www.aai.org/About/Publications/JI/copyright.html Submit copyright permission requests at: Email Alerts http://jimmunol.org/alerts Receive free email-alerts when new articles cite this article. Sign up at: Print ISSN: 0022-1767 Online ISSN: 1550-6606. Immunologists All rights reserved. Copyright © 2006 by The American Association of 1451 Rockville Pike, Suite 650, Rockville, MD 20852 The American Association of Immunologists, Inc., is published twice each month by The Journal of Immunology by guest on March 13, 2018 http://www.jimmunol.org/ Downloaded from by guest on March 13, 2018 http://www.jimmunol.org/ Downloaded from

Transcript of Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

Page 1: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

of March 13, 2018.This information is current as as a New Hypoxia-Inducible Gene

Identification Of CC-Chemokine Ligand 20 Novel Immune-Related Genes andPrimary Human Monocytes: Modulation of Hypoxia Modifies the Transcriptome of

Luigi VaresioandAnfosso, Ulrich Pfeffer, Paolo Fardin, Florinda Battaglia

Maria Carla Bosco, Maura Puppo, Clara Santangelo, Luca

http://www.jimmunol.org/content/177/3/1941doi: 10.4049/jimmunol.177.3.1941

2006; 177:1941-1955; ;J Immunol 

Referenceshttp://www.jimmunol.org/content/177/3/1941.full#ref-list-1

, 33 of which you can access for free at: cites 81 articlesThis article

        average*  

4 weeks from acceptance to publicationFast Publication! •    

Every submission reviewed by practicing scientistsNo Triage! •    

from submission to initial decisionRapid Reviews! 30 days* •    

Submit online. ?The JIWhy

Subscriptionhttp://jimmunol.org/subscription

is online at: The Journal of ImmunologyInformation about subscribing to

Permissionshttp://www.aai.org/About/Publications/JI/copyright.htmlSubmit copyright permission requests at:

Email Alertshttp://jimmunol.org/alertsReceive free email-alerts when new articles cite this article. Sign up at:

Print ISSN: 0022-1767 Online ISSN: 1550-6606. Immunologists All rights reserved.Copyright © 2006 by The American Association of1451 Rockville Pike, Suite 650, Rockville, MD 20852The American Association of Immunologists, Inc.,

is published twice each month byThe Journal of Immunology

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 2: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

Hypoxia Modifies the Transcriptome of Primary HumanMonocytes: Modulation of Novel Immune-Related Genes andIdentification Of CC-Chemokine Ligand 20 as a NewHypoxia-Inducible Gene1

Maria Carla Bosco,2* Maura Puppo,* Clara Santangelo,* Luca Anfosso,‡ Ulrich Pfeffer,†

Paolo Fardin,* Florinda Battaglia,* and Luigi Varesio*

Peripheral blood monocytes migrate to and accumulate in hypoxic areas of inflammatory and tumor lesions. To characterize themolecular bases underlying monocyte functions within a hypoxic microenvironment, we investigated the transcriptional profileinduced by hypoxia in primary human monocytes using high-density oligonucleotide microarrays. Profound changes in the geneexpression pattern were detected following 16 h exposure to 1% O2, with 536 and 677 sequences showing at least a 1.5-fold increaseand decrease, respectively. Validation of this analysis was provided by quantitative RT-PCR confirmation of expression differencesof selected genes. Among modulated genes, 74 were known hypoxia-responsive genes, whereas the majority were new genes whoseresponsiveness to hypoxia had not been previously described. The hypoxic transcriptome was characterized by the modulation ofa significant cluster of genes with immunological relevance. These included scavenger receptors (CD163, STAB1, C1qR1, MSR1,MARCO, TLR7), immunoregulatory, costimulatory, and adhesion molecules (CD32, CD64, CD69, CD89, CMRF-35H, ITGB5,LAIR1, LIR9), chemokines/cytokines and receptors (CCL23, CCL15, CCL8, CCR1, CCR2, RDC1, IL-23A, IL-6ST). Furthermore,we provided conclusive evidence of hypoxic induction of CCL20, a chemoattractant for immature dendritic cells, activated/memoryT lymphocytes, and naive B cells. CCL20 mRNA up-regulation was paralleled by increased protein expression and secretion. Thisstudy represents the first transcriptome analysis of hypoxic primary human monocytes, which provides novel insights into mono-cyte functional behavior within ischemic/hypoxic tissues. CCL20 up-regulation by hypoxia may constitute an important mecha-nism to promote recruitment of specific leukocyte subsets at pathological sites and may have implications for the pathogenesis ofchronic inflammatory diseases. The Journal of Immunology, 2006, 177: 1941–1955.

P eripheral blood monocytes (Mn)3 represent the earlymononuclear phagocyte component of the leukocyte infil-trate at sites of inflammation, infection, and tumor growth,

where they differentiate into inflammatory and tumor-associatedmacrophages (Mf) (1). Mn/Mf are potent regulators of immuneand inflammatory reactions. They orchestrate the coordinated re-cruitment and activation of specific leukocyte subsets at patholog-ical sites through the local secretion of low m.w. structurally re-lated proteins, termed chemokines (1). Chemokines are classifiedinto CXC, CC, C, and CX3C families, which bind to and activatemembers of a superfamily of 7-transmembrane domain, G protein-

coupled receptors differentially expressed and regulated in leuko-cytes (2). CCL20 (also known as MIP-3�, liver and activationregulated chemokine, and Exodus) is a recently described Mn-derived CC-chemokine which selectively attracts immature den-dritic cells (iDC), effector/memory T lymphocytes, and naive Bcells through its specific receptor, CCR6, expressed on these cells(for a review, see Ref. 3).

Mononuclear phagocyte reactivity in pathological tissues isfinely tuned by a complex interplay between stimulatory and in-hibitory signals of various nature that include immune-derivedstimuli (4, 5), viral/bacterial products (5, 6), cell metabolites (4, 7),and tissue-specific signals (8). A common denominator of manypathological processes and an important regulator of gene expres-sion is represented by low partial oxygen pressure (pO2) (reviewedin Ref. 9). Hypoxia occurs in cardiovascular, hematological, andpulmonary disorders, ischemic wounds, arthritic joints, atheroscle-rotic plaques, and microbial infections, and experimental and clin-ical studies point toward its fundamental role in the pathogenesisof these diseases (8–11). Areas of low pO2 are also present in solidtumors, where they have been associated with malignant progres-sion, metastasis formation, resistance to therapy, and poor clinicaloutcome (8, 12–14). Transcriptional response to hypoxia is medi-ated primarily by the hypoxia-inducible factor-1 (HIF-1), a het-erodimeric basic helix-loop-helix (bHLH) transcription factorcomposed of HIF-1� (also known as the aryl hydrocarbon receptornuclear translocator (ARNT)), the constitutive subunit, and HIF-1�, 2�, or 3�, the oxygen-sensitive subunits (9, 15). The � sub-units are posttranslationally stabilized under hypoxia and translo-cate to the nucleus where they dimerize with HIF-1�,

*Laboratory of Molecular Biology, G. Gaslini Institute, and †Functional Genomics,National Cancer Research Institute, Genova, Italy; and ‡University of Insubria, Va-rese, Italy

Received for publication November 4, 2005. Accepted for publication May 18, 2006.

The costs of publication of this article were defrayed in part by the payment of pagecharges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.1 This work was supported by grants from the Italian Association for Cancer Re-search, Fondazione Italiana per la Lotta al Neuroblastoma, San Paolo Company, Ital-ian Health Ministry, and Ministero Istruzione Universita’ e Ricerca.2 Address correspondence and reprint requests to Dr. Maria Carla Bosco, Laboratoriodi Biologia Molecolare, Istituto Giannina Gaslini, Padiglione 2, L.go GerolamoGaslini 5, 16147 Genova Quarto, Italy. E-mail address: [email protected] Abbreviations used in this paper: Mn, monocyte; Mf, macrophage; iDC, immaturedendritic cell; pO2, partial oxygen pressure; HIF-1, hypoxia-inducible factor-1; GO,Gene Ontology; EASE, Expression Analysis Systematic Explorer; qRT-PCR, real-time quantitative PCR; VEGF, vascular endothelial growth factor; HMG, hypoxia-modulated gene; IRS, immunoregulatory signaling; ARNT, aryl hydrocarbon receptornuclear translocator; ECM, extracellular matrix; hMDM, human monocyte-derivedmacrophage; MMP, matrix metalloproteinase; bHLH, basic helix-loop-helix.

The Journal of Immunology

Copyright © 2006 by The American Association of Immunologists, Inc. 0022-1767/06/$02.00

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 3: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

transactivating the hypoxia responsive element present in the pro-moter of many O2-sensitive genes (9, 15). Regulation of HIF-1expression and activity by hypoxia is a tightly regulated processwhich results from the activity of several oxygen-dependent en-zymes and requires interaction and cooperation with various tran-scriptional cofactors and other transcription factors (15).

Mononuclear phagocytes accumulate preferentially in hypoxic/ischemic areas of diseased tissues (1), and hypoxic conditions havebeen shown to profoundly affect their proinflammatory and immu-noregulatory responses by modulating the expression of genescoding for angiogenic factors, inflammatory cytokines, and extra-cellular matrix (ECM) components/regulators (reviewed in Ref. 1).Recent evidence indicates that hypoxia can also strictly control thechemokine network in cells of the monocytic lineage not only byregulating the production of specific chemokines but also control-ling their action through the modulation of their receptors. Up-regulation of CCL3 (16), CXCL1 (1), CXCL8 (1), and CXCR4(17), and inhibition of CCL2 (16) and CCR5 (18) under hypoxiawere reported.

In the last few years, microarray technology has become animportant tool for the characterization of the molecular bases un-derlying cell response to stimulation (19). Recent investigationshave defined the transcriptional profile induced by hypoxia in invitro-derived human Mf (hMDM) (20, 21). Given their critical rolein the regulation of the initial phases of inflammation (1), it wasimportant to study primary Mn as a model of the early response tothe hypoxic environment. In this study, we report the first transcrip-tome analysis of primary human Mn following hypoxia exposure. Ourresults reveal the regulation by hypoxia of a cluster of novel genesrelevant to inflammation and immunity coding for surface molecules/markers, inflammatory cytokines/chemokines, and their receptors,and identify CCL20 as a new hypoxia-inducible gene.

Materials and MethodsCells and culture conditions

PBMC were isolated from platelet apheresis of healthy donors, obtained bythe Blood Transfusion Center of the Gaslini Institute (Genova, Italy), bydensity gradient centrifugation over a Ficoll cushion (Ficoll-Paque PLUS;Amersham Biosciences). Mn were then purified by countercurrent centrif-ugal elutriation using a Beckman JE-6 elutriation chamber and AvantiJ-20XP rotor system (Beckman Coulter), as described (22), followed byMACS magnetic bead separation (Human Monocyte Isolation kit-II; Milte-nyi Biotec). The purity of Mn preparations was �95%, as assessed bymorphology on Giemsa-stained cytocentrifuge slides and flow cytometrywith anti-CD14 mAb. Viability, determined by flow cytometry after DNAstaining with propidium iodide (5 �g/ml in PBS), was �98%.

Mn were plated in Costar plates (Celbio) in RPMI 1640 (Euroclone;Celbio) supplemented with 10% heat-inactivated FCS (HyClone; Celbio),2 mM L-glutamine, 100 U/ml penicillin, and 100 �g/ml streptomycin (Cel-bio) and maintained at 37°C in a humidified incubator containing 21% O2,5% CO2, and 75% N2, referred to as normoxic conditions. Hypoxic con-ditions (i.e., 1% O2) were achieved by incubating and handling the cells at37°C in a humidified anaerobic work station incubator (Bug Box; ALCInternational) flushed with a mixture of 94% N2, 5% CO2, and 1% O2.Culture medium was allowed to equilibrate for 3 h in a loosely capped flaskin the hypoxic incubator before cell exposure, and pO2 was monitoredusing a portable trace oxygen analyzer (Oxi 315i/set; WTW). The pO2 innormoxic medium ranged between 149 and 150 mm Hg, values that cor-respond to a 21% O2 concentration in an aqueous solution at 37°C and ata barometric pressure of 760 mm Hg, whereas the pO2 attained in themedium under hypoxic conditions was �7.1 mm Hg, which is equivalentto an O2 concentration of 1% and is in the range of the hypoxic levels foundin inflammatory tissues (8–12). The endotoxin content, determined by theLimulus amebocyte lysate test (QCL-1000; Bio-Whittaker), was �0.125endotoxin units/ml in all reagents.

RNA isolation and cRNA synthesis

Total RNA was purified from different donor-derived Mn using the RNeasyMini kit from Qiagen. The physical quality control of RNA integrity was

conducted by electrophoresis with an Agilent Bioanalyzer 2100 (AgilentTechnologies Europe). For each experimental condition, equal amounts ofMn RNA from 15 different donors were randomly pooled into three sub-sets, and the RNA pools were used for probe preparation. Briefly, 20 �g ofRNA were reverse transcribed into double-stranded cDNA on a GeneAmpPCR System 2700 thermal cycler (Applied Biosystems), using the Super-Script Double-Stranded cDNA Synthesis kit (Invitrogen Life Technolo-gies) according to the manufacturer’s instructions, except that a T7-(dT)24

primer (high purity salt-free purified) was used in place of the oligo pro-vided with the kit. cDNA was purified and used for in vitro transcriptionwith the BioArray High Efficiency RNA Transcript Labeling kit (Enzo LifeSciences) in the presence of biotin-11-CTP and biotin-16-UTP. LabeledcRNA was cleaned up using the Qiagen RNeasy Mini kit, checked forquality, and fragmented by incubation in mild alkaline buffer.

GeneChip hybridization and data analysis

Fragmented cRNA probes were used for hybridization to Affymetrix Hu-man Genome-U133A 2.0 GeneChips (Genopolis) containing 22,283 probesets corresponding to 18,400 transcripts. Each RNA pool was hybridized toan individual chip, and hybridization was performed at 45°C in the pres-ence of herring sperm DNA (0.1 mg/ml; Sigma-Aldrich). Chips were thenwashed with 6� standard saline citrate phosphate/EDTA (1� is 0.15 MNaCl, 0.01 M sodium phosphate (pH 7.4), and 1 mM EDTA), stained withstreptavidin-PE, and scanned using a confocal microscope scanner (HPGeneArray Scanner 2500) according to Affymetrix’s guidelines. Data cap-turing was conducted with standard Affymetrix analysis software algo-rithms (Microarray Suite 5.0), which selects the spots representative of atranscript and subtracts the background from the significant signals (23).Comparative analysis of hypoxic relative to normoxic expression profileswas conducted on GeneSpring 7.2 software (Silicon Genetics). Gene ex-pression data for each replicate experiment were normalized using the “perchip normalization” and “per gene normalization” algorithms implementedin the GeneSpring program. First, each signal was normalized based uponthe median signal in that chip (“per chip normalization”). Each correctedvalue was then normalized based upon the median of the measurements forthat gene in all samples (“per gene normalization”). This normalizationmethod, which removes the differing intensity scales and binding ratesfrom multiple experimental readings, allows the comparison of multipleGeneChip hybridizations (24). Finally, gene expression levels of replicateexperiments were averaged, and only genes that were modulated by at least1.5-fold in hypoxic relative to normoxic samples (means of three experi-ments) were considered differentially expressed. The significance of geneexpression differences between the two experimental conditions was cal-culated using a one-way ANOVA. Only genes that passed a Student’stwo-sample t test at a confidence level of 95% ( p value �0.05) wereconsidered significant. The complete data set for each microarray ex-periment was lodged in the ArrayExpress website �www.ebi.ac.uk/arrayexpress�. Gene Ontology (GO) data mining (25) for biological processat level 3 and Expression Analysis Systematic Explorer (EASE) biologicaltheme analysis (26) were conducted online at �http://david.niaid.nih.gov�using the Database for Annotation, Visualization, and Integrated Discovery(DAVID) 2.0 program (27).

Real-time RT-PCR

Real time quantitative PCR (qRT-PCR) of reverse-transcribed cDNA wasperformed on an I-Cycler (Bio-Rad), using iQ Supermix supplementedwith 10 nM fluorescein (Bio-Rad), 0.1� Sybr-Green I (Sigma-Aldrich),and 300 nM sense and antisense oligonucleotide primers (TIBMolbiol). Allprimer pairs (listed in Table I) were designed using Primer-3 software (28)from sequences in GenBank with a Tm optimum of 60°C and a productlength of 80–150 nt and tested before use to confirm appropriate productsize and optimal concentrations. qRT-PCR was conducted in triplicate foreach target transcript under the following cycling conditions: initial dena-turation of 3 min during which the well factor was measured, 50 cycles of15 s at 95°C followed by 30 s at 60°C. Fluorescence was measured duringthe annealing step in each cycle. After amplification, melting curves with80 steps of 15 s and 0.5°C increase were performed to monitor ampliconidentity. Expression data were normalized on the values obtained in par-allel for three reference genes (actin-related protein 2/3 complex 1B(ARPC1B); lysosomal-associated multispanning membrane protein-5(LAPTM5); and thrombospondin 1 (THBS1)) selected among those notaffected by hypoxia in the Affymetrix analysis, using the Bestkeeper soft-ware (29). Relative expression values were calculated using Q-gene soft-ware (30).

For semiquantitative PCR, reverse-transcribed cDNA was amplified intriplicate with recombinant TaqDNA polymerase (Invitrogen Life Tech-nologies) using the following cycling conditions: denaturation at 94°C,

1942 TRANSCRIPTIONAL PROFILE OF HYPOXIC MONOCYTES

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 4: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

annealing at 61°C, and extension at 72°C for 60 s for 30 cycles. Productswere separated by electrophoresis on a 1.2% agarose gel and visualized byethidium bromide staining.

Immunocytochemistry

A total of 1 � 105 Mn were applied to polysine glass slides by cytocen-trifuging at 900 rpm for 5 min. Cytospin preparations were fixed in 4%paraformaldehyde in PBS for 30 min at room temperature, and permeabil-

ized with 0.1% Triton X-100 in PBS for 5 min. Endogenous peroxidaseswere blocked with 0.3% hydrogen peroxide for 15 min. After rinsing inPBS, the slides were preincubated for 30 min in blocking buffer (PBSsupplemented with 2% human AB serum) and then incubated for 1 h withanti-human CCL20 mAb or an isotype-matched mAb (IgG1; R&D System)in blocking buffer. mAbs were detected with DakoCytomation Envision�

System Labeled Polymer-HRP anti-mouse. Peroxidase staining was re-vealed by 3-amino-9-ethylcarbazole (DakoCytomation), as a substrate.

Table I. Primer pairs used for real-time quantitative RT-PCRa

Geneb Primers Product (bp)

Actin-related protein 2/3 complex, subunit 1B (ARPC1B*) For 5�-aacgagaacaagtttgctgtg-3� 106Rev 5�-gatgggcttcttgatgtgc-3�

Activating transcription factor 5 (ATF5) For 5�-cccaccttctttcttcagc-3� 119Rev 5�-acgaggctctggaggatg-3�

Adrenomedullin (ADM) For 5�-cctgatgtacctgggttc-3� 118Rev 5�-ttccctcttcccacgact-3�

Arginase (ARG1) For 5�-attgagaaaggctggtctgc-3� 95Rev 5�-cattagggatgtcagcaaagg-3�

B cell activation gene (RGS1) For 5�-tgctgaagtaatgcaatggtct-3� 127Rev 5�-caagccagccagaactcaat-3�

BCL2/adenovirus E1B 19-kDa-interacting protein 3 (BNIP3) For 5�-ttccatctctgctgctctc-3� 80Rev 5�-tggtggaggttgtcagac-3�

Carbonic anhydrase XII (CA12) For 5�-cttggcatctgtattgtggtg-3� 121Rev 5�-tgggcctcagtctccatc-3�

CD163 Ag (CD163) For 5�-ttcgtcgcattattcttcttgac-3� 181Rev 5�-ggcaatagaccctccaag-3�

CD69 Ag (CD69) For 5�-gaacatggtgctactcttgc-3� 81Rev 5�-ttcctctctacctgcgtatc-3�

Chemokine (CC motif) ligand 20 (CCL20) For 5�-aatttattgtgggcttcacacg-3� 115Rev 5�-acccaagtctgttttggatttg-3�

Chemokine (CC motif) ligand 23 (CCL23) For 5�-ctttgaaacgaacagcgagtg-3� 179Rev 5�-cttgtgtcccttcaccttg-3�

Chemokine (CXC motif) receptor 4 (CXCR4) For 5�-gcatctggagaaccagcg-3� 111Rev 5�-gaaacagggttccttcatgg-3�

Fc fragment of IgG, high-affinity Ia, receptor for (FCGR1A) For 5�-atcgctacacatcagcagg-3� 137Rev 5�-ctgcaagagcaactttgtttc-3�

Fc fragment of IgG, low-affinity IIc, receptor for (FCGR2A) For 5�-acttctccatcccacaagc-3� 112Rev 5�-gagcttggacagtgatgg-3�

Fc fragment of IgG, low-affinity IIb, receptor for (FCGR2B) For 5�-ttacctgtccttgccactg-3� 124Rev 5�-agtttcagcacagcctttgg-3�

Hypoxia-inducible factor 1, � subunit (HIF1A) For 5�-aaatctcatccaagaagcccta-3� 118Rev 5�-cgctttctctgagcattctg-3�

IL-1R antagonist (IL1RN) For 5�-tcatgctctgttcttgggaat-3� 132Rev 5�-gcttgtcctgctttctgttc-3�

IL-6 signal transducer (IL-6ST) For 5�-ggcctaatgttccagatcc-3� 145Rev 5�-tcatttgcttctatttccacaaca-3�

Leukocyte-associated Ig-like receptor 1 (LAIR1) For 5�-cagattccgcattgactcag-3� 122Rev 5�-gaggtttctttcaccagcag-3�

Leukocyte Ig-like receptor, subfamily B, member 7 (LIR9) For 5�-gtatggtcagaacccagtg-3� 121Rev 5�-tgcgtaatcctgaaggtgtg-3�

Leukocyte membrane Ag (CMRF-35H) For 5�-gcactacgcaaatctggag-3� 163Rev 5�-tctgagcagctatcctgttg-3�

Lysosomal-associated multispanning membrane protein-5 (LAPTM5*) For 5�-ggtcacacctctgagtatg-3� 131Rev 5�-gtggaggagaagagaaactc-3�

Macrophage migration inhibitory factor (MIF) For 5�-gtccttctgccatcatgc-3� 166Rev 5�-gaaggccatgagctggt-3�

MAX-interacting protein 1 (MXI1) For 5�-agatggaacgaatacgaatgg-3� 110Rev 5�-gggagaactctgtgctttc-3�

N-myc downstream-regulated gene 1 (NDRG1) For 5�-ccttatcaacgtgaacccttg-3� 132Rev 5�-gttactctgcatttcttccttc-3�

Secreted phosphoprotein 1 (SPP1) For 5�-tgacccatctcagaagcag-3� 111Rev 5�-atggctttcgttggacttac-3�

Stabilin 1 (STAB1) For 5�-actcttcgtccctgtcaatg-3� 157Rev 5�-tcactgatgatgaggctgag-3�

Thrombospondin 1 (THBS1*) For 5�-cagcattctccatcaggaac-3� 125Rev 5�-gaggaatggactgttgatagc-3�

Vascular endothelial growth factor (VEGF) For 5�-gcagcttgagttaaacgaacg-3� 150Rev 5�-gcagcgtggtttctgtatc-3�

Vav 3 oncogene (VAV3) For 5�-tgttgtgagacgtttggaatg-3� 112Rev 5�-tgttcgagaaagtcgtgataatg-3�

a The expected PCR product size for each gene is shown. For, forward; Rev, reverse.b The * indicates the reference genes used for data normalization.

1943The Journal of Immunology

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 5: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

Slides were counterstained with hematoxylin, coverslipped with 80%glycerol in PBS, and examined with a phase contrast microscope (OlympusItalia). Photomicrographs were taken with a Zeiss camera.

ELISA

Secreted CCL20 was measured in cell-free supernatants using the Quan-tikine human CCL20 immunoassay kit from R&D Systems (Space ImportExport, according to the manufacturer’s instructions. The OD of the plateswere determined using a Spectrafluor Plus plate reader from Tecan at 450nm. All assays were done in duplicate and repeated three times.

ResultsGene expression profile of hypoxic Mn

Mn purified from 15 independent donors were cultured for 16 h in1% O2, a condition previously shown to effectively modulate geneexpression in these cells (17, 31), and the mRNA for the angio-genic factor, vascular endothelial growth factor (VEGF), was as-sessed by RT-PCR as an index of the response to hypoxia. Fig. 1shows VEGF mRNA levels in a representative subset of samples.Mn exposed to normoxic conditions expressed basal levels ofVEGF mRNA, though showing some degree of donor-to-donorvariation. Incubation under hypoxia caused a strong and consistentVEGF up-regulation in all the samples, in agreement with previousobservations (31).

The transcriptional profile of hypoxic Mn was then investigatedby microarray analysis. Equal amounts of RNA from the differentMn preparations were randomly combined into three pools thatwere independently hybridized to human Affymetrix HG-U133AGeneChips, obtaining three biological replicates for each experi-mental condition. Statistical analysis was performed as describedin Materials and Methods. Pairwise comparison among the datasets from normoxic and hypoxic samples (average of three exper-iments) revealed the differential modulation by hypoxia of a largenumber of transcripts (data not shown). After restricting the profileto those sequences exhibiting �1.5-fold expression differences inhypoxic relative to normoxic samples, we identified 536 up-regu-lated and 677 down-regulated genes. The majority of differentiallyexpressed genes were identified as unique and named in GenBank,whereas the remaining transcripts were either identified as un-named expressed sequence tags or were hypothetical. A selec-tion of hypoxia-modulated genes (HMGs) is presented in TablesII and III.

Functional classification of HMGs

Genes displaying at least 1.5-fold differential expression levelswere classified into various categories based on the biologicalfunction(s) of the encoded protein to determine the global directionof the molecular response to hypoxia. According to GO data min-ing for biological processes (Fig. 2), the transcriptional profile in-duced by hypoxia in Mn was mainly related to cell growth and/ormaintenance, signal transduction, nucleic acid, and protein metab-olism, these functional categories being the most enriched in both

up- and down-regulated genes. Moreover, hypoxic Mn showed aprominent expression of genes involved in organogenesis, re-sponse to stress, biosynthesis, and phosphorus metabolism. Inter-estingly, a significant number of HMGs coded for proteins impli-cated in cell response to external stimuli, immune response, celladhesion and motility, and cell-cell signaling, revealing a trendtoward inflammation and immunity (Fig. 2).

Comparison of microarray data with known hypoxia-inducedchanges in gene expression

In an initial verification of microarray data, we have cross-refer-enced our results with those of other studies investigating HMGs.As summarized in Table II, a large cluster of known HMGs wasaffected by hypoxia. In addition to the reference gene VEGF, wedemonstrated up-regulation of 20 genes related to angiogenesis,cell adhesion, transcription, and inflammatory responses, whichhave been previously identified as hypoxia inducible in cells of theMn lineage (Table II). The majority of them, including ad-renomedullin (ADM), arginase-1 (ARG1), coagulation factor III(F3), fibronectin-1 (FN1), IL-1� (IL-1A), IL-6, TNF-�, macro-phage migration inhibitory factor (MIF), matrix metalloprotein-ase-1 (MMP-1), osteopontin (SPP1), and VEGF receptor-1(FLT1), were reported to be increased by hypoxia in hMDM and/ormouse Mf (20, 32), but not in primary Mn. The expression ofanother 46 known HMGs characterized in cells types other thanmononuclear phagocytes was also triggered in hypoxic Mn (TableII), including classical genes involved in glycolytic metabolismand glucose transport (e.g., glucose transporter 1 and 3 (GLUT1, 3)(9, 13, 15, 33), or associated with nonglycolytic metabolism andion transport (e.g., carbonic anhydrase XII (CA12) (13, 33). Genescoding for two novel HMGs, hypoxia-inducible protein 2 (HIG2)(34) and HIF-1-responsive RTP801 (DDIT4) (15), and other HIF-1target genes implicated in HIF-1� hydroxylation, such as EGL-nine homolog-1 (EGLN1) (35) and proline 4-hydroxylases-AI,II(P4HA1, A2) (33, 36), were increased in hypoxic Mn (Table II).Finally, several genes related to apoptosis, cell cycle, transcription,and immune responses, whose responsiveness to hypoxia was pre-viously demonstrated in normal and malignant cells, were alsoup-regulated in hypoxic Mn. BACH1 transcription repressor (37),bHLH domain-containing B2 transcription factor (BHLHB2/DEC1) (15), BCL2/adenovirus E1B-interacting protein 3 (BNIP-3)(15), hepatocyte growth factor receptor (MET) (15), IL-4 (38), IL-1receptor antagonist (IL-1RN) (39), MAX-interacting protein-1(MXI1) (33), and N-myc downstream-regulated gene-1 (NDRG1)(33) represent a few examples (Table II). Interestingly, we alsodemonstrated inhibition by hypoxia of a cluster of inflammatorygenes previously found down-regulated in mMf and hMDM, suchas CCL2 (16), CCR5 (18), cathepsin C (CTSC) (21), 2,5-oligoad-enylate synthetase (OASE) (40), and the ras family member, RAB7(21). Overall, these data demonstrate that primary Mn share withMf and cells of other lineages a cluster of hypoxia-responsivegenes.

Identification of novel hypoxia-modulated genes in primary Mn

To identify genes not previously characterized in terms of respon-siveness to hypoxia, the hypoxic transcriptome of primary Mn wasfurther investigated after excluding known HMGs listed in TableII. A list of selected novel hypoxia-modulated genes is presentedin Table III. Of interest, several were enzymes or other moleculesimplicated in lipid metabolism with a role in the regulation of fattyacid and/or cholesterol biosynthesis or transport (Table III). Themost relevant are: apolipoprotein B48 receptor (APOB48R, up-regulated), apolipoprotein E (APOE, down-regulated), fatty acid

FIGURE 1. RT-PCR analysis of VEGF mRNA expression in humanMn. Human peripheral blood Mn purified from different donors were cul-tured for 16 h under normoxic () or hypoxic conditions (�), and totalRNA was reverse transcribed and tested for VEGF mRNA expression bysemiquantitative RT-PCR, as detailed in Materials and Methods. �-actinmRNA was assayed in parallel as an internal control for input RNA. Arepresentative experiment of two performed is shown

1944 TRANSCRIPTIONAL PROFILE OF HYPOXIC MONOCYTES

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 6: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

Table II. Relative expression of genes previously reported to be modulated by hypoxiaa

Function Gene Symbolb Gene Description Fold Changec Known in Mf/Mnd References

Angiogenesis ADM* Adrenomedullin 2.8 � (hMDM, peritoneal Mf) 9, 13, 15, 20, 33CALCRL Calcitonin receptor-like 1.5 79F3* Coagulation factor III (tissue factor) 4.5 � (hMDM) 20FGF1 Acid fibroblast growth factor 1 1.9 � (hMDM) 43FLT1 Vascular endothelial growth factor receptor 1 14.8 � (hMDM) 9, 13, 15, 20MIF* Macrophage migration inhibitory factor 10.1 � (hMDM) 13, 20SPP1* Secreted phosphoprotein 1 (osteopontin) 28.0 �/(hMDM) 13, 20VEGF Vascular endothelial growth factor 7.5 � (hMDM, hMn) 9, 13, 15, 20, 33WNT5A Wingless-type 5A 1.7 � (hMDM) 20

Apoptosis BCAT1* Branched chain aminotransferase 1 3.0 33BNIP3 BCL2/adenovirus E1B 19-kDa-interacting protein 3 16.5 15BNIP3L BCL2/adenovirus E1B 19-kDa-interacting protein 3-like 4.0 33MCL1 Myeloid cell leukemia sequence 1 (BCL2-related) 1.8 15TNF* Tumor necrosis factor 1.6 � (hMDM, peritoneal mMf) 43

Cell adhesion/matrix/ F3* Coagulation factor III (tissue factor) 4.5 � (hMDM) 20MMP1 Matrix metalloproteinase 1 (interstitial collagenase) 15.6 � (hMDM) 20FN1 Fibronectin 1 2.0 � (hMDM) 20SPP1* Secreted phosphoprotein 1 (osteopontin) 28.0 �/(hMDM) 13, 20TIMP-1 Tissue inhibitor of metalloproteinase 1 1.5 � (hMDDC) 44

Cell cycle/differentiation

ADM* Adrenomedullin 2.8 � (hMDM, peritoneal Mf) 9, 13, 15, 20, 33

BCAT1* Branched chain aminotransferase 1 3.0 33CCNG2 Cyclin G2 1.8 13, 33INHBA Inhibin, � A (activin A) 4.0 � (hMDM) 20MET met protooncogene (hepatocyte growth factor receptor) 1.8 15NDRG1 N-myc downstream regulated gene 1 4.1 33NPM1* Nucleophosmin 1.5 15

Glucose transport SLC2A1 (GLUT1) Solute carrier family 2 (glucose transporter), member 1 3.3 9, 13, 15SLC2A3 (GLUT3) Solute carrier family 2 (glucose transporter), member 3 5.8 9, 13, 33

Glycolytic metabolism ALDOA Aldolase A 1.6 9, 13, 15ALDOC Aldolase C 5.3 9ENO1* Enolase 1 3.0 9, 13, 15, 33ENO2 Enolase 2 38.9 33GAPDH Glyceraldehyde-3-phosphate dehydrogenase 1.6 9, 13, 15GPI Glucose phosphate isomerase 7.2 15HK2 Hexokinase 2 1.8 9LDHA Lactate dehydrogenase A 2.1 9, 13, 15PDK1 Pyruvate dehydrogenase kinase, isoenzyme 1 24.5 33PFKFB4 6-phosphofructo-2-kinasefructose-2,6-biphosphatase 4 2.0 15PFKP Phosphofructokinase, platelet 7.0 9, 13, 15, 33PGAM1 Phosphoglycerate mutase 1 2.4 15PGK1 Phosphoglycerate kinase 1 2.6 9, 13, 15, 33TPI1 Triose-phosphate isomerase 1 2.3 9

Immune/ responses ADORA2B Adenosine A2b receptor 2.5 80ARG1* Arginase I 2.8 � (peritoneal mMf) 32CCL2 Chemokine (CC motif) ligand 2 (MCP-1) 0.6 � (mMf) 16CCR5 Chemokine (CC motif) receptor 5 0.6 � (mMf) 18CD55 (DAF) Decay accelerating factor for complement 1.8 81CTSC Cathepsin C 0.3 � (hMDM) 21CTSD Cathepsin D 0.5 82CXCR4 Chemokine (CXC motif) receptor 4 1.9 � (hMn, hMf) 15, 17, 33IL1A Interleukin 1-� 3.2 � (peritoneal, alveolar Mf) 43IL4 Interleukin 4 1.8 38IL1RN Interleukin 1 receptor antagonist 3.1 39IL6 Interleukin 6 3.8 � (peritoneal mMf) 13, 43MIF* Macrophage migration inhibitory factor 10.1 � (hMDM) 13, 20OASE1 2,5-Oligoadenylate synthetase 1 0.5 � (mMf) 40OASE2 2,5-Oligoadenylate synthetase 2 0.5 � (mMf) 40SPP1* Secreted phosphoprotein 1 (osteopontin) 28.0 �/ (hMDM) 13, 20TNF* Tumor necrosis factor 1.6 � (hMDM, peritoneal mMf) 43

Metabolism (nonglycolytic) ADM* Adrenomedullin 2.8 � (hMDM, peritoneal Mf) 13, 15, 20, 33AK3 Adenylate kinase 3 2.5 9ARGI* Arginase I 2.8 � (peritoneal mMf) 32BCAT1* Branched chain aminotransferase 1 3.0 33CA12 Carbonic anhydrase XII 3.9 13, 33EGLN1 Egl nine homolog 1 4.6 35ERO1L ERO1-like 3.3 83GBE1 Glucan (1,4-�), branching enzyme 1 3.0 33P4HA1 Procollagen-proline, 2-oxoglutarate 4-dioxygenasa I 4.5 33, 36P4HA2 Procollagen-proline, 2-oxoglutarate 4-dioxygenasea II 2.2 33, 36PAM Peptidylglycine �-amidating monooxygenase 4.1 33PTGS2 Prostaglandin synthase and cyclooxygenase 2 3.6 � (hMDM) 20VLDLR Very low density lipoprotein receptor 5.3 � (hMn, hMDM) 41

Stress response DD1T4 HIF-1 responsive RTP801 3.1 15HIG2 Hypoxia-inducible protein 2 9.7 34WSB1 SOCS box-containing WD protein SWiP-1 2.4 33

Transcription/signaling BACH1 Bach1 transcription repressor 1.8 37BHLHB2 Basic helix-loop-helix domain containing, class B, 2 2.4 15EGR1 Early growth response 1 2.2 � (hMn) 43ENO1* Enolase 1 3.0 9, 13, 15, 33ID2 Inhibitor of DNA binding 2 1.6 � (hMDM) 15, 21, 33JMJD1A Zinc finger protein 3.2 33JUN Jun oncogene 2.2 13MXI1 MAX-interacting protein 1 4.5 33RAB7 Member RAS oncogene family 0.6 � (hMDM) 21NPM1* Nucleophosmin 1.5 15SNAPC1 Small nuclear RNA activating complex, polypeptide 1 3.6 33

a Gene expression profiling was carried out independently on three RNA pools, each composed by RNA from five different donor-derived Mn cultured under normoxic and hypoxicconditions, and comparative analysis of gene expression differences between the two experimental conditions was conducted as described in Materials and Methods. A function, a commongene symbol, a brief gene description, the fold change value, and published references are specified for each gene.

b The* indicates genes with more than one function appearing in multiple functional categories.c The indicated values represent the ratio of hypoxic/normoxic signals (mean of three experiments). A mean ratio �1.5 refers to an increase in hypoxic relative to normoxic expression

and a mean ratio �0.67 refers to a decrease in relative expression.d The � sign indicates genes previously shown to be induced by hypoxia in cells of the monocytic lineage; �/ indicates genes shown to require HIF-2� overexpression. hMn, human

monocytes; hMDM, human monocyte-derived macrophages; hMDDC, human monocyte-derived dendritic cells; hMf, human macrophages; mMf, mouse macrophages.

1945The Journal of Immunology

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 7: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

Tab

leII

I.R

elat

ive

expr

essi

onof

sele

cted

nove

lge

nes

defin

ing

the

tran

scri

ptom

eof

hypo

xic

prim

ary

mon

ocyt

esa

Up-

regu

late

dD

own-

regu

late

d

Gen

Ban

kSy

mbo

lG

ene

Des

crip

tion

Fold

Upb

Gen

Ban

kSy

mbo

lG

ene

Des

crip

tion

Fold

Dow

nb

Cyt

okin

es/c

hem

okin

esan

dre

cept

ors

NM

_020

327

AC

VR

1BA

ctiv

inA

rece

ptor

,ty

peIB

1.5

AF0

3158

7C

CL

15C

hem

okin

e(C

Cm

otif

)lig

and

14(M

IP-1

�)0.

18N

M_0

0459

1C

CL

20C

hem

okin

e(C

Cm

otif

)lig

and

20(M

IP-3

�)

7.4

AB

0002

21C

CL

18C

hem

okin

e(C

Cm

otif

)lig

and

18(P

AR

C,

MIP

-4)

0.30

NM

_016

326

CK

LF

Che

mok

ine-

like

fact

or3

1.7

U88

321

CC

L19

Che

mok

ine

(CC

mot

if)

ligan

d19

(Exo

dus-

3)0.

65N

M_0

0075

7C

SF1

Col

ony

stim

ulat

ing

fact

or1

(mac

roph

age)

1.6

U58

913

CC

L23

Che

mok

ine

(CC

mot

if)

ligan

d23

(CK

�8,

MP1

F-1)

0.33

NM

_000

759

CSF

3C

olon

yst

imul

atin

gfa

ctor

3(g

ranu

locy

te)

4.8

A19

8498

0C

CL

8C

hem

okin

e(C

Cm

otif

)lig

and

8(M

CP-

2)0.

26M

5773

1C

XC

L2

Che

mok

ine

(C�

Cm

otif

)lig

and

2(G

RO

-�)

1.9

NM

_001

295

CC

R1

Che

mok

ine

(CC

mot

if)

rece

ptor

10.

53N

M_0

0209

0C

XC

L3

Che

mok

ine

(C�

Cm

otif

)lig

and

3(G

RO

-�)

2.4

NM

_000

647

CC

R2

Che

mok

ine

(CC

mot

if)

rece

ptor

20.

64N

M_0

0299

4C

XC

L5

Che

mok

ine

(C�

Cm

otif

)lig

and

5(E

NA

-78)

3.1

NM

_005

211

CSF

1RC

olon

yst

imul

atin

gfa

ctor

1re

cept

or(v

fms)

0.63

U20

350

CX

3CR

1C

hem

okin

e(C

�3C

)re

cept

or1

1.5

NM

_000

760

CSF

3RC

olon

yst

imul

atin

gfa

ctor

3re

cept

or(g

ranu

locy

te)

0.66

NM

_019

618

IL1F

9In

terl

euki

n-1

fam

ily,

mem

ber

93.

5A

F002

985

CX

CL

11C

hem

okin

e(C

�C

mot

if)

ligan

d11

(1-T

AC

)0.

49N

M_0

0218

2IL

1RA

PIn

terl

euki

n1

rece

ptor

acce

ssor

ypr

otei

n3.

2N

M_0

0299

3C

XC

L6

Che

mok

ine

(C�

Cm

otif

)lig

and

6(G

CP-

2)0.

46N

M_0

1658

4IL

23A

Inte

rleu

kin

23p1

9su

buni

t2.

3N

M_0

0195

3E

CG

F1

Plat

elet

-der

ived

endo

thel

ial

cell

grow

thfa

ctor

10.

54N

M_0

0218

4IL

6ST

Inte

rleu

kin

6si

gnal

tran

sduc

er(g

p130

)1.

8N

M_0

0156

0IL

13R

A1

Inte

rleu

kin

13re

cept

or,

�1

0.65

NM

_007

199

IRA

K3

Inte

rleu

kin-

1re

cept

or-a

ssoc

iate

dki

nase

M1.

5A

I337

584

IL18

BP

Inte

rleu

kin

18bi

ndin

gpr

otei

n0.

63A

I817

041

RD

C1

Gpr

otei

n-co

uple

dch

emok

ine

orph

anre

cept

or4.

3N

M_0

0385

5IL

18R

1In

terl

euki

n18

rece

ptor

10.

50N

M_0

0461

2T

GF

BR

1T

rans

form

ing

grow

thfa

ctor

,�

rece

ptor

12.

7N

M_0

2179

8IL

21R

Inte

rleu

kin

21re

cept

or0.

45B

C00

1281

TN

FR

SF10

BT

umor

necr

osis

fact

orre

cept

orsu

perf

amily

10b

3.5

AI9

8311

5IL

27R

aIn

terl

euki

n27

rece

ptor

a0.

63B

F433

902

TN

FR

SF11

BT

umor

necr

osis

fact

orre

cept

orsu

perf

amily

11b

2.2

NM

_002

253

KD

R/F

Ik1

Kin

ase

dom

ain

rece

ptor

(VE

GFR

-2)

0.64

BE

5681

34T

NF

RSF

21T

umor

necr

osis

fact

orre

cept

orsu

perf

amily

213.

7N

M_0

0324

2T

GF

BR

2T

rans

form

ing

grow

thfa

ctor

,�

rece

ptor

II0.

48B

C00

6196

TN

FR

SF9

Tum

orne

cros

isfa

ctor

rece

ptor

supe

rfam

ily9

1.5

BC

0050

43T

NF

RSF

10C

Tum

orne

cros

isfa

ctor

rece

ptor

supe

rfam

ily10

c0.

51N

M_0

0380

7T

NF

SF14

Tum

orne

cros

isfa

ctor

(lig

and)

supe

rfam

ily14

4.9

NM

_003

810

TN

FSF

10T

umor

necr

osis

fact

orsu

perf

amily

100.

51N

M_0

0511

8T

NF

SF15

Tum

orne

cros

isfa

ctor

(lig

and)

supe

rfam

ily15

1.7

AF1

1401

3T

NF

SF13

Tum

orne

cros

isfa

ctor

supe

rfam

ily13

0.47

NM

_021

138

TR

AF

2T

NF

rece

ptor

-ass

ocia

ted

fact

or2

0.52

Cyt

oske

leto

nan

dE

CM

com

pone

nts

orre

gula

tors

AI8

1452

7A

DA

M8

Dis

inte

grin

and

met

allo

prot

eina

se8

(CD

156a

)2.

3U

4873

4A

CT

N4

Act

inin

,�

40.

60A

B01

1536

CE

LSR

3C

adhe

rin,

EG

FL

AG

seve

n-pa

ssG

-typ

ere

cept

or3

1.8

NM

_021

777

AD

AM

28D

isin

tegr

inan

dm

etal

lopr

otei

nase

dom

ain

280.

49N

M_0

0009

4C

OL

7A1

Col

lage

n,ty

peV

II,

�1

3.7

AB

0023

64A

DA

MT

S3D

isin

tegr

inan

dm

etal

lopr

otea

sew

ithth

rom

bosp

ondi

n0.

52A

L12

1981

DL

G1

Dis

cspr

otei

n2.

0A

I796

813

AV

ILA

dvill

in0.

33A

I797

281

DSC

3D

esm

ocol

lin3

1.6

NM

_004

342

CA

LD

1C

alde

smon

10.

55N

M_0

0441

5D

SPD

esm

opla

kin

1.6

AI3

4123

4C

OR

O1B

Cor

onin

,ac

tin-b

indi

ngpr

otei

n,1B

0.21

M62

994

FL

NB

Fila

min

B,

�3.

1L

3559

4E

NP

P2

Aut

otax

in0.

19N

M_0

0152

3H

AS1

Hya

luro

nan

synt

hase

13.

2N

M_0

1328

1F

LR

T3

Fibr

onec

tinle

ucin

e-ri

chtr

ansm

embr

ane

prot

ein

30.

55A

J406

939

KR

TA

P4-

7K

erat

in-

asso

ciat

edpr

otei

n4.

71.

9A

I633

888

FY

BFY

N-b

indi

ngpr

otei

n0.

42L

2554

1L

AM

B3

Lam

inin

SB

3ch

ain

1.5

NM

_021

991

JUP

Junc

tion

plak

oglo

bin

0.33

AF3

4281

6L

GA

LS8

Gal

ectin

-8,

lect

inga

lact

osid

e-bi

ndin

g,so

lubl

e,8

2.8

U76

549

KR

T8

Cyt

oker

atin

80.

45N

M_0

2256

4M

MP

16M

atri

xm

etal

lopr

otei

nase

162.

4J0

3202

LA

MC

1L

amin

in�

10.

62N

M_0

0242

9M

MP

19M

atri

xm

etal

lopr

otei

nase

191.

5N

M_0

0649

8L

GA

LS2

Gal

ectin

2,le

ctin

,ga

lact

osid

e-bi

ndin

g,so

lubl

e,2

0.24

NM

_015

368

PA

NX

1Pa

nnex

in1

2.4

L16

895

LO

XH

uman

lysy

lox

idas

e0.

20A

A18

7563

PA

RV

BPa

rvin

,�

2.1

NM

_022

718

MM

P25

Mat

rix

met

allo

prot

eina

se25

0.65

BC

0064

39P

CD

HG

C3

Prot

ocad

heri

n�

subf

amily

A,

52.

3N

M_0

2287

0M

YH

11M

yosi

n,he

avy

poly

pept

ide

110.

12N

M_0

0029

7P

KD

2Po

lycy

stic

kidn

eydi

seas

e2

2.3

NM

_004

819

SYM

PK

Gsy

mpl

ekin

0.54

NM

_002

92R

GS1

Reg

ulat

orof

G-p

rote

insi

gnal

ing

12.

4N

M_0

0324

7T

HB

S2T

hrom

bosp

ondi

n2

0.45

NM

_006

932

SMT

NSm

ooth

elin

6.4

NM

_005

428

VA

V1

vav

1on

coge

ne0.

65N

M_0

0309

8SN

TA

1Sy

ntro

phin

,�

12.

3A

F118

887

VA

V3

vav

3on

coge

ne0.

55J0

3225

TF

PI1

Tis

sue

fact

orpa

thw

ayin

hibi

tor

11.

6N

M_0

1400

0V

CL

Vin

culin

0.59

L27

624

TF

PI2

Tis

sue

fact

orpa

thw

ayin

hibi

tor

25.

5N

M_0

0346

1Z

YX

Zyx

in0.

55N

M_0

2264

8T

NS1

Ten

sin

116

.7(T

able

cont

inue

s)

1946 TRANSCRIPTIONAL PROFILE OF HYPOXIC MONOCYTES

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 8: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

Tab

leII

I.C

onti

nued

Up-

regu

late

dD

own-

regu

late

d

Gen

Ban

kSy

mbo

lG

ene

Des

crip

tion

Fold

Upb

Gen

Ban

kSy

mbo

lG

ene

Des

crip

tion

Fold

Dow

nb

Lip

idm

etab

olis

mN

M_0

1836

1A

GP

AT

5A

cid

acyl

tran

sfer

asee

psilo

n2.

0N

M_0

0199

5A

CSL

1Fa

ttyac

idac

yl-C

oAsy

ntha

selo

ng-c

hain

fam

ily1

0.56

NM

_018

690

AP

OB

48R

Apo

lipop

rote

inB

48re

cept

or1.

7N

M_0

0445

7A

CSL

3Fa

ttyac

idac

yl-C

oAsy

ntha

selo

ng-c

hain

fam

ily3

0.53

NM

_001

646

AP

OC

4A

polip

opro

tein

C-I

V1.

8N

M_0

0164

5A

PO

C1

Apo

lipop

rote

inC

I0.

62N

M_0

0070

9B

CK

DH

AB

ranc

hed

chai

nke

toac

idde

hydr

ogen

ase

El�

1.6

N33

009

AP

OE

Apo

lipop

rote

inE

0.32

NM

_024

306

FA

2HFa

ttyac

idhy

drox

ylas

e(F

AA

H)

1.6

BC

0043

95A

PO

L2

Apo

lipop

rote

inL

20.

40N

M_0

0144

2F

AB

P4

Fatty

acid

bind

ing

prot

ein

41.

8N

M_0

0078

4C

YP

27A

1St

eroi

d27

-hyd

roxy

lase

(cyt

ochr

ome

P450

,X

XV

IIA

)0.

30A

I927

993

OSB

PO

xyst

erol

bind

ing

prot

ein

1.7

NM

_001

444

FA

BP

5Fa

ttyac

id-b

indi

ngpr

otei

n5

0.62

NM

_017

784

OSB

PL

10O

xyst

erol

bind

ing

prot

ein-

like

102.

1A

I861

942

LD

LR

Low

dens

itylip

opro

tein

rece

ptor

0.65

U78

577

PIP

5K1A

Phos

phat

idyl

inos

itol-

4-ph

osph

ate

5-ki

nase

�1.

6B

F304

759

LR

P1

Low

dens

itylip

opro

tein

-rel

ated

prot

ein

10.

54B

C00

3393

PIK

3CB

Phos

phoi

nosi

tide-

3-ki

nase

,ca

taly

tic,

�3.

0N

M_0

0463

1L

RP

8L

owde

nsity

lipop

rote

inre

cept

or-r

elat

edpr

otei

n8

0.61

M61

906

PIK

3R1

Phos

phoi

nosi

tide-

3-ki

nase

poly

pept

ide

11.

5W

1998

3O

SBP

L1A

Oxy

ster

ol-b

indi

ngpr

otei

n-lik

e1A

0.41

AK

0235

46P

LC

L2

Phos

phol

ipas

eC

-lik

e2

2.1

AW

7710

15P

LC

E1

Phos

phol

ipas

eC

,�

10.

58A

F297

052

PT

GIS

Pros

tacy

clin

synt

hase

(CY

P8A

1)2.

0N

M_0

0622

6P

LC

L1

Phos

phol

ipas

eC

-lik

e1

0.48

AL

1175

15P

LC

L2

Phos

phol

ipas

eC

-lik

e2

0.54

Surf

ace

mol

ecul

es/r

ecep

tors

W72

082

C1Q

R1

Com

plem

ent

com

pone

nt1q

,re

cept

or1

4.2

M81

695

CD

11C

Inte

grin

,�

X,

leuk

ocyt

ead

hesi

onp1

500.

61N

M_0

0613

9C

D28

CD

28A

g(T

p44)

1.7

NM

_004

334

CD

157/

BST

1B

one

mar

row

stro

mal

cell

Ag

10.

66N

M_0

0061

1C

D59

CD

59A

gp1

8-20

(pro

tect

in)

1.6

Z22

970

CD

163

CD

163

Ag

0.16

L07

555

CD

69E

arly

activ

atio

nA

gC

D69

1.8

M98

399

CD

36C

D36

Ag

(thr

ombo

spon

din

rece

ptor

)0.

41N

M_0

0178

3C

D79

AC

D79

aA

g1.

6B

G23

0614

CD

47C

D47

Ag

(int

egri

n-as

soci

ated

prot

ein)

0.55

NM

_006

889

CD

86C

D86

Ag

(B7-

2A

g)1.

9N

M_0

0387

4C

D84

CD

84A

g(l

euko

cyte

Ag)

0.31

NM

_006

678

CM

RF

35L

euko

cyte

Ig-l

ike

rece

ptor

2.1

AF2

0073

8C

LE

CSF

6C

-typ

ele

ctin

supe

rfam

ilym

embe

r6

(DC

IR)

0.56

AJ0

1010

2C

MR

F-3

5HL

euko

cyte

mem

bran

eA

g26

.0X

1435

5F

CG

R1A

Fcfr

agm

ent

ofIg

G,

Ia,

rece

ptor

for

(CD

64)

0.33

U73

304

CN

R1

CB

1ca

nnab

inoi

dre

cept

or3.

4N

M_0

2391

4G

PR

86G

prot

ein-

coup

led

rece

ptor

860.

11N

M_0

3078

1C

OL

EC

12Sc

aven

ger

rece

ptor

with

G-t

ype

lect

in.1

1.7

M90

686

HL

A-G

HL

A-G

hist

ocom

patib

ility

Ag

clas

sIG

0.61

U56

237

FC

AR

Fcfr

agm

ent

ofIg

A,

rece

ptor

for

(CD

89)

4.2

NM

_002

162

ICA

M3

Inte

rcel

lula

rad

hesi

onm

olec

ule

30.

62N

M_0

2164

2F

CG

R2A

Fcfr

agm

ent

ofIg

G,

IIa,

rece

ptor

for

(CD

32)

1.9

AI3

3520

8IT

GB

5In

tegr

in,

�5

0.17

M31

933

FC

GR

2BFc

frag

men

tof

IgG

,II

b,re

cept

orfo

r(C

D32

)4.

3N

M_0

0088

9IT

GB

7In

tegr

in,

�7

0.61

NM

_000

570

FC

GR

3AFc

frag

men

tof

IgG

,II

Ib,

rece

ptor

for

(CD

16)

1.9

AF1

0968

3L

AIR

1L

euko

cyte

-ass

ocia

ted

Ig-l

ike

rece

ptor

10.

49N

M_0

2162

4H

RH

4H

ista

min

eH

4re

cept

or1.

7A

F009

007

LIL

RB

1L

euko

cyte

Ig-l

ike

rece

ptor

B1

(CD

85j)

0.65

NM

_003

259

ICA

M5

Inte

rcel

lula

rad

hesi

onm

olec

ule

52.

0A

F004

231

LIL

RB

2L

euko

cyte

Ig-l

ike

rece

ptor

B2

(CD

85d)

0.52

L76

666

KIR

3DL

2K

iller

cell

imm

unog

lobu

lin-l

ike

rece

ptor

1.8

AF0

0963

5L

ILR

B3

Leu

kocy

teIg

-lik

ere

cept

orB

30.

56N

M_0

1652

3K

LR

F1

Kill

erce

llle

ctin

-lik

ere

cept

orF1

1.9

U82

979

LIL

RB

4L

euko

cyte

Ig-l

ike

rece

ptor

B4

0.57

NM

_006

770

MA

RC

OM

acro

phag

ere

cept

orw

ithco

llage

nous

stru

ctur

e2.

0A

F212

842

LIR

9Ig

-lik

etr

ansc

ript

11pr

otei

n(I

LT

11)

0.62

NM

_002

445

MSR

1M

acro

phag

esc

aven

ger

rece

ptor

12.

1N

M_0

0234

6L

Y6E

Lym

phoc

yte

Ag

6co

mpl

ex,

locu

sE

0.59

NM

_003

693

SCA

RF

1A

cety

lL

DL

rece

ptor

,sc

aven

ger

rece

ptor

F11.

8N

M_0

0634

3M

ER

TK

c-m

erpr

oto-

onco

gene

tyro

sine

kina

se0.

15A

I949

392

SEM

A4C

Sem

apho

rin

4C1.

7N

M_0

2306

8SN

Sial

oadh

esin

(CD

169)

0.09

NM

_006

378

SEM

A4D

Sem

apho

rin

4D(C

D10

0)1.

8N

M_0

1513

STA

B1

Stab

ilin

10.

25N

M_0

0426

3SE

MA

4FSe

map

hori

n4F

1.6

AF0

5115

1T

LR

5T

oll-

like

rece

ptor

50.

56N

M_0

1864

3T

RE

M1

Tri

gger

ing

rece

ptor

expr

esse

don

mye

loid

cells

12.

4N

M_0

1656

2T

LR

7T

oll-

like

rece

ptor

70.

24(T

able

cont

inue

s)

1947The Journal of Immunology

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 9: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

Tab

leII

I.C

onti

nued

Up-

regu

late

dD

own-

regu

late

d

Gen

Ban

kSy

mbo

lG

ene

Des

crip

tion

Fold

Upb

Gen

Ban

kSy

mbo

lG

ene

Des

crip

tion

Fold

Dow

nb

Tra

nscr

iptio

nfa

ctor

sN

M_0

0188

0A

TF

2A

ctiv

atin

gtr

ansc

ript

ion

fact

or2

1.5

NM

_001

621

AH

RA

ryl

hydr

ocar

bon

rece

ptor

0.57

NM

_012

068

AT

F5

Act

ivat

ing

tran

scri

ptio

nfa

ctor

51.

8N

M_0

1486

2A

RN

T2

Ary

lhy

droc

arbo

nre

cept

ornu

clea

rtr

ansl

ocat

or2

0.37

U79

751

BL

ZF

1B

asic

leuc

ine

zipp

ernu

clea

rfa

ctor

1(J

EM

-1)

1.6

U85

962

CB

PC

RE

B-b

indi

ngpr

otei

n0.

49N

M_0

0443

0E

GR

3E

arly

grow

thre

spon

se3

2.4

NM

_004

364

CE

BP

AC

CA

AT

enha

ncer

bind

ing

prot

ein

(CE

BP)

,�

0.51

NM

_006

732

FO

SBFo

sho

mol

ogB

2.0

AV

6556

40C

EB

PD

CC

AA

T/e

nhan

cer

bind

ing

prot

ein

(C/E

BP)

,�

0.55

NM

_024

530

FO

SL2

Fos-

like

Ag

21.

7N

M_0

0490

4C

RE

B5

cAM

Pre

spon

seel

emen

t-bi

ndin

gpr

otei

n5

0.46

BC

0053

29H

SF2

Hea

tsh

ock

tran

scri

ptio

nfa

ctor

22.

7N

M_0

0039

9E

GR

2E

arly

grow

thre

spon

se2

0.55

D13

889

ID1

Inhi

bito

rof

DN

Abi

ndin

g1

2.2

NM

_005

890

GA

S7G

row

thar

rest

-spe

cific

70.

20N

M_0

0246

0IR

F4

IFN

regu

lato

ryfa

ctor

41.

5N

M_0

0153

0H

IF1A

Hyp

oxia

-ind

ucib

lefa

ctor

1,�

subu

nit

0.50

NM

_005

655

KL

F10

TG

FBin

duci

ble

earl

ygr

owth

resp

onse

4.5

NM

_002

383

MA

ZM

YC

-ass

ocia

ted

zinc

finge

rpr

otei

n0.

41N

M_0

0250

5N

FY

AN

ucle

artr

ansc

ript

ion

fact

orY

,�

1.8

NM

_000

248

MIT

FM

icro

phth

alm

ia-a

ssoc

iate

dtr

ansc

ript

ion

fact

or0.

55M

2198

5N

R2C

1T

R2

nucl

ear

rece

ptor

2C1

3.3

NM

_005

933

ML

LM

yelo

id/ly

mph

oid

orm

ixed

-lin

eage

leuk

emia

0.28

NM

_006

186

NR

4A2

Nuc

lear

rece

ptor

subf

amily

4A2

2.2

NM

_002

432

MN

DA

Mye

loid

cell

nucl

ear

diff

eren

tiatio

nA

g0.

20L

3459

8R

UN

X1

Run

t-re

late

dtr

ansc

ript

ion

fact

or1

1.9

NM

_002

467

MY

Cv-

myc

avia

nm

yelo

cyto

mat

osis

vira

lon

coge

ne0.

47L

4099

2R

UN

X2

Run

t-re

late

dtr

ansc

ript

ion

fact

or2

1.7

U19

179

NC

OA

1N

ucle

arre

cept

orco

activ

ator

10.

49N

M_0

0311

2SP

4Sp

4tr

ansc

ript

ion

fact

or1.

7N

M_0

1625

0N

DR

G2

N-m

ycdo

wns

trea

m-r

egul

ated

gene

20.

53A

F077

053

TA

F9L

Neu

rona

lce

llde

ath-

rela

ted

prot

ein

2.9

BF2

0950

7P

C4

Act

ivat

edR

NA

poly

mer

ase

IItr

ansc

ript

ion

cofa

ctor

40.

56A

I703

074

TC

F7L

2T

rans

crip

tion

fact

or7-

like

22.

8A

V72

7449

PC

AF

p300

/CB

P-as

soci

ated

fact

or0.

65B

F056

105

TC

FL

4T

rans

crip

tion

fact

or-l

ike

41.

6B

C00

0080

PM

LPr

omye

locy

ticle

ukem

ia0.

34N

M_0

1590

5T

IF1

Tra

nscr

iptio

nal

inte

rmed

iary

fact

or1

1.7

NM

_003

120

SPI1

Vir

uspr

ovir

alin

tegr

atio

non

coge

nesp

il0.

37K

0319

9T

P53

p53

cellu

lar

tum

orA

g0.

40N

M_0

0699

1Z

NF

197

Zin

cfin

ger

prot

ein

197

0.62

aG

ene

expr

essi

onpr

ofilin

gw

asca

rrie

dou

tin

depe

nden

tlyon

thre

eR

NA

pool

s,ea

chco

mpo

sed

byR

NA

from

five

diff

eren

tdo

nor-

deri

ved

Mn

cultu

red

unde

rno

rmox

ican

dhy

poxi

cco

nditi

ons,

and

com

para

tive

anal

ysis

ofge

neex

pres

sion

diff

eren

ces

betw

een

the

two

expe

rim

enta

lco

nditi

ons

was

cond

ucte

das

desc

ribe

din

Mat

eria

lsan

dM

etho

ds.

Eac

hge

neis

give

na

repr

esen

tativ

eG

enB

ank

acce

ssio

nnu

mbe

r,a

com

mon

gene

sym

bol,

abr

ief

gene

desc

ript

ion,

and

the

fold

chan

geva

lue.

Und

erlin

edge

nes

wer

eva

lidat

edby

qRT

-PC

R.

bT

hein

dica

ted

valu

esre

pres

ent

the

ratio

ofhy

poxi

c/no

rmox

icsi

gnal

s(m

eans

ofth

ree

expe

rim

ents

).A

mea

nra

tio�

1.5

refe

rsto

anin

crea

sein

hypo

xic

rela

tive

tono

rmox

icex

pres

sion

(fol

dup

)an

da

mea

nra

tio�

0.67

refe

rsto

ade

crea

sein

rela

tive

expr

essi

on(f

old

dow

n).

1948 TRANSCRIPTIONAL PROFILE OF HYPOXIC MONOCYTES

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 10: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

acyl-CoA synthase long-chain family 1 and 3 (ACSL1,3, down-regulated), fatty acid binding protein 4 (FABP4, up-regulated), lowdensity lipoprotein receptor (LDLR, down-regulated), oxysterolbinding protein-like 10 (OSBPL10, up-regulated), and steroid 27-hydroxylase (CYP27A1, down-regulated). Modulation of thesegenes under low pO2 may have implications for the pathogenesisof atherosclerosis and Alzheimer’s disease, where a role for hyp-oxia has been suggested (9, 10, 41).

Other genes differentially expressed in hypoxic Mn coded forcytoskeleton and ECM components/regulators (Table III). Induc-ible genes include adhesive proteins of the desmosome type ofcell-cell junction, actin-interacting/regulatory transmembrane mol-ecules, and secreted proteins. Of note are galectin 8 (LGALS8),MMP-16 (MT3-MMP), MMP-19, regulator of G-protein signaling1 (RGS1), tensin 1 (TNS1), and tissue factor pathway inhibitors 1and 2 (TFPI1,2). Among the most significantly down-regulatedgenes, we identified advillin (AVIL), autotaxin (ENPP2), galectin2 (LGALS2), myosin heavy polypeptide 11 (MYH11), and VAV3.Modulation of these genes is likely to regulate Mn adhesion, mo-tility, and tissue remodeling.

The hypoxic profile also revealed hypoxia inducibility of a set ofgenes with transcription regulatory activity, including activatingtranscription factor (ATF)-2 and ATF-5, Fos homolog B (FOSB),Fos-like Ag 2 (FRA2), and runt-related transcription factors 1 and2 (RUNX1,2) (Table III). Moreover, several transcription factor-and cofactor-encoding genes were inhibited by hypoxia. Of rele-vance are the aryl-hydrocarbon receptor (AHR), ARNT2, CCAAT

enhancer-binding protein � (C/EBP�), CREB-binding protein(CBP), HIF-1�, microphthalmia-associated transcription factor(MITF), MYC oncogene, nuclear receptor coactivator 1 (NCOA1/SRC-1), p53 tumor Ag (TP53), and zinc finger protein 197(ZNF197/VHLaK) (Table III). These data are indicative of major,coordinated changes in transcription and suggest the existence ofboth positive and negative O2-driven feedback regulatory mecha-nisms of hypoxia transcriptional response

Differential modulation by hypoxia of immune-related genesin Mn

As summarized in Table III, a prominent set of novel HMGs haveimmunological relevance. These include genes encoding surfaceimmunoregulatory signaling (IRS) receptors, such as early activa-tion Ag (CD69), leukocyte membrane Ag CMRF-35H, low-affinityIgG receptors Fc�RIIA,B (CD32), IgA receptor Fc�R (CD89), andtriggering receptor expressed on myeloid cells 1 (TREM1), thatwere up-regulated, and high-affinity IgG receptor Fc�RIA (CD64),histocompatibility Ag class IG (HLA-G), leukocyte-associated Ig-like receptor 1 (LAIR1), leukocyte Ig-like receptor 9 (LIR9), andB1,B2,B3,B4 (LILRB1–4, CD85), that were down-regulated. Sev-eral scavenging and pattern recognition receptors were also selec-tively induced (complement component 1q receptor 1 (C1qR1);macrophage receptor with collagenous structure (MARCO); mac-rophage scavenger receptor 1 (MSR1); scavenger receptor-FI(SCARF1); scavenger receptor with C-type lectin 1 (COLEC12/SRCL)) or repressed (CD163 Ag; stabilin, STAB-1; TLR-5 and -7)

FIGURE 2. GO data mining. The gene expression profile of hypoxic vs normoxic primary human Mn was analyzed using high-density oligonucleotidearrays, as described in Materials and Methods. Genes showing at least a 1.5-fold change in expression levels between hypoxic and normoxic cells (meanof three experiments) were selected and characterized according to their biological process classification (at level 3) in the GO database. Based on thisclassification scheme, genes were placed in more than one category if more than one function of the encoded protein had been established. Transcriptswithout a GO classification were categorized as unclassified. Bars on the right of the y-axis represent up-regulated genes; bars on the left of the y-axisrepresent down-regulated genes.

1949The Journal of Immunology

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 11: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

under hypoxia (Table III). Other differentially expressed genescoded for costimulatory and adhesion molecules involved in cell-cell and cell-matrix interaction, such as bone marrow stromal cellAg (CD157), CD36, CD84, and CD86 Ags, integrin � X (CD11C),integrin �5 and �7 (ITGB5,7), ICAM3,5, semaphorin 4D (CD100),and sialoadhesin (CD169) (Table III).

The hypoxic transcriptome was also characterized by the mod-ulation of a cluster of genes coding for cytokines/chemokinesand/or their receptors. Within the chemokine system, we identifiedfor the first time CCL20, CXCL2, CXCL3, CXCL5, the fractalkinereceptor CX3CR1, and the G protein-coupled chemokine orphanreceptor (RDC1) as hypoxia-inducible genes, whereas CCL15,CCL18, CCL23, CCL8, CXCL6, CCR1, and CCR2 were the mosthighly hypoxia-repressed genes (Table III). Finally, Mn hypoxicprofile included various components of the IL-1 system (IL-1 fam-ily member 9 (IL1F9); IL-1 receptor accessory protein (IL1RAP);IL-1R associated kinase 3 (IRAK-3); IL-18R1) and members of theTNF receptor and ligand superfamilies, as well as IL-23A, IL-6signal transducer (IL-6ST), IL-13RA1, IL-21R, CSF1,3, andCSF1,3Rs (Table III).

Confirmation of microarray data by qRT-PCR analysis ofselected hypoxia-modulated genes

To validate the microarray results using a different technique,mRNA levels for a subset of known and novel hypoxia-modulatedgenes were quantified by qRT-PCR on a new RNA pool (Fig. 3),using the primer pairs listed in Table I. For this analysis, we ran-domly selected 27 genes involved in immune regulation, inflam-matory responses, and transcription. Three reference genes (TableI) were used for data normalization. We found a 100% concor-dance between qRT-PCR and Affymetrix data with respect to thedirection of the expression changes. For the majority of the genes,fold-differences were also of comparable magnitude (Fig. 3), al-though they were higher according to qRT-PCR for six genes

(CCL23, FCGR1A, LIR9, BNIP-3, Mxl1, and VEGF), in agreementwith previous findings showing that microarray can often under-estimate the extent of gene regulation compared with qRT-PCR(Varesio et al., unpublished observations). For other genes, such asCMRF-35H, FCGR2B, STAB-1, and MIF, however, higher expres-sion differences were detected by microarray. These results con-firm hypoxia responsiveness of novel genes identified bymicroarray.

Hypoxia induces CCL20 expression and secretion by human Mn

We then selected CCL20, one of the novel immune-related genesmost strongly up-regulated in hypoxic Mn, for further analysis.According to microarray and qRT-PCR data, CCL20 mRNA levelswere increased by an average of 7.4- and 5.7-fold, respectively(Fig. 3). To address the issue of donor-to-donor variability, weanalyzed CCL20 mRNA expression by RT-PCR in a subset ofsamples comprising the pools used for microarrays (Fig. 4A).CCL20 basal expression was detected in all Mn preparations cul-tured under normal pO2, although with some variations amongindividual donors. Consistent CCL20 transcript up-regulation wastriggered by hypoxia in every donor, independently of the baselinelevels (Fig. 4A), indicating the general inducibility of the gene inprimary Mn. qRT-PCR was also performed to quantify the mag-nitude of hypoxia-induced changes. As shown in Fig. 4B, the ex-tent of CCL20 mRNA increase ranged from 3.1- to 11.4-foldamong the samples examined. mRNA up-regulation was paralleledby increased protein expression, as determined by immunocyto-chemistry (Fig. 5A). Mn isolated from the three donors analyzedby qRT-PCR were cytocentrifuged and immunostained with amAb directed to CCL20. Low levels of CCL20 immunoreactivitywere detectable in the cytoplasm of normoxic Mn. Hypoxia expo-sure for 16 h resulted in a marked increase in intracellular proteincontent in all the samples. No staining was detected when an iso-type-matched control Ab was used (Fig. 5A).

FIGURE 3. qRT-PCR analysis of genes selected from the microarray profile. Equal amounts of total RNA from four donor-derived normoxic or hypoxicMn were pooled, and RNA pools were subjected to qRT-PCR analysis, as detailed in Materials and Methods. Expression changes of 27 selected genes wereevaluated in relation to the values obtained in parallel for three reference genes. Results from one representative experiment of three performed areexpressed as log2 ratio (1% O2 relative to 21% O2) and are the mean of triplicate determinations for each target transcript (f). Microarray data are shownfor comparison (u). The number associated with each bar indicates the linear fold-change of mRNA expression in hypoxic relative to normoxic cells(arbitrarily defined as equal to 1). Positive values indicate that the mRNA level of a particular gene was up-regulated, whereas negative values indicate thatthe transcript was down-regulated. Known hypoxia-inducible genes identified for the first time in cells of the Mn lineage are marked with an asterisk (�).

1950 TRANSCRIPTIONAL PROFILE OF HYPOXIC MONOCYTES

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 12: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

Conditioned medium from normoxic and hypoxic Mn cultureswas then analyzed for CCL20 content by ELISA to determinewhether a parallel rise in chemokine secretion was triggered byhypoxia (Fig. 5B). Normoxic Mn constitutively secreted variableamounts of CCL20, which accumulated in the culture mediumranging from 10 to 410 pg/106 cells/ml in six different donors.Consistent with the mRNA data, chemokine secretion increased by2.4- to 3.5-fold upon incubation to 1% O2 for 16 h (Fig. 5B) andwas further augmented at longer time points, with almost 7-foldhigher amounts detectable in the conditioned medium at 72 h ofculture (Fig. 5C, donor 12). CCL20 is a LPS-inducible chemokine(3) and the response of Mn to LPS was evaluated in parallel ex-periments as a positive control. Interestingly, the extent of CCL20secretion elicited by hypoxia exceeded that triggered by stimula-tion with LPS at all the time points analyzed (Fig. 5C), suggestingthat hypoxia is a more potent stimulus than LPS in inducingCCL20 production by Mn.

Taken together, these results indicate that primary human Mnisolated from different donors produce variable baseline levels ofCCL20 but respond to low pO2 with consistent CCL20 up-regu-lation, confirming CCL20 as a hypoxia-inducible gene.

DiscussionMn responses that ensue following recruitment at pathologicalsites begin in the setting of reduced pO2 (1). Hypoxia regulatoryeffects on Mn gene expression have not been characterized in de-tail, and only a limited number of hypoxia-inducible genes havebeen identified in these cells (17, 31, 41). In this study, we providethe first transcriptome analysis of hypoxic primary peripheralblood human Mn.

Microarray analysis was conducted on RNA pooled from dif-ferent donor-derived Mn, whose response to hypoxia was demon-strated by VEGF up-regulation. Three pools, each composed byRNA from five different donors, were investigated per each ex-perimental condition. RNA pooling is a powerful, cost-effective,

and statistically valid mean of identifying common changes in agene expression profile. Previous studies have established the fea-sibility of using this type of approach to overcome interindividualvariability and provide reliable results that reflect gene expressionin individual donors (42). The suitability of our experimental pro-tocol for identifying bona fide hypoxia-responsive genes in pri-mary cells was inferred by the demonstration that hypoxia modu-lated 74 genes known from the literature to be responsive tohypoxia. The majority of these genes, involved in glycolysis, ap-optosis, cell cycle, and transcription, may constitute a gene clustercommonly induced by hypoxia in cells of different lineages. Thegeneral representativeness of the gene expression changes detectedby microarray was further established by qRT-PCR, which con-firmed the differential expression of 27 genes randomly selectedfrom the microarray profile in a fourth set of pooled RNAs.

Two recent reports investigated the hypoxia transcriptome ofhMDM and demonstrated induction of genes encoding angiogen-esis and ECM regulators/components (20, 21), some of whichwere also present in our profile. Expression of ADM, F3, FLT1,FN1, MIF, INHBA, MMP-1, PTGS2, SPP1, and VEGF, amongothers, was detectable at significant levels in both primary humanMn (this report) and MDM (20), and exposure to hypoxia resultedin their strong up-regulation in both cell populations. Moreover,we demonstrated up-regulation of other genes previously shown tobe inducible by hypoxia in mononuclear phagocytes and codingfor immunoregulatory/proinflammatory proteins and transcriptionfactors, such as ARG1, CXCR4, EGR1, FGF1, ID2, TIMP-1, andVLDR (17, 21, 32, 41, 43, 44). Interestingly, IL-1�, IL-6, andTNF-�, induction in Mn was observed in response to hypoxiaalone, whereas up-regulation in Mf and MDM required LPS co-stimulation or a reoxygenation period (43). Furthermore, we con-firmed in primary Mn hypoxic inhibition of a cluster of inflam-matory genes found down-regulated in mMf and hMDM, such asCCL2 (16), CCR5 (18), CTSC (21), OASE (40), and RAB7 (21).These findings indicate that hypoxia is active on different types ofmononuclear phagocytes and across species in regulating the ex-pression of selected target genes, which may be critical for theiradaptation to the hypoxic environment and ability to function in itand could be representative of the hypoxic transcriptome of cellsbelonging to the monocytic lineage. In contrast, the observationthat other genes up-regulated in hMDM (20, 21) were not inducedor were even down-regulated in primary Mn, e.g., the angiogenesisinducers endothelial cell growth factor-1 (ECGF1) and IL-18 bind-ing protein (IL-18BP), suggests that hypoxia may also activate inmononuclear phagocytes a specific transcriptional response de-pending on their differentiation stage.

Several HMGs identified in our analysis were endowed withtranscription regulatory activity and/or encoded important compo-nents of the HIF-1 transcription pathway. Hypoxia increased theexpression of P4HA1, P4HA2, and EGLN1, three members of theprolyl-hydroxylase family which mediate HIF-� hydroxylation inwell-oxygenated cells targeting the protein for proteosomal deg-radation by the von Hippel Lindau tumor suppressor protein(pVHL) (15), and of BHLHB2, a bHLH family member implicatedin the regulation of pVHL/HIF pathways (15). These data are inagreement with previous findings in other cell types (15, 35, 36)and suggest an O2-driven HIF-1-dependent autoregulatory mech-anism, required to ensure fast HIF-1� degradation upon reoxygen-ation, also in Mn. Of note is the novel evidence that hypoxia pro-motes the expression of ATF2 and ATF5, two members of the ATFfamily of transcription factors (45) required for efficient activationof gene transcription by hypoxia (15). Moreover, we demonstratedup-regulation of FosB and FRA-2, two components of the AP-1complex (45), which is activated by hypoxia and was recently

FIGURE 4. Up-regulation of CCL20 mRNA expression by hypoxia inhuman Mn. Total RNA was isolated from human peripheral blood Mncultured under normoxic () or hypoxic (�) conditions for 16 h. A, RNAfrom six different donor-derived Mn was reverse transcribed and tested forCCL20 and �-actin mRNA expression by semiquantitative RT-PCR. B,The extent of CCL20 mRNA up-regulation by hypoxia was determined byqRT-PCR analysis on the RNA isolated from the indicated donors, andrelative transcript expression was calculated in relation to the values ob-tained in parallel for three reference genes, as detailed in Materials andMethods. Results from one representative experiment are expressed as foldincrease of mRNA levels in hypoxic relative to normoxic cells (arbitrarilydefined as 1) and are the mean of triplicate determinations. Fold-changevalues are indicated by a number associated with each bar.

1951The Journal of Immunology

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 13: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

shown to cooperate with HIF-1� in the transactivation of hypoxiainducible genes (46), and of JEM-1, a novel transcriptional cofac-tor which enhances AP-1 activity (47). Up-regulation of thesegenes together with down-regulation of ZnF197, a novel VHL-interacting protein that functions as a repressor of HIF-1� trans-activation (48), may represent a positive regulatory mechanism ofhypoxia transcriptional response in Mn. Down-regulation of sev-eral HIF-1 transcriptional cofactors, including CBP, C/EBP�,MITF, and NCOA1/SRC-1, was also observed in hypoxic Mn. Allthese proteins physically interact with HIF-1� enhancing its trans-activation function under hypoxia (9, 15, 49), and their down-regulation suggests the existence of a negative feedback mecha-nism to control Mn hypoxic response. Consistent with thishypothesis, we observed a parallel decrease of the mRNAs encod-ing HIF-1� and one of its dimerization partners, ARNT-2 (9)

GO data mining characterized a significant cluster of genes asbeing associated with immune regulation and inflammatory re-sponses, chemotaxis, cell adhesion, and ECM remodeling, the ma-jority of which has not been previously identified as responsive tohypoxia. Profound changes were observed in the expression ofscavenger receptors. Of relevance is the up-regulation of MSR1and SCARF1, because these molecules functions as endocytic re-

ceptors for acetylated LDL and may thus be implicated in lipid-loaded foam cell formation contributing to atherosclerotic plaquesdevelopment (50). Consistent with the view that hypoxia may exerta pathogenetic role in atherosclerosis and Alzheimer’s disease (9,10, 41) is also the down-regulation of CD163 and STAB1 scaven-ger receptors, which are endowed with atheroprotective activity(51, 52), and of various genes involved in the regulation of fattyacid and/or cholesterol biosynthesis/transport and acting as anti-atherogenic factors, e.g., LDLR, ApoE, and CYP27A1 (53, 54). Mnhypoxic profile also showed up-regulation of a number of otherpattern recognition receptors critical to host defense, the most rel-evant of which are the C1qR1, COLEC12, and MARCO. Thesemolecules bind specific Ags on bacteria facilitating their recogni-tion and phagocytosis (55–57). Interestingly, the TLR familymembers TLR5 and TLR7, whose function is also to recognizepathogen or their products and hence initiate innate immune re-sponses (58), were down-regulated by hypoxia. Differential mod-ulation of receptors with similar functions is intriguing and islikely to contribute to the fine tuning of Mn antimicrobial activitiesat sites of infection.

Myeloid cell immune functions are regulated by a balance ofinhibitory and activating signals transduced by multiple families of

FIGURE 5. Hypoxia induces CCL20 production by primary Mn. Human peripheral blood Mn purified from the indicated donors were cultured undernormoxic () or hypoxic conditions (�) for 16 h (A and B) or for different time points (C), in the presence or absence of 100 ng/ml LPS. A, CCL20 proteinexpression was evaluated on cytospin preparations by immunocytochemical analysis using a specific mAb. Staining was conducted with the Envision�

System as described in the Materials and Methods. Hematoxylin-counterstained slides were examined under a phase-contrast microscope and photomi-crographed (magnification, �40). CCL20 immunoreactivity is detectable in the cell cytoplasm (brown staining). Staining with an isotype-matched controlmAb (IgG1) is shown for donor 7. B and C, Supernatants were harvested and assayed for secreted CCL20 by ELISA. Results from one experiment,representative of three performed, in which each sample was tested in triplicate, are expressed as picogram per 1 � 106 cells/ml. The number associatedwith each bar indicates the fold induction of CCL20 secretion in hypoxic relative to normoxic cells (arbitrarily defined as equal to 1).

1952 TRANSCRIPTIONAL PROFILE OF HYPOXIC MONOCYTES

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 14: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

cell surface IRS receptors belonging to the Ig superfamily of in-hibitory/stimulatory pairs of molecules (59). These receptors areeither characterized by ITIM/ITAM regulatory components intheir cytoplasmic domain or pair with ITAM-containing trans-membrane adapter proteins (59). Mn exposure to hypoxia resultedin the selective modulation of various members of these receptorfamilies. Of particular interest is the up-regulation of Fc�RIIA,Band down-regulation of Fc�RIA. Fc�RIIA contains a functionalITAM motif, thus triggering cell activation, whereas Fc�RIIB en-codes an ITIM component, leading to repression of cellular re-sponses (60). Fc�RIA, which associates for signaling with anITAM-containing �-chain, also belongs to the activatory Fc�Rsubclass (60). Fc�Rs involvement in various autoimmune inflam-matory states was reported (60), and our findings suggest the po-tential role of hypoxia in influencing the pathogenesis of thesediseases through the modulation of distinct Fc�R genes.

Hypoxic modulation of other ITIM/ITAM Ig family members,specifically up-regulation of FCAR, CMRF-35H, and TREM-1 anddown-regulation of LIR9 and LAIR1, was also observed. Thesemolecules mediate a variety of effector functions vital to the ad-aptative immune response, and their ligation can have both proin-flammatory and immunosuppressive consequences by differen-tially modulating the secretion of pro- or anti-inflammatorymediators (59–63). The differential expression of inhibitory andactivating isoforms of a given receptor family with similar speci-ficities on the same cell is another example of the tight regulatoryrole of hypoxia on Mn inflammatory responses. In particular, theconcomitant down-regulation of the Ig-like inhibitory receptorsLILRB1–4 and of their ligand HLA-G, a nonclassical inhibitoryHLA class I Ag, is noteworthy given the role of these molecules inimmune tolerance and immune escape (64). It is conceivable thatinhibition of these molecules may decrease the activation thresholdof Mn retained at hypoxic sites. Mn activation under conditions ofreduced O2 availability may also be controlled by the selectivedown- and up-regulation of two C-type lectin receptors, the ITIM-containing inhibitory molecule CLECSF6 (65) and CD69, a mem-ber of the NK receptor family and a potent inducer of Mn inflam-matory mediator production and cytotoxic activity (66). Althougha ligand for CD69 has not been identified, previous studies sug-gested a pathogenetic role for CD69 in certain inflammatory statescharacterized by hypoxia, such as rheumatoid arthritis, chronic in-flammatory liver diseases, and asthma (66).

Mononuclear phagocyte migratory activity is a highly regulatedprocess which depends on a defined repertoire of chemokines/re-ceptors and adhesion molecules, and dysregulated expression ofthese proteins may alter their recruitment and activation (67). Var-ious studies have investigated the mechanisms whereby mononu-clear phagocytes are retained/concentrated at hypoxic pathologicalsites (1). One possibility is that hypoxia inhibits their migration inresponse to chemokines by decreasing the expression of specificchemokine receptors, as demonstrated for CCR5 in Mf (18). Inagreement with this hypothesis and with previous findings show-ing impaired Mn migratory ability to CCL2 under conditions oflow pO2 (1), we observed CCR5, CCR1, and CCR2 down-regula-tion. This study also suggests other potential mechanisms for Mnentrapment in hypoxic areas, such as the up-regulation of RGS1, amember of a new class of G protein-signaling deactivators whichinactivates several chemotactic receptors inhibiting chemoattrac-tant-induced Mn migration (68). Furthermore, repression of thesecreted cell motility-promoting factor, ENPP2 (69), together withinduction of the antichemotactic cytokine, MIF (13), and the GROfamily chemokines, CXCL2 and CXCL3, which are specializedMn-arrest chemokines (70), may also provide a “stop” signal toMn within hypoxic tissues.

The complexity of the regulation of Mn migratory behavior byhypoxia is further emphasized by the demonstration of dysregu-lated expression of several other migration-related genes. Mn hy-poxic profile was associated with down-regulation of the adhesionmolecules CD11C, CD57, and ITGB5,7, which mediate Mn adhe-sion to the endothelium and/or to the ECM (67), and with up-regulation of both the fractalkine receptor, which binds to theCX3C chemokine fractalkine expressed on endothelial cells func-tioning as a potent adhesion molecule (71), and the RDC1 receptor,which share with CXCR4 the chemokine CXCL12/SDF-1 as anatural ligand (72). A critical role in the control of mononuclearphagocyte motility is also exerted by MMPs, a group of secretedenzymes that trigger ECM degradation facilitating leukocytemovement in tissues (73). Recent reports have shown up-regula-tion by hypoxia of MMP-1, MMP-7, and MMP12 in hMDM (20,21). Interestingly, only MMP1 was induced also in primary Mnthat showed specific up-regulation of MMP16 and MMP19 anddown-regulation of MMP25. Moreover, the MMP inhibitors,TIMP1 and TFPI2, which reduce ECM degradation inhibiting cellmigratory activity (73, 74), were also modulated in hypoxic Mn.Collectively, these studies indicate that regulation of MMPs andtheir inhibitors is a common denominator of mononuclear phago-cyte response to hypoxia, but that different components of thesefamilies are controlled depending on the cell differentiation stage.Altered expression of MMPs has been associated with a variety ofacute and chronic inflammatory states (73), and hypoxia can prob-ably play a role in the pathogenesis of these diseases by modulat-ing MMP production by infiltrating Mn

Various cytokines/chemokine and/or receptors are modulated byhypoxia in mononuclear phagocytes (1, 43). The Mn hypoxic tran-scriptome confirmed and extended those findings showing differ-ential expression of other components of the cytokine/chemokinesystem. Various members of the IL-1 and the TNFR/ligand super-families were selectively up- or down-regulated in hypoxic Mn,including molecules associated with and mediating signal trans-duction from their receptors (e.g., IL1RAP, IRAK3, and the TNFR-associated factor, TRAF). Because of their pleiotropic effects onalmost every types of cells, coordinated regulation of the TNF andIL-1 systems is likely to represent an important mechanism tocontrol the amplitude and the duration of inflammatory responses.The demonstration that hypoxia induces CSF1 and CSF3, whileinhibiting their receptors, is noteworthy given the role of thesefactors in the regulation of myeloid cell production, differentiation,and function (75) and is consistent with previous findings suggest-ing a reciprocal and divergent action of hypoxia on receptor vsligand expression (16, 18). Accordingly, IL-4 up-regulation andconcomitant inhibition of its receptor IL-13RA1 was observed.This interplay is likely to serve as a negative feedback mechanismto control the autocrine activation of producing Mn. However, notall the data presented in this study are consistent with this scenario.Concomitant down-regulation of the Mn chemoattractants CCL2and CCL8 and of their receptors CCR2 and CCR5 (2) was in factobserved in response to hypoxia. Similarly, we found down-reg-ulation of CCL15, a chemoattractant for neutrophils, monocytes,and lymphocytes (76), and CCL23, a chemokine mediating restingT cell and Mn chemotaxis (77), and of their common receptorCCR1. Collectively, these data indicate that a dynamic change inchemokine/receptor expression profile occurs in Mn within hy-poxic tissues. This tight and complex level of control exerted bylow O2 tension is clearly of pathophysiologic relevance, represent-ing an important mechanism of regulation of leukocyte traffickingand function at sites of inflammation. Because some of the mod-ulated chemokines have angiogenic activity (1, 2, 76), their altered

1953The Journal of Immunology

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 15: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

expression under low pO2 may also influence neoangiogenesis inpathological tissues.

One of the most important findings of this study was the dem-onstration that hypoxia strongly induced the expression of theCCL20-encoding gene in primary Mn. Marked differences in thebasal mRNA expression were detected among individual donors.However, mRNA up-regulation under hypoxia was consistentlydemonstrated in all Mn preparation analyzed, and gene expressionresults were associated to a parallel augmentation of protein ex-pression and secretion. Previous reports have shown that theCCL20 gene is markedly up-regulated in PBMC by LPS and in-ducible in other cell types in response to various mediators ofinflammation, including cytokines, growth factors, bacterial, viraland plant products, whereas it is poorly expressed in the absenceof inflammatory stimuli (3). This study is the first to identify hyp-oxia as a new CCL20 inducer. Interestingly, the extent of CCL20up-regulation by hypoxia exceeded that triggered by LPS, suggest-ing that hypoxia is a more potent stimulus than LPS for CCL20production by Mn. Given its role in the recruitment of iDC, effec-tor/memory T lymphocytes, and naive B cells, CCL20 has beenproposed as an important mediator for both the initiation and ef-fector phases of the inflammatory reactions, linking innate andacquired immunity (3). Hence, by producing CCL20, Mn may con-trol the kinetics and composition of the cellular infiltrate undervarious inflammatory conditions and at tumor sites. The identifi-cation of CCL20 as a hypoxia-inducible gene may explain, in part,the high levels of this chemokine present in areas of inflammationand in various chronic inflammatory conditions, such as rheuma-toid arthritis, inflammatory skin disorders, and tumors (for a re-view, see Refs. 3 and 78), as these sites are known to be hypoxicand infiltrated by Mn, and is indicative of a pathogenetic role forthis molecule in these diseases. Further studies are ongoing in thelaboratory to elucidate the molecular mechanisms underlyingCCL20 induction by hypoxia.

In summary, we have described the hypoxia transcriptome ofprimary human Mn and identified a large number of genes notpreviously known to change as a result of reduced O2 concentra-tions. Our findings contribute to the definition of the gene clustercommonly induced by hypoxia in cells of different lineage. Thisstudy provides novel insights into the molecular responses to thehypoxic stress and the mechanisms linking low pO2 to the regu-lation of immune and inflammatory responses, leading to new per-spectives of the role of hypoxia in programming Mn functionswithin pathological conditions and identifying potential moleculartargets for the development of rational therapeutic approaches.

AcknowledgmentsWe thank S. Delfino for technical assistance and C. Dabizzi for secretarialwork.

DisclosuresThe authors have no financial conflict of interest.

References1. Murdoch, C., A. Giannoudis, and C. E. Lewis. 2004. Mechanisms regulating the

recruitment of macrophages into hypoxic areas of tumors and other ischemictissues. Blood 104: 2224–2234.

2. Rossi, D., and A. Zlotnik. 2000. The biology of chemokines and their receptors.Annu. Rev. Immunol. 18: 217–242.

3. Schutyser, E., S. Struyf, and J. Van Damme. 2003. The CC chemokine CCL20and its receptor CCR6. Cytokine Growth Factor Rev. 14: 409–426.

4. Varesio, L., I. Espinoza-Delgado, G. Gusella, G. Cox, G. Melillo, T. Musso, andM. C. Bosco. 1995. Role of cytokines in the activation of monocytes. In HumanCytokines: Their Role in Disease and Therapy. B. B. Aggarwal and R. K. Puri,eds. Blackwell Scientific Publication, Cambridge, pp. 55–70.

5. Paulnock, D. M., K. P. Demick, and S. P. Coller. 2000. Analysis of interferon-�-dependent and-independent pathways of macrophage activation. J. LeukocyteBiol. 67: 677–682.

6. Kopydlowski, K. M., C. A. Salkowski, M. J. Cody, N. Van Rooijen, J. Major,T. A. Hamilton, and S. N. Vogel. 1999. Regulation of macrophage chemokineexpression by lipopolysaccharide in vitro and in vivo. J. Immunol. 163:1537–1544.

7. Bosco, M. C., A. Rapisarda, S. Massazza, G. Melillo, H. Young, and L. Varesio.2000. The tryptophan catabolite picolinic acid selectively induces the chemokinesmacrophage-inflammatory protein-1� and -1� in macrophages. J. Immunol. 164:3283–3291.

8. Crowther, M., N. J. Brown, E. T. Bishop, and C. E. Lewis. 2001. Microenviron-mental influence on macrophage regulation of angiogenesis in wounds and ma-lignant tumors. J. Leukocyte Biol. 70: 478–490.

9. Semenza, G. L. 2001. Hypoxia-inducible factor 1: oxygen homeostasis and dis-ease pathophysiology. Trends Mol. Med. 7: 345–350.

10. Bjornheden, T., M. Levin, M. Evaldsson, and O. Wiklund. 1999. Evidence ofhypoxic areas within the arterial wall in vivo. Arterioscler. Thromb. Vasc. Biol.19: 870–876.

11. Distler, J. H., R. H. Wenger, M. Gassmann, M. Kurowska, A. Hirth, S. Gay, andO. Distler. 2004. Physiologic responses to hypoxia and implications for hypoxia-inducible factors in the pathogenesis of rheumatoid arthritis. Arthritis. Rheum. 50:10–23.

12. Vaupel, P., and M. Hockel. 2003. Tumor oxygenation and its relevance to tumorphysiology and treatment. Adv. Exp. Med. Biol. 510: 45–49.

13. Harris, A. L. 2002. Hypoxia—a key regulatory factor in tumour growth. Nat. Rev.Cancer 2: 38–47.

14. Le, Q. T., N. C. Denko, and A. J. Giaccia. 2004. Hypoxic gene expression andmetastasis. Cancer Metastasis Rev. 23: 293–310.

15. Wenger, R. H., D. P. Stiehl, and G. Camenisch. 2005. Integration of oxygensignaling at the consensus HRE. Sci. STKE 2005: re12.

16. Bosco, M. C., M. Puppo, S. Pastorino, Z. Mi, G. Melillo, S. Massazza,A. Rapisarda, and L. Varesio. 2004. Hypoxia selectively inhibits monocyte che-moattractant protein-1 production by macrophages. J. Immunol. 172: 1681–1690.

17. Schioppa, T., B. Uranchimeg, A. Saccani, S. K. Biswas, A. Doni, A. Rapisarda,S. Bernasconi, S. Saccani, M. Nebuloni, L. Vago, et al. 2003. Regulation of thechemokine receptor CXCR4 by hypoxia. J. Exp. Med. 198: 1391–1402.

18. Bosco, M. C., G. Reffo, M. Puppo, and L. Varesio. 2004. Hypoxia inhibits theexpression of the CCR5 chemokine receptor in macrophages. Cell. Immunol.228: 1–7.

19. Lockhart, D. J., and E. A. Winzeler. 2000. Genomics, gene expression and DNAarrays. Nature 405: 827–836.

20. White, J. R., R. A. Harris, S. R. Lee, M. H. Craigon, K. Binley, T. Price,G. L. Beard, C. R. Mundy, and S. Naylor. 2004. Genetic amplification of thetranscriptional response to hypoxia as a novel means of identifying regulators ofangiogenesis. Genomics 83: 1–8.

21. Burke, B., A. Giannoudis, K. P. Corke, D. Gill, M. Wells, L. Ziegler-Heitbrock,and C. E. Lewis. 2003. Hypoxia-induced gene expression in human macrophages:implications for ischemic tissues and hypoxia-regulated gene therapy.Am. J. Pathol. 163: 1233–1243.

22. Bosco, M. C., I. Espinoza-Delgado, T. K. Rowe, M. G. Malabarba, L. Long, andL. Varesio. 1997. Functional role for the myieloid differentiation antigen CD14in the activation of human monocytes by IL-2. J. Immunol. 159: 2922–2931.

23. Irizarry, R. A., B. M. Bolstad, F. Collin, L. M. Cope, B. Hobbs, and T. P. Speed.2003. Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res.31: e15.

24. Quackenbush, J. 2002. Microarray data normalization and transformation. Nat.Genet. 32(Suppl.): 496–501.

25. Ashburner, M., C. A. Ball, J. A. Blake, D. Botstein, H. Butler, J. M. Cherry,A. P. Davis, K. Dolinski, S. S. Dwight, J. T. Eppig, et al. 2000. Gene ontology:tool for the unification of biology. The Gene Ontology Consortium. Nat. Genet.25: 25–29.

26. Hosack, D. A., G. Dennis, Jr., B. T. Sherman, H. C. Lane, and R. A. Lempicki.2003. Identifying biological themes within lists of genes with EASE. GenomeBiol. 4: R70.

27. Dennis, G., Jr., B. T. Sherman, D. A. Hosack, J. Yang, W. Gao, H. C. Lane, andR. A. Lempicki. 2003. DAVID: database for annotation, visualization, and inte-grated discovery. Genome Biol. 4: 3.

28. Rozen, R., and H. J. Skaletsky. 2000. Primer 3 on the WWW for general usersand for biological programmers. In Bioinformatics Methods and Protocols: Meth-ods in Molecular Biology. S. Krawets and S. Misener, eds. Humana, Totowa,p. 365.

29. Pfaffl, M. W., A. Tichopad, C. Prgomet, and T. P. Neuvians. 2004. Determinationof stable housekeeping genes, differentially regulated target genes and sampleintegrity: BestKeeper–Excel-based tool using pair-wise correlations. Biotechnol.Lett. 26: 509–515.

30. Muller, P. Y., H. Janovjak, A. R. Miserez, and Z. Dobbie. 2002. Processing ofgene expression data generated by quantitative real-time RT-PCR. BioTechniques32: 1372–1379.

31. Melillo, G., E. A. Sausville, K. Cloud, T. Lahusen, L. Varesio, andA. M. Senderovicz. 1999. Flavopiridol, a protein kinase inhibitor, down-regulateshypoxic induction of vascular endothelial growth factor expression in humanmonocytes. Cancer Res. 59: 5433–5437.

32. Louis, C. A., J. S. Reichner, W. L. Henry, Jr., B. Mastrofrancesco, T. Gotoh,M. Mori, and J. E. Albina. 1998. Distinct arginase isoforms expressed in primaryand transformed macrophages: regulation by oxygen tension. Am. J. Physiol. 274:R775–R782.

33. Jogi, A., J. Vallon-Christersson, L. Holmquist, H. Axelson, A. Borg, andS. Pahlman. 2004. Human neuroblastoma cells exposed to hypoxia: induction of

1954 TRANSCRIPTIONAL PROFILE OF HYPOXIC MONOCYTES

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from

Page 16: Hypoxia Modifies the Transcriptome of Primary Human Monocytes ...

genes associated with growth, survival, and aggressive behavior. Exp. Cell Res.295: 469–487.

34. Denko, N., C. Schindler, A. Koong, K. Laderoute, C. Green, and A. Giaccia.2000. Epigenetic regulation of gene expression in cervical cancer cells by thetumor microenvironment. Clin. Cancer Res. 6: 480–487.

35. del Peso, L., M. C. Castellanos, E. Temes, S. Martin-Puig, Y. Cuevas, G. Olmos,and M. O. Landazuri. 2003. The von Hippel Lindau/hypoxia-inducible factor(HIF) pathway regulates the transcription of the HIF-proline hydroxylase genesin response to low oxygen. J. Biol. Chem. 278: 48690–48695.

36. Hofbauer, K. H., B. Gess, C. Lohaus, H. E. Meyer, D. Katschinski, and A. Kurtz.2003. Oxygen tension regulates the expression of a group of procollagen hy-droxylases. Eur. J. Biochem. 270: 4515–4522.

37. Kitamuro, T., K. Takahashi, K. Ogawa, R. Udono-Fujimori, K. Takeda,K. Furujama, M. Nakayama, J. Sun, H. Fujita, W. Hida, et al. 2003. Bach 1functions as a hypoxia-inducible repressor for the heme oxygenase-1 gene inhuman cells. J. Biol. Chem. 278: 9125–9133.

38. Naldini, A., F. Carraro, S. Silvestri, and V. Bocci. 1997. Hypoxia affects cytokineproduction and proliferative responses by human peripheral mononuclear cells.J. Cell. Physiol. 173: 335–342.

39. Naldini, A., A. Pucci, and F. Carraro. 2001. Hypoxia induces the expression andrelease of interleukin 1 receptor antagonist in mitogen-activated mononuclearcells. Cytokine 13: 334–341.

40. Carta, L., S. Pastorino, G. Melillo, M. C. Bosco, S. Massazza, and L. Varesio.2001. Engineering of macrophages to produce IFN-� in response to hypoxia.J. Immunol. 166: 5374–5380.

41. Nakazato, K., T. Ishibashi, K. Nagata, Y. Seino, Y. Wada, T. Sakamoto,R. Matsuoka, T. Teramoto, M. Sekimata, Y. Homma, and Y. Maruyama. 2001.Expression of very low density lipoprotein receptor mRNA in circulating humanmonocytes: its up-regulation by hypoxia. Atherosclerosis 155: 439–444.

42. Peng, X., C. L. Wood, E. M. Blalock, K. C. Chen, P. W. Landfield, andA. J. Stromberg. 2003. Statistical implications of pooling RNA samples for mi-croarray experiments. BMC Bioinformatics 4: 26.

43. Lewis, J. S., J. A. Lee, J. C. E. Underwood, A. L. Harris, and C. E. Lewis. 1999.Macrophage responses to hypoxia: relevance to disease mechanisms. J. Leuko-cyte Biol. 66: 889–900.

44. Qu, X., M. X. Yang, B. H. Kong, L. Qi, Q. L. Lam, S. Yan, P. Li, M. Zhang, andL. Lu. 2005. Hypoxia inhibits the migratory capacity of human monocyte-deriveddendritic cells. Immunol. Cell Biol. 83: 668–673.

45. Herdegen, T., and J. D. Leah. 1998. Inducible and constitutive transcription fac-tors in the mammalian nervous system: control of gene expression by Jun, Fosand Krox, and CREB/ATF proteins. Brain Res. Brain Res. Rev. 28: 370–490.

46. Laderoute, K. R., J. M. Calaoagan, C. Gustafson-Brown, A. M. Knapp, G. C. Li,H. L. Mendonca, H. E. Ryan, Z. Wang, and R. S. Johnson. 2002. The responseof c-jun/AP-1 to chronic hypoxia is hypoxia-inducible factor 1 � dependent. Mol.Cell. Biol. 22: 2515–2523.

47. Tong, J. H., E. Duprez, and M. Lanotte. 1999. JEM-1, a novel nuclear co-factor:localisation and functional interaction with AP-1. Leukemia. 13: 1982–1992.

48. Li, Z., D. Wang, X. Na, S. R. Schoen, E. M. Messing, and G. Wu. 2003. The VHLprotein recruits a novel KRAB-A domain protein to repress HIF-1� transcrip-tional activity. EMBO J. 22: 1857–1867.

49. Busca, R., E. Berra, C. Gaggioli, M. Khaled, K. Bille, B. Marchetti, R. Thyss,G. Fitsialos, L. Larribere, C. Bertolotto, et al. 2005. Hypoxia-inducible factor 1�is a new target of microphthalmia-associated transcription factor (MITF) in mel-anoma cells. J. Cell Biol. 170: 49–59.

50. Fukuhara-Takaki, K., M. Sakai, Y. Sakamoto, M. Takeya, and S. Horiuchi. 2005.Expression of class A scavenger receptor is enhanced by high glucose in vitro andunder diabetic conditions in vivo: one mechanism for an increased rate of ath-erosclerosis in diabetes. J. Biol. Chem. 280: 3355–3364.

51. Moestrup, S. K., and H. J. Moller. 2004. CD163: a regulated hemoglobin scav-enger receptor with a role in the anti-inflammatory response. Ann. Med. 36:347–354.

52. Prevo, R., S. Banerji, J. Ni, and D. G. Jackson. 2004. Rapid plasma membrane-endosomal trafficking of the lymph node sinus and high endothelial venule scav-enger receptor/homing receptor stabilin-1 (FEEL-1/CLEVER-1). J. Biol. Chem.279: 52580–52592.

53. Greenow, K., N. J. Pearce, and D. P. Ramji. 2005. The key role of apolipoproteinE in atherosclerosis. J. Mol. Med. 83: 329–342.

54. Vainio, S., and E. Ikonen. 2003. Macrophage cholesterol transport: a criticalplayer in foam cell formation. Ann. Med. 35: 146–155.

55. Arredouani, M. S., A. Palecanda, H. Koziel, Y. C. Huang, A. Imrich,T. H. Sulahian, Y. Y. Ning, Z. Yang, T. Pikkarainen, M. Sankala, et al. 2005.MARCO is the major binding receptor for unopsonized particles and bacteria onhuman alveolar macrophages. J. Immunol. 175: 6058–6064.

56. Nakamura, K., H. Funakoshi, K. Miyamoto, F. Tokunaga, and T. Nakamura.2001. Molecular cloning and functional characterization of a human scavengerreceptor with C-type lectin (SRCL), a novel member of a scavenger receptorfamily. Biochem. Biophys. Res. Commun. 280: 1028–1035.

57. Geertsma, M. F., P. H. Nibbering, H. P. Haagsman, M. R. Daha, and R. van Furth.1994. Binding of surfactant protein A to C1q receptors mediates phagocytosis ofStaphylococcus aureus by monocytes. Am. J. Physiol. 267: L578–L584.

58. Pasare, C., and R. Medzhitov. 2005. Toll-like receptors: linking innate and adap-tive immunity. Adv. Exp. Med. Biol. 560: 11–18.

59. Clark, G. J., B. Cooper, S. Fitzpatrick, B. J. Green, and D. N. Hart. 2001. Thegene encoding the immunoregulatory signaling molecule CMRF-35A localized tohuman chromosome 17 in close proximity to other members of the CMRF-35family. Tissue Antigens 57: 415–423.

60. Gerber, J. S., and D. M. Mosser. 2001. Stimulatory and inhibitory signals orig-inating from the macrophage Fc� receptors. Microbes Infect. 3: 131–139.

61. Meyaard, L., J. Hurenkamp, H. Clevers, L. L. Lanier, and J. H. Phillips. 1999.Leukocyte-associated Ig-like receptor-1 functions as an inhibitory receptor oncytotoxic T cells. J. Immunol. 162: 5800–5804.

62. Dietrich, J., H. Nakajima, and M. Colonna. 2000. Human inhibitory and activat-ing Ig-like receptors which modulate the function of myeloid cells. MicrobesInfect. 2: 323–329.

63. Bleharski, J. R., V. Kiessler, C. Buonsanti, P. A. Sieling, S. Stenger, M. Colonna,and R. L. Modlin. 2003. A role for triggering receptor expressed on myeloidcells-1 in host defense during the early-induced and adaptive phases of the im-mune response. J. Immunol. 170: 3812–3818.

64. LeMaoult, J., K. Zafaranloo, C. Le Danff, and E. D. Carosella. 2005. HLA-Gup-regulates ILT2, ILT3, ILT4, and KIR2DL4 in antigen presenting cells, NKcells, and T cells. FASEB J. 19: 662–664.

65. Bates, E. E., N. Fournier, E. Garcia, J. Valladeau, I. Durand, J. J. Pin,S. M. Zurawski, S. Patel, J. S. Abrams, S. Lebecque, et al. 1999. APCs expressDCIR, a novel C-type lectin surface receptor containing an immunoreceptor ty-rosine-based inhibitory motif. J. Immunol. 163: 1973–1983.

66. De Maria, R., M. G. Cifone, R. Trotta, M. R. Rippo, C. Festuccia, A. Santoni, andR. Testi. 1994. Triggering of human monocyte activation through CD69, a mem-ber of the natural killer cell gene complex family of signal transducing receptors.J. Exp. Med. 180: 1999–2004.

67. Imhof, B. A., and M. Aurrand-Lions. 2004. Adhesion mechanisms regulating themigration of monocytes. Nat. Rev. Immunol. 4: 432–444.

68. Denecke, B., A. Meyerdierks, and E. C. Bottger. 1999. RGS1 is expressed inmonocytes and acts as a GTPase-activating protein for G-protein-coupled che-moattractant receptors. J. Biol. Chem. 274: 26860–26868.

69. Black, E. J., T. Clair, J. Delrow, P. Neiman, and D. A. Gillespie. 2004. Microar-ray analysis identifies Autotaxin, a tumour cell motility and angiogenic factorwith lysophospholipase D activity, as a specific target of cell transformation byv-Jun. Oncogene 23: 2357–2366.

70. Smith, D. F., E. Galkina, K. Ley, and Y. Huo. 2005. GRO family chemokines arespecialized for monocyte arrest from flow. Am. J. Physiol. 289: H1976–H1984.

71. Umehara, H., S. Goda, T. Imai, Y. Nagano, Y. Minami, Y. Tanaka, T. Okazaki,E. T. Bloom, and N. Domae. 2001. Fractalkine, a CX3C-chemokine, functionspredominantly as an adhesion molecule in monocytic cell line THP-1. Immunol.Cell Biol. 79: 298–302.

72. Balabanian, K., B. Lagane, S. Infantino, K. Y. Chow, J. Harriague, B. Moepps,F. Arenzana-Seisdedos, M. Thelen, and F. Bachelerie. 2005. The chemokineSDF-1/CXCL12 binds to and signals through the orphan receptor RDC1 in Tlymphocytes. J. Biol. Chem. 280: 35760–35766.

73. Shapiro, S. D., and R. M. Senior. 1999. Matrix metalloproteinases: matrix deg-radation and more. Am. J. Respir. Cell Mol. Biol. 20: 1100–1102.

74. Herman, M. P., G. K. Sukhova, W. Kisiel, D. Foster, M. R. Kehry, P. Libby, andU. Schonbeck. 2001. Tissue factor pathway inhibitor-2 is a novel inhibitor ofmatrix metalloproteinases with implications for atherosclerosis. J. Clin. Invest.107: 1117–1126.

75. Pimentel, E. 1990. Colony-stimulating factors. Ann. Clin. Lab. Sci. 20: 36–55.76. Hwang, J., C. W. Kim, K. N. Son, K. Y. Han, K. H. Lee, H. K. Kleinman, J. Ko,

D. S. Na, B. S. Kwon, Y. S. Gho, and J. Kim. 2004. Angiogenic activity of humanCC chemokine CCL15 in vitro and in vivo. FEBS Lett. 570: 47–51.

77. Forssmann, U., M. B. Delgado, M. Uguccioni, P. Loetscher, G. Garotta, andM. Baggiolini. 1997. CK�8, a novel CC chemokine that predominantly acts onmonocytes. FEBS Lett. 408: 211–216.

78. Ruth, J. H., S. Shahrara, C. C. Park, J. C. Morel, P. Kumar, S. Qin, andA. E. Koch. 2003. Role of macrophage inflammatory protein-3� and its ligandCCR6 in rheumatoid arthritis. Lab. Invest. 83: 579–588.

79. Nikitenko, L. L., D. M. Smith, R. Bicknell, and M. C. Rees. 2003. Transcriptionalregulation of the CRLR gene in human microvascular endothelial cells by hyp-oxia. FASEB J. 17: 1499–1501.

80. Eltzschig, H. K., J. C. Ibla, G. T. Furuta, M. O. Leonard, K. A. Jacobson,K. Enjyoji, S. C. Robson, and S. P. Colgan. 2003. Coordinated adenine nucleotidephosphohydrolysis and nucleoside signaling in posthypoxic endothelium: role ofectonucleotidases and adenosine A2B receptors. J. Exp. Med. 198: 783–796.

81. Louis, N. A., K. E. Hamilton, T. Kong, and S. P. Colgan. 2005. HIF-dependentinduction of apical CD55 coordinates epithelial clearance of neutrophils. FASEBJ. 19: 950–959.

82. Cosio, G., M. C. Jeziorski, F. Lopez-Barrera, G. M. De La Escalera, andC. Clapp. 2003. Hypoxia inhibits expression of prolactin and secretion of ca-thepsin-D by the GH4C1 pituitary adenoma cell line. Lab. Invest. 83: 1627–1636.

83. May, D., A. Itin, O. Gal, H. Kalinski, E. Feinstein, and E. Keshet. 2005. Ero1-L�plays a key role in a HIF-1-mediated pathway to improve disulfide bond forma-tion and VEGF secretion under hypoxia: implication for cancer. Oncogene 24:1011–1020.

1955The Journal of Immunology

by guest on March 13, 2018

http://ww

w.jim

munol.org/

Dow

nloaded from