Dissecting interferon-induced transcriptional programs in human peripheral blood cells

Simon J Waddell, Stephen J Popper, Kathleen H Rubins, Michael J Griffiths, Patrick O Brown, Michael Levin, David A Relman, Simon J Waddell, Stephen J Popper, Kathleen H Rubins, Michael J Griffiths, Patrick O Brown, Michael Levin, David A Relman

Abstract

Interferons are key modulators of the immune system, and are central to the control of many diseases. The response of immune cells to stimuli in complex populations is the product of direct and indirect effects, and of homotypic and heterotypic cell interactions. Dissecting the global transcriptional profiles of immune cell populations may provide insights into this regulatory interplay. The host transcriptional response may also be useful in discriminating between disease states, and in understanding pathophysiology. The transcriptional programs of cell populations in health therefore provide a paradigm for deconvoluting disease-associated gene expression profiles.We used human cDNA microarrays to (1) compare the gene expression programs in human peripheral blood mononuclear cells (PBMCs) elicited by 6 major mediators of the immune response: interferons alpha, beta, omega and gamma, IL12 and TNFalpha; and (2) characterize the transcriptional responses of purified immune cell populations (CD4+ and CD8+ T cells, B cells, NK cells and monocytes) to IFNgamma stimulation. We defined a highly stereotyped response to type I interferons, while responses to IFNgamma and IL12 were largely restricted to a subset of type I interferon-inducible genes. TNFalpha stimulation resulted in a distinct pattern of gene expression. Cell type-specific transcriptional programs were identified, highlighting the pronounced response of monocytes to IFNgamma, and emergent properties associated with IFN-mediated activation of mixed cell populations. This information provides a detailed view of cellular activation by immune mediators, and contributes an interpretive framework for the definition of host immune responses in a variety of disease settings.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. PBMC transcriptional programs elicited by…
Figure 1. PBMC transcriptional programs elicited by cytokine exposure.
The PBMC response to stimulation with 0.6 pM IFNα, β, ω, γ, IL12 and TNFα from 30 minutes to 24 h after treatment. 1857 genes were identified to be significantly differentially expressed in response to one or more stimuli compared to mock-treated PBMC (0.1% BSA/PBS). The expression profiles are ordered by hierarchical clustering; the genes are displayed as rows, time points/conditions as columns, in temporal order (see yellow key). Red coloring signifies the up-regulation of expression; blue coloring denotes down-regulation. Samples were taken at time intervals of 0.5, 1, 4, 8, 12 and 24 h. The column marked SAM indicates (in red) which genes were identified to be significantly differentially expressed in response to each cytokine.
Figure 2. Induction of IFNγ in PBMCs…
Figure 2. Induction of IFNγ in PBMCs after IL12 exposure.
The mean expression profile of IFNγ transcripts after exposure of PBMCs to 0.6 pM IFNα, β, ω, and γ, IL12 and TNFα; as determined by microarray analysis at time intervals of 0.5, 1, 4, 8, 12 and 24 h post stimulation. Standard deviations are indicated with error bars.
Figure 3. Cell-type associated gene expression.
Figure 3. Cell-type associated gene expression.
A (left). Cell type-associated genes identified from the transcriptional profiles of unstimulated isolated cell types (time zero and 0.1% BSA/PBS-treated time series). Genes were defined as cell type-associated if they were identified as significantly more highly expressed in a single cell type compared to all other cell types. The expression profiles are ordered by hierarchical clustering; the genes are displayed as rows, cell type/time points as columns. Red coloring signifies high expression; blue coloring denotes low expression. Cell type-associated gene clusters are marked. B (right). The cell type-specific nature of IFNγ transcriptional responses. Purified subsets of B cells, CD4+ T cells, CD8+ T cells, NK cells and monocytes were stimulated with 0.6 pM IFNγ and sampled at 0.5, 1, 4, 8, 12 and 24 h. 807 significantly induced genes were identified, and ordered by cell type and hierarchical clustering. The genes are displayed as rows, time points/cell subsets as columns. Red coloring signifies up-regulation, and blue coloring signifies down-regulation of expression after IFNγ exposure relative to the mock-treated discrete cell population. The column marked SAM indicates (in red) which genes were significantly differentially expressed by IFNγ in each cell type. Genes of interest are marked as annotated in Source .
Figure 4. Monocyte responses to IFNγ.
Figure 4. Monocyte responses to IFNγ.
A (top). The temporal transcriptional responses of monocytes to IFNγ stimulation. Of 363 genes that were significantly differentially expressed following treatment with 0.6 pM IFNγ, 264 genes were assigned to 7 significantly represented expression profiles, A–G. The temporal response (measured in mean fold change) of the genes assigned to each cluster is plotted at 0.5, 1, 4, 8, 12 and 24 h post treatment. Clusters A (square), B (triangle) and C (diamond) colored red; D (triangle) and E (square) in green; F (square) in purple; and G (square) in blue. B (bottom). A network of predicted crosstalk downstream of IFNγ exposure. Previously characterized interactions between IFNγ and the 363 genes identified as differentially expressed after IFNγ treatment in monocytes were mapped. The expression of 109 genes were predicted to be directly affected by IFNγ. Secondary regulatory events were characterized by the expression of 65 additional genes (identified to be differentially expressed by IFNγ treatment) whose expression was modified by those 109 genes but not by IFNγ directly. This network illustrates the complexity of possible interplay downstream of IFNγ stimulation. In addition, 61 proteins reported to have an effect on IFNγ expression itself are highlighted in yellow, and may represent positive and negative feedback loops mediating cell activation. Gene identifiers are colored by temporal expression pattern after IFNγ treatment as detailed in Figure 4A (dark red (profiles A–C), lighter red (D and E), pink (F), blue (G), grey (unassigned)). Gene identifiers are also shaped by putative function, transcription factors (ellipse), kinase/phosphatases (triangle), ligands (rhombus), receptors (cross). The nature of the interactions are indicated with connecting lines either reflecting a positive (green), negative (blue), or undefined (grey) impact on downstream gene or protein expression.
Figure 5. The transcriptional plasticity of immune…
Figure 5. The transcriptional plasticity of immune cellular responses to IFNγ stimulation.
79 genes associated with a single isolated immune cell subtype (without stimulation) were induced after IFNγ treatment in other cell types. Genes defined as cell type-associated (by comparing unstimulated time series, as detailed in Figure 3A) are marked on the left. The top panel detailing B cell associated genes, followed by T cell, NK cell, and monocyte associated genes illustrated in the bottom panel. The differential regulation of these genes after IFNγ treatment in each cell type (as in Figure 3B) is described on the right. The expression profiles are ordered by hierarchical clustering; the genes are displayed as rows, cell type/time points as columns. Red coloring in the cell-type associated panels (on left) signifies highly expressed in a single cell type compared to all other cell types; blue coloring denotes low expression. Red coloring in the IFNγ responsive panels (on right) signifies up-regulation, and blue coloring signifies down-regulation of expression after IFNγ exposure relative to the mock-treated discrete cell population. B (B cells); T4 (CD4+ T cells); T8 (CD8+ T cells); NK (NK cells); M (monocytes). The column marked SAM details (in red) which genes were significantly differentially expressed by IFNγ in each cell type. Genes are marked as annotated in Source .

References

    1. Isaacs A, Lindenmann J. Virus interference. I. The interferon. Proc R Soc Lond B Biol Sci. 1957;147:258–267.
    1. Bach EA, Aguet M, Schreiber RD. The IFN gamma receptor: a paradigm for cytokine receptor signaling. Annu Rev Immunol. 1997;15:563–591.
    1. Kotenko SV, Gallagher G, Baurin VV, Lewis-Antes A, Shen M, et al. IFN-lambdas mediate antiviral protection through a distinct class II cytokine receptor complex. Nat Immunol. 2003;4:69–77.
    1. Sheppard P, Kindsvogel W, Xu W, Henderson K, Schlutsmeyer S, et al. IL-28, IL-29 and their class II cytokine receptor IL-28R. Nat Immunol. 2003;4:63–68.
    1. Pestka S, Krause CD, Walter MR. Interferons, interferon-like cytokines, and their receptors. Immunol Rev. 2004;202:8–32.
    1. van den Broek MF, Muller U, Huang S, Aguet M, Zinkernagel RM. Antiviral defense in mice lacking both alpha/beta and gamma interferon receptors. J Virol. 1995;69:4792–4796.
    1. Stark GR, Kerr IM, Williams BR, Silverman RH, Schreiber RD. How cells respond to interferons. Annu Rev Biochem. 1998;67:227–264.
    1. Sadler AJ, Williams BR. Interferon-inducible antiviral effectors. Nat Rev Immunol. 2008;8:559–568.
    1. Ortaldo JR, Herberman RB, Harvey C, Osheroff P, Pan YC, et al. A species of human alpha interferon that lacks the ability to boost human natural killer activity. Proc Natl Acad Sci U S A. 1984;81:4926–4929.
    1. Hilkens CM, Schlaak JF, Kerr IM. Differential responses to IFN-alpha subtypes in human T cells and dendritic cells. J Immunol. 2003;171:5255–5263.
    1. Jaitin DA, Roisman LC, Jaks E, Gavutis M, Piehler J, et al. Inquiring into the differential action of interferons (IFNs): an IFN-alpha2 mutant with enhanced affinity to IFNAR1 is functionally similar to IFN-beta. Mol Cell Biol. 2006;26:1888–1897.
    1. Takaoka A, Yanai H. Interferon signalling network in innate defence. Cell Microbiol. 2006;8:907–922.
    1. Baig E, Fish EN. Distinct signature type I interferon responses are determined by the infecting virus and the target cell. Antivir Ther. 2008;13:409–422.
    1. Boehm U, Klamp T, Groot M, Howard JC. Cellular responses to interferon-gamma. Annu Rev Immunol. 1997;15:749–795.
    1. Newport MJ, Huxley CM, Huston S, Hawrylowicz CM, Oostra BA, et al. A mutation in the interferon-gamma-receptor gene and susceptibility to mycobacterial infection. N Engl J Med. 1996;335:1941–1949.
    1. Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nat Rev Immunol. 2006;6:836–848.
    1. Schiepers OJ, Wichers MC, Maes M. Cytokines and major depression. Prog Neuropsychopharmacol Biol Psychiatry. 2005;29:201–217.
    1. Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling [see comments]. Nature. 2000;403:503–11.
    1. Lock C, Hermans G, Pedotti R, Brendolan A, Schadt E, et al. Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis. Nat Med. 2002;8:500–508.
    1. Griffiths MJ, Shafi MJ, Popper SJ, Hemingway CA, Kortok MM, et al. Genomewide analysis of the host response to malaria in Kenyan children. J Infect Dis. 2005;191:1599–1611.
    1. Popper SJ, Shimizu C, Shike H, Kanegaye JT, Newburger JW, et al. Gene-expression patterns reveal underlying biological processes in Kawasaki disease. Genome Biol. 2007;8:R261.
    1. Ramilo O, Allman W, Chung W, Mejias A, Ardura M, et al. Gene expression patterns in blood leukocytes discriminate patients with acute infections. Blood. 2007;109:2066–2077.
    1. Jacobsen M, Repsilber D, Gutschmidt A, Neher A, Feldmann K, et al. Candidate biomarkers for discrimination between infection and disease caused by Mycobacterium tuberculosis. J Mol Med. 2007;85:613–621.
    1. Watson MA, Ylagan LR, Trinkaus KM, Gillanders WE, Naughton MJ, et al. Isolation and molecular profiling of bone marrow micrometastases identifies TWIST1 as a marker of early tumor relapse in breast cancer patients. Clin Cancer Res. 2007;13:5001–5009.
    1. Rubins KH, Hensley LE, Jahrling PB, Whitney AR, Geisbert TW, et al. The host response to smallpox: analysis of the gene expression program in peripheral blood cells in a nonhuman primate model. Proc Natl Acad Sci U S A. 2004;101:15190–15195.
    1. Simmons CP, Popper S, Dolocek C, Chau TN, Griffiths M, et al. Patterns of host genome-wide gene transcript abundance in the peripheral blood of patients with acute dengue hemorrhagic fever. J Infect Dis. 2007;195:1097–1107.
    1. Zimmerer JM, Lesinski GB, Ruppert AS, Radmacher MD, Noble C, et al. Gene expression profiling reveals similarities between the in vitro and in vivo responses of immune effector cells to IFN-alpha. Clin Cancer Res. 2008;14:5900–5906.
    1. Jenner RG, Young RA. Insights into host responses against pathogens from transcriptional profiling. Nat Rev Microbiol. 2005;3:281–294.
    1. Der SD, Zhou A, Williams BR, Silverman RH. Identification of genes differentially regulated by interferon alpha, beta, or gamma using oligonucleotide arrays. Proc Natl Acad Sci U S A. 1998;95:15623–15628.
    1. Dolken L, Ruzsics Z, Radle B, Friedel CC, Zimmer R, et al. High-resolution gene expression profiling for simultaneous kinetic parameter analysis of RNA synthesis and decay. RNA. 2008;14:1959–1972.
    1. Sana TR, Janatpour MJ, Sathe M, McEvoy LM, McClanahan TK. Microarray analysis of primary endothelial cells challenged with different inflammatory and immune cytokines. Cytokine. 2005;29:256–269.
    1. Hartman SE, Bertone P, Nath AK, Royce TE, Gerstein M, et al. Global changes in STAT target selection and transcription regulation upon interferon treatments. Genes Dev. 2005;19:2953–2968.
    1. Darnell JE, Jr, Kerr IM, Stark GR. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science. 1994;264:1415–1421.
    1. Platanias LC. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol. 2005;5:375–386.
    1. Brinkmann V, Geiger T, Alkan S, Heusser CH. Interferon alpha increases the frequency of interferon gamma-producing human CD4+ T cells. J Exp Med. 1993;178:1655–1663.
    1. de Jong R, Altare F, Haagen IA, Elferink DG, Boer T, et al. Severe mycobacterial and Salmonella infections in interleukin-12 receptor-deficient patients. Science. 1998;280:1435–1438.
    1. Wajant H, Pfizenmaier K, Scheurich P. Tumor necrosis factor signaling. Cell Death Differ. 2003;10:45–65.
    1. Kempe S, Kestler H, Lasar A, Wirth T. NF-kappaB controls the global pro-inflammatory response in endothelial cells: evidence for the regulation of a pro-atherogenic program. Nucleic Acids Res. 2005;33:5308–5319.
    1. Palmer C, Diehn M, Alizadeh AA, Brown PO. Cell-type specific gene expression profiles of leukocytes in human peripheral blood. BMC Genomics. 2006;7:115.
    1. Holling TM, Schooten E, van den Elsen PJ. Function and regulation of MHC class II molecules in T-lymphocytes: of mice and men. Hum Immunol. 2004;65:282–290.
    1. Ramsey SA, Klemm SL, Zak DE, Kennedy KA, Thorsson V, et al. Uncovering a macrophage transcriptional program by integrating evidence from motif scanning and expression dynamics. PLoS Comput Biol. 2008;4:e1000021.
    1. Ernst J, Bar-Joseph Z. STEM: a tool for the analysis of short time series gene expression data. BMC Bioinformatics. 2006;7:191.
    1. Vasanwala FH, Kusam S, Toney LM, Dent AL. Repression of AP-1 function: a mechanism for the regulation of Blimp-1 expression and B lymphocyte differentiation by the B cell lymphoma-6 protooncogene. J Immunol. 2002;169:1922–1929.
    1. Dimitriou ID, Clemenza L, Scotter AJ, Chen G, Guerra FM, et al. Putting out the fire: coordinated suppression of the innate and adaptive immune systems by SOCS1 and SOCS3 proteins. Immunol Rev. 2008;224:265–283.
    1. Ram PA, Waxman DJ. Role of the cytokine-inducible SH2 protein CIS in desensitization of STAT5b signaling by continuous growth hormone. J Biol Chem. 2000;275:39487–39496.
    1. Chae HD, Lee KE, Williams DA, Gu Y. Cross-talk between RhoH and Rac1 in regulation of actin cytoskeleton and chemotaxis of hematopoietic progenitor cells. Blood. 2008;111:2597–2605.
    1. Daryadel A, Yousefi S, Troi D, Schmid I, Schmidt-Mende J, et al. RhoH/TTF negatively regulates leukotriene production in neutrophils. J Immunol. 2009;182:6527–6532.
    1. Ogilvie RL, Sternjohn JR, Rattenbacher B, Vlasova IA, Williams DA, et al. Tristetraprolin Mediates Interferon-{gamma} mRNA Decay. J Biol Chem. 2009;284:11216–11223.
    1. Kovarik P, Sauer I, Schaljo B. Molecular mechanisms of the anti-inflammatory functions of interferons. Immunobiology. 2007;212:895–901.
    1. LeibundGut-Landmann S, Waldburger JM, Krawczyk M, Otten LA, Suter T, et al. Mini-review: Specificity and expression of CIITA, the master regulator of MHC class II genes. Eur J Immunol. 2004;34:1513–1525.
    1. Rubins KH, Hensley LE, Wahl-Jensen V, Daddario DiCaprio KM, Young HA, et al. The temporal program of peripheral blood gene expression in the response of nonhuman primates to Ebola hemorrhagic fever. Genome Biol. 2007;8:R174.
    1. Pathan N, Hemingway CA, Alizadeh AA, Stephens AC, Boldrick JC, et al. Role of interleukin 6 in myocardial dysfunction of meningococcal septic shock. Lancet. 2004;363:203–209.
    1. Popper SJ, Watson VE, Shimizu C, Kanegaye JT, Burns JC, et al. Gene transcript abundance profiles distinguish Kawasaki disease from adenovirus infection. J Infect Dis. 2009;200:657–666.
    1. Schlaak JF, Hilkens CM, Costa-Pereira AP, Strobl B, Aberger F, et al. Cell-type and donor-specific transcriptional responses to interferon-alpha. Use of customized gene arrays. J Biol Chem. 2002;277:49428–49437.
    1. Whitney AR, Diehn M, Popper SJ, Alizadeh AA, Boldrick JC, et al. Individuality and variation in gene expression patterns in human blood. Proc Natl Acad Sci U S A. 2003;100:1896–1901.
    1. Boldrick JC, Alizadeh AA, Diehn M, Dudoit S, Liu CL, et al. Stereotyped and specific gene expression programs in human innate immune responses to bacteria. Proc Natl Acad Sci U S A. 2002;99:972–977.
    1. Thompson LJ, Dunstan SJ, Dolecek C, Perkins T, House D, et al. Transcriptional response in the peripheral blood of patients infected with Salmonella enterica serovar Typhi. Proc Natl Acad Sci U S A. 2009;106:22433–22438.
    1. Aberger F, Costa-Pereira AP, Schlaak JF, Williams TM, O'Shaughnessy RF, et al. Analysis of gene expression using high-density and IFN-gamma-specific low-density cDNA arrays. Genomics. 2001;77:50–57.
    1. Indraccolo S, Pfeffer U, Minuzzo S, Esposito G, Roni V, et al. Identification of genes selectively regulated by IFNs in endothelial cells. J Immunol. 2007;178:1122–1135.
    1. Kalveram B, Schmidtke G, Groettrup M. The ubiquitin-like modifier FAT10 interacts with HDAC6 and localizes to aggresomes under proteasome inhibition. J Cell Sci. 2008;121:4079–4088.
    1. Lukasiak S, Schiller C, Oehlschlaeger P, Schmidtke G, Krause P, et al. Proinflammatory cytokines cause FAT10 upregulation in cancers of liver and colon. Oncogene. 2008;27:6068–6074.
    1. Radomska HS, Huettner CS, Zhang P, Cheng T, Scadden DT, et al. CCAAT/enhancer binding protein alpha is a regulatory switch sufficient for induction of granulocytic development from bipotential myeloid progenitors. Mol Cell Biol. 1998;18:4301–4314.
    1. Nerlov C. C/EBPalpha mutations in acute myeloid leukaemias. Nat Rev Cancer. 2004;4:394–400.
    1. Dimas AS, Deutsch S, Stranger BE, Montgomery SB, Borel C, et al. Common regulatory variation impacts gene expression in a cell type-dependent manner. Science. 2009;325:1246–1250.
    1. Stanford Microarray Database. Available: .
    1. Edgar R, Domrachev M, Lash AE. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30:207–210.
    1. Gene Expression Omnibus Accession Number GSE17762. Available: .
    1. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A. 2001;98:5116–5121.
    1. Eisen MB, Spellman PT, Brown PO, Botstein D. Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci U S A. 1998;95:14863–8.
    1. Dennis G, Jr, Sherman BT, Hosack DA, Yang J, Gao W, et al. DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 2003;4:3.
    1. Huang dW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4:44–57.
    1. Heinemeyer T, Wingender E, Reuter I, Hermjakob H, Kel AE, et al. Databases on transcriptional regulation: TRANSFAC, TRRD and COMPEL. Nucleic Acids Res. 1998;26:362–367.
    1. Diehn M, Sherlock G, Binkley G, Jin H, Matese JC, et al. SOURCE: a unified genomic resource of functional annotations, ontologies, and gene expression data. Nucleic Acids Res. 2003;31:219–223.

Source: PubMed

3
Suscribir