RNA-Seq analysis of chikungunya virus infection and identification of granzyme A as a major promoter of arthritic inflammation

Jane A C Wilson, Natalie A Prow, Wayne A Schroder, Jonathan J Ellis, Helen E Cumming, Linden J Gearing, Yee Suan Poo, Adam Taylor, Paul J Hertzog, Francesca Di Giallonardo, Linda Hueston, Roger Le Grand, Bing Tang, Thuy T Le, Joy Gardner, Suresh Mahalingam, Pierre Roques, Phillip I Bird, Andreas Suhrbier, Jane A C Wilson, Natalie A Prow, Wayne A Schroder, Jonathan J Ellis, Helen E Cumming, Linden J Gearing, Yee Suan Poo, Adam Taylor, Paul J Hertzog, Francesca Di Giallonardo, Linda Hueston, Roger Le Grand, Bing Tang, Thuy T Le, Joy Gardner, Suresh Mahalingam, Pierre Roques, Phillip I Bird, Andreas Suhrbier

Abstract

Chikungunya virus (CHIKV) is an arthritogenic alphavirus causing epidemics of acute and chronic arthritic disease. Herein we describe a comprehensive RNA-Seq analysis of feet and lymph nodes at peak viraemia (day 2 post infection), acute arthritis (day 7) and chronic disease (day 30) in the CHIKV adult wild-type mouse model. Genes previously shown to be up-regulated in CHIKV patients were also up-regulated in the mouse model. CHIKV sequence information was also obtained with up to ≈8% of the reads mapping to the viral genome; however, no adaptive viral genome changes were apparent. Although day 2, 7 and 30 represent distinct stages of infection and disease, there was a pronounced overlap in up-regulated host genes and pathways. Type I interferon response genes (IRGs) represented up to ≈50% of up-regulated genes, even after loss of type I interferon induction on days 7 and 30. Bioinformatic analyses suggested a number of interferon response factors were primarily responsible for maintaining type I IRG induction. A group of genes prominent in the RNA-Seq analysis and hitherto unexplored in viral arthropathies were granzymes A, B and K. Granzyme A-/- and to a lesser extent granzyme K-/-, but not granzyme B-/-, mice showed a pronounced reduction in foot swelling and arthritis, with analysis of granzyme A-/- mice showing no reductions in viral loads but reduced NK and T cell infiltrates post CHIKV infection. Treatment with Serpinb6b, a granzyme A inhibitor, also reduced arthritic inflammation in wild-type mice. In non-human primates circulating granzyme A levels were elevated after CHIKV infection, with the increase correlating with viral load. Elevated granzyme A levels were also seen in a small cohort of human CHIKV patients. Taken together these results suggest granzyme A is an important driver of arthritic inflammation and a potential target for therapy.

Trial registration: ClinicalTrials.gov NCT00281294.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Global analysis of gene expression.
Fig 1. Global analysis of gene expression.
(A) Venn diagram [70] of up-regulated genes in feet. Only genes with q2 and FPKM >1 were included. Numbers in parenthesis represent the mean fold change of all the genes in the segment; note genes which are shared between all time points show the highest mean fold changes. (B) Heat map of−log10 z scores from Ingenuity upstream regulator analysis of the genes shown in A. Scores are ranked highest to lowest (mean of the 3 time points). The top and bottom 10 upstream regulators are shown on the figure; the full data set is shown in S3 Table. (C) Venn diagram of down-regulated genes in feet. Numbers in parenthesis represent the mean fold change. (D) Heat map of p values from Ingenuity canonical pathway analysis of the genes shown in C, with day 7 pathways ranked most to least significant. A DAVID analysis of the same genes (bar chart) showing similarity scores. The full data sets for these figures are provided in S3 Table. (E) Venn diagram of up-regulated genes in lymph nodes on days 2 and 7. (F) Venn diagram of up-regulated genes on day 2 in lymph nodes (LN) and feet (Ft); and heat map of−log10 z scores from Ingenuity upstream regulator analysis of these genes. The first column of the heat map shows z scores from day 2 lymph nodes ranked highest to lowest (the full data set is provided in S3 Table). (G) Venn diagram of up-regulated genes in lymph nodes on days 7 and feet on day 7. (H) Venn diagram of down-regulated genes in lymph nodes on days 2 and 7.
Fig 2. CHIKV genome.
Fig 2. CHIKV genome.
(A) The percentage of all the reads that aligned to the mouse genome for each tissue and time point. Read alignment numbers are provided in S1D Fig. (B) Examples of alignments of RNA-Seq reads from 3 foot samples mapped to the CHIKV genome (map quality threshold 20) viewed using Integrated Genomics Viewer (IGV version 2.3.34). The CHIKV genome is shown at the top for reference (arrow represents position of the sub-genomic promoter). Upper graphs (dark grey) show sequence coverage (log scale) for each nucleotide position in the CHIKV genome with y axis scale ranges (e.g. 0–300,000) shown in the left hand corners. The bottom graphs are “squished” views of the 100 bp reads aligned to the CHIKV genome (each grey horizontal bar represents one read). Black spots represent deletions/insertions within each read.
Fig 3. The interferon signature.
Fig 3. The interferon signature.
(A) Expression of IFNα6 and IFNβ as determined by qRT-PCR in different tissues on day 2 post CHIKV infection (n = 3 mice), the time of the peak IFNα/β response [28]. Values are normalized to RPL13a mRNA levels and expressed as fold induction relative to mock infected controls (n = 3). CHIKV titres in the tissues were determined as described [28]. Spearman’s correlation tests showed a significant relationship between IFN mRNA levels and viral titres (IFNα6 Spearman’s rho = 0.829 p = 0.042, IFNβ Spearman’s rho = 0.943 p = 0.005). (B) For all samples the up- and down-regulated DEGs (for which FC>2, q1, see S1 Table) were analyzed using Interferome and the percentage of these DEGs that are interferon regulated genes (IRGs) is shown. (Interferome does not distinguish between genes directly or indirectly stimulated by IFNs, and some type I and/or II IRGs may not be identified by Interferome). (C) Heat map of FPKM values for all IFN genes identified by the RNA-Seq analysis (the same data is plotted as bar chart in S4 Fig). (D) Transcription factors associated with IFN responses in feet. Fold change of indicated transcription factors with vertical numbers representing the mean FKPM values (for the 3 biological replicates). Horizontal bold numbers represent the activation Z scores for the indicated transcription factor as determined by the direct function of the upstream regulator analysis of IPA; corresponding p values are provided in S5 Fig. (E) CiiiDER analysis of putative transcription factor site enrichment in the up-regulated type II IRGs in feet (as identified by Interferome). Color and size of circles reflect p values of the enrichment. Calculations for x and y values and the input/output data for the labeled transcription factors are provided in S7 Fig.
Fig 4. Granzyme gene expression and CHIKV…
Fig 4. Granzyme gene expression and CHIKV infection in granzyme deficient mice.
(A) Volcano plots of host gene expression from the RNA-Seq analysis of feet and lymph nodes (LN) at the indicated day post infection. Only genes where FPKM ≥1 in at least one sample in the pair wise comparisons were included in the plots. Positions of the granzyme (Gzm) genes are indicated by arrows, with values in parenthesis representing FPKM values. (B) Foot swelling in granzyme deficient mice. Mice were infected as above and foot swelling monitored. GzmA-/- mice; * p<0.03, ** p<0.001 (Kolmogorov-Smirnov tests, n = 8–10 mice per group). GzmK-/- mice; *p<0.031 (Mann Whitney U tests, mean of two independent experiments is shown, n = 12–14 per group). (C) Viraemia in granzyme deficient mice. Granzyme deficient mice were infected with CHIKV and the viraema measure on the indicated days. No significant differences in viraema were apparent: GzmA-/- vs C57BL/6 controls (n = 6 mice per group); GzmB-/- vs C57BL/6 controls (n = 8–10 mice per group); GzmK-/- vs C57BL/6 controls (n = 12–14 mice per group). (D) Tissue CHIKV titers in GzmA-/- and C57BL/6 mice. Feet; n = 6–12 GzmA-/- and n = 12–20 C57BL/6 feet per time point; data obtained from 2 independent experiments. Muscle; n = 3–6 mice per time point.
Fig 5. Histology and immunohistochemistry of GzmA…
Fig 5. Histology and immunohistochemistry of GzmA-/- mice feet.
(A) H&E staining of feet on day 6 post infection in C57BL/6 and GzmA-/- mice. Cellular infiltrates are characterized by high densities of blue staining nuclei; areas with pronounced cellular infiltrates are indicated by white oval outlines. (B) Aperio Positive Pixel Count determination of the ratio of blue (nuclear) to red (cytoplasmic) staining areas in whole foot sections day 6 post infection. Leukocytes tend to have a higher nuclear/cytoplasmic area ratio, so elevated ratios reflect increased leukocyte infiltrates [13]; (n = 6 feet from 6 mice per group, 3 sections per foot; statistics by 2 way ANOVA including a term for section). (C) Immunohistochemical staining for NK cells (anti-CD335/NKp46) clearly visible (brown staining) in muscle tissue of feet from CHIKV-infected C57BL/6 mice 6 days post infection (left). NK cell staining was less pronounced in GzmA-/- mice (right). Blue counter staining with haematoxylin. (D) Aperio Positive Pixel Count determination of NK cell staining; strong brown pixels per μm2 in whole feet sections (3 sections per foot; n = 11–12 feet from 11–12 mice per group from 2 independent experiments. Statistics by Mann Whitney U test). (E) As for C for T cell (anti-CD3) staining. (F) As for D for T cell staining. Statistics by Kolmogrov Smirnov test. (G) As for C for monocyte/macrophage (F4/80) staining, which was prominent in subcutaneous tissues (* indicates epidermis). (H) As for D for monocyte/macrophage staining.
Fig 6. Granzyme A inhibitor and granzyme…
Fig 6. Granzyme A inhibitor and granzyme levels in CHIKV infected primates.
(A) On days 2 to 6 post infection with CHIKV, C57BL/6 mice were treated i.v. with 10 μg of the granzyme inhibitor, Serpinb6b, or Trypsinized (inactivated) GzmA inhibitor (green arrows) or were left untreated. The data represents results from 2 independent experiments: GzmA inhibitor (n = 14 feet, 7 mice); Trypsinized GzmA inhibitor (n = 6 feet, 3 mice); untreated mice (n = 30 feet, 15 mice). Statistics by t test; p values provided for comparisons between granzyme A inhibitor and Trypsinized granzyme A inhibitor and are only provided where comparisons between granzyme A inhibitor and untreated mice were also significant. (B) Viraemia for the same mice as shown in A. (C) Aperio Positive Pixel Count determination of the ratio of blue (nuclear) to red (cytoplasmic) pixels in H&E stained whole foot sections day 6 post infection (as in Fig 5B). (n = 6 feet from 3 mice per group, 3 sections per foot; statistics by Kolmogorov Smirnov test). (D) Granzyme A levels in plasma samples from CHIKV-infected non-human primates (NHPs) measured using an ELISA kit. Data for 9 NHPs is shown; all NHPs had samples collected day -1, one day prior to CHIKV infection. (E) Granzyme A levels on day -1 and peak granzyme A levels plotted for the 9 NHPs. Statistics by paired t test. (F) Correlation between peak viral load (S9D Fig) and the increase in granzyme A levels from day -1 to peak (i.e. peak levels minus day -1 levels). Statistics by Spearman correlation. (G) Serum granzyme A levels in healthy controls and IgM positive symptomatic CHIKV patients. Granzyme A levels were determined using cytokine bead array and FACs. The limit of detection is deemed to be 3.7 pg/ml. Statistics by Kruskal-Wallis test.

References

    1. Suhrbier A, Jaffar-Bandjee MC, Gasque P. Arthritogenic alphaviruses—an overview. Nat Rev Rheumatol. 2012;8: 420–429. 10.1038/nrrheum.2012.64
    1. Weaver SC, Lecuit M. Chikungunya virus and the global spread of a mosquito-borne disease. N Engl J Med. 2015;372: 1231–1239. 10.1056/NEJMra1406035
    1. Nunes MR, Faria NR, de Vasconcelos JM, Golding N, Kraemer MU, de Oliveira LF, et al. Emergence and potential for spread of chikungunya virus in Brazil. BMC Med. 2015;13: 102 10.1186/s12916-015-0348-x
    1. Simon F, Javelle E, Cabie A, Bouquillard E, Troisgros O, Gentile G, et al. French guidelines for the management of chikungunya (acute and persistent presentations). November 2014. Med Mal Infect. 2015;45: 243–263. 10.1016/j.medmal.2015.05.007
    1. Padmakumara B, Jayana JB, Menona RMR, Krishnankuttyd B, Payippallilc R, Nishab RS. Comparative evaluation of four therapeutic regimes in chikungunya arthritis: a prospective randomized parallel-group study. Indian J Rheumatol 2009;4: 94–101.
    1. Ramsauer K, Schwameis M, Firbas C, Mullner M, Putnak RJ, Thomas SJ, et al. Immunogenicity, safety, and tolerability of a recombinant measles-virus-based chikungunya vaccine: a randomised, double-blind, placebo-controlled, active-comparator, first-in-man trial. Lancet Infect Dis. 2015;15: 519–527. 10.1016/S1473-3099(15)70043-5
    1. Chang LJ, Dowd KA, Mendoza FH, Saunders JG, Sitar S, Plummer SH, et al. Safety and tolerability of chikungunya virus-like particle vaccine in healthy adults: a phase 1 dose-escalation trial. Lancet. 2014;384: 2046–2052. 10.1016/S0140-6736(14)61185-5
    1. Ryman KD, Klimstra WB. Host responses to alphavirus infection. Immunol Rev. 2008;225: 27–45. 10.1111/j.1600-065X.2008.00670.x
    1. Rudd PA, Wilson J, Gardner J, Larcher T, Babarit C, Le TT, et al. Interferon response factors 3 and 7 protect against chikungunya virus hemorrhagic fever and shock. J Virol. 2012;86: 9888–9898. 10.1128/JVI.00956-12
    1. Schilte C, Couderc T, Chretien F, Sourisseau M, Gangneux N, Guivel-Benhassine F, et al. Type I IFN controls chikungunya virus via its action on nonhematopoietic cells. J Exp Med. 2010;207: 429–442. 10.1084/jem.20090851
    1. Couderc T, Chretien F, Schilte C, Disson O, Brigitte M, Guivel-Benhassine F, et al. A mouse model for chikungunya: young age and inefficient type-I interferon signaling are risk factors for severe disease. PLoS Pathog. 2008;4: e29 10.1371/journal.ppat.0040029
    1. Yoon IK, Alera MT, Lago CB, Tac-An IA, Villa D, Fernandez S, et al. High rate of subclinical chikungunya virus infection and association of neutralizing antibody with protection in a prospective cohort in the Philippines. PLoS Negl Trop Dis. 2015;9: e0003764 10.1371/journal.pntd.0003764
    1. Poo YS, Rudd PA, Gardner J, Wilson JA, Larcher T, Colle MA, et al. Multiple immune factors are involved in controlling acute and chronic chikungunya virus infection. PLoS Negl Trop Dis. 2014;8: e3354 10.1371/journal.pntd.0003354
    1. Fox JM, Long F, Edeling MA, Lin H, van Duijl-Richter MK, Fong RH, et al. Broadly neutralizing alphavirus antibodies bind an epitope on E2 and inhibit entry and egress. Cell. 2015;163: 1095–1107. 10.1016/j.cell.2015.10.050
    1. Lum FM, Teo TH, Lee WW, Kam YW, Renia L, Ng LF. An essential role of antibodies in the control of Chikungunya virus infection. J Immunol. 2013;190: 6295–6302. 10.4049/jimmunol.1300304
    1. Teng TS, Kam YW, Lee B, Hapuarachchi HC, Wimal A, Ng LC, et al. A systematic meta-analysis of immune signatures in patients with acute chikungunya virus infection. J Infect Dis. 2015;211: 1925–1935. 10.1093/infdis/jiv049
    1. Ng LF, Chow A, Sun YJ, Kwek DJ, Lim PL, Dimatatac F, et al. IL-1beta, IL-6, and RANTES as biomarkers of chikungunya severity. PLoS ONE. 2009;4: e4261 10.1371/journal.pone.0004261
    1. Chow A, Her Z, Ong EK, Chen JM, Dimatatac F, Kwek DJ, et al. Persistent arthralgia induced by chikungunya virus infection is associated with interleukin-6 and granulocyte macrophage colony-stimulating factor. J Infect Dis. 2011;203: 149–157. 10.1093/infdis/jiq042
    1. Wauquier N, Becquart P, Nkoghe D, Padilla C, Ndjoyi-Mbiguino A, Leroy EM. The acute phase of Chikungunya virus infection in humans is associated with strong innate immunity and T CD8 cell activation. J Infect Dis. 2011;204: 115–123. 10.1093/infdis/jiq006
    1. Hoarau JJ, Jaffar Bandjee MC, Krejbich Trotot P, Das T, Li-Pat-Yuen G, Dassa B, et al. Persistent chronic inflammation and infection by chikungunya arthritogenic alphavirus in spite of a robust host immune response. J Immunol. 2010;184: 5914–5927. 10.4049/jimmunol.0900255
    1. Chopra A, Saluja M, Venugopalan A. Effectiveness of chloroquine and inflammatory cytokine response in patients with early persistent musculoskeletal pain and arthritis following chikungunya virus infection. Arthritis Rheumatol. 2014;66: 319–326. 10.1002/art.38221
    1. Chaaitanya IK, Muruganandam N, Sundaram SG, Kawalekar O, Sugunan AP, Manimunda SP, et al. Role of proinflammatory cytokines and chemokines in chronic arthropathy in CHIKV infection. Viral Immunol. 2011;24: 265–271. 10.1089/vim.2010.0123
    1. Reddy V, Mani RS, Desai A, Ravi V. Correlation of plasma viral loads and presence of Chikungunya IgM antibodies with cytokine/chemokine levels during acute chikungunya virus infection. J Med Virol. 2014;86: 1393–1401. 10.1002/jmv.23875
    1. Chirathaworn C, Poovorawan Y, Lertmaharit S, Wuttirattanakowit N. Cytokine levels in patients with chikungunya virus infection. Asian Pac J Trop Med. 2013;6: 631–634. 10.1016/S1995-7645(13)60108-X
    1. Long KM, Heise MT. Protective and pathogenic responses to chikungunya virus infection. Curr Trop Med Rep. 2015;2: 13–21. 10.1007/s40475-015-0037-z
    1. Petitdemange C, Wauquier N, Vieillard V. Control of immunopathology during chikungunya virus infection. J Allergy Clin Immunol. 2015;135: 846–855. 10.1016/j.jaci.2015.01.039
    1. Nakaya HI, Gardner J, Poo YS, Major L, Pulendran B, Suhrbier A. Gene profiling of Chikungunya virus arthritis in a mouse model reveals significant overlap with rheumatoid arthritis. Arthritis Rheum. 2012;64: 3553–3563. 10.1002/art.34631
    1. Gardner J, Anraku I, Le TT, Larcher T, Major L, Roques P, et al. Chikungunya virus arthritis in adult wild-type mice. J Virol. 2010;84: 8021–8032. 10.1128/JVI.02603-09
    1. Gasque P, Couderc T, Lecuit M, Roques P, Ng LF. Chikungunya virus pathogenesis and immunity. Vector Borne Zoonotic Dis. 2015;15: 241–249. 10.1089/vbz.2014.1710
    1. Teo TH, Lum FM, Claser C, Lulla V, Lulla A, Merits A, et al. A pathogenic role for CD4+ T cells during chikungunya virus infection in mice. J Immunol. 2013;190: 259–269. 10.4049/jimmunol.1202177
    1. Lidbury BA, Rulli NE, Suhrbier A, Smith PN, McColl SR, Cunningham AL, et al. Macrophage-derived proinflammatory factors contribute to the development of arthritis and myositis after infection with an arthrogenic alphavirus. J Infect Dis. 2008;197: 1585–1593. 10.1086/587841
    1. Cardona-Ospina JA, Villamil-Gomez WE, Jimenez-Canizales CE, Castaneda-Hernandez DM, Rodriguez-Morales AJ. Estimating the burden of disease and the economic cost attributable to chikungunya, Colombia, 2014. Trans R Soc Trop Med Hyg. 2015;109: 793–802. 10.1093/trstmh/trv094
    1. Labadie K, Larcher T, Joubert C, Mannioui A, Delache B, Brochard P, et al. Chikungunya disease in nonhuman primates involves long-term viral persistence in macrophages. J Clin Invest. 2010;120: 894–906. 10.1172/JCI40104
    1. Perri S, Driver DA, Gardner JP, Sherrill S, Belli BA, Dubensky TW Jr., et al. Replicon vectors derived from Sindbis virus and Semliki forest virus that establish persistent replication in host cells. J Virol. 2000;74: 9802–9807.
    1. Atkinson T, Barrett AD, Mackenzie A, Dimmock NJ. Persistence of virulent Semliki Forest virus in mouse brain following co-inoculation with defective interfering particles. J Gen Virol. 1986;67: 1189–1194. 10.1099/0022-1317-67-6-1189
    1. Garcia-Arriaza J, Cepeda V, Hallengard D, Sorzano CO, Kummerer BM, Liljestrom P, et al. A novel poxvirus-based vaccine, MVA-CHIKV, is highly immunogenic and protects mice against chikungunya infection. J Virol. 2014;88: 3527–3547. 10.1128/JVI.03418-13
    1. Wang D, Suhrbier A, Penn-Nicholson A, Woraratanadharm J, Gardner J, Luo M, et al. A complex adenovirus vaccine against chikungunya virus provides complete protection against viraemia and arthritis. Vaccine. 2011;29: 2803–2809. 10.1016/j.vaccine.2011.01.108
    1. Selvarajah S, Sexton NR, Kahle KM, Fong RH, Mattia KA, Gardner J, et al. A neutralizing monoclonal antibody targeting the acid-sensitive region in chikungunya virus E2 protects from disease. PLoS Negl Trop Dis. 2013;7: e2423 10.1371/journal.pntd.0002423
    1. Goh LY, Hobson-Peters J, Prow NA, Gardner J, Bielefeldt-Ohmann H, Pyke AT, et al. Neutralizing monoclonal antibodies to the E2 protein of chikungunya virus protects against disease in a mouse model. Clin Immunol. 2013;149: 487–497. 10.1016/j.clim.2013.10.004
    1. Prow TW, Chen X, Prow NA, Fernando GJ, Tan CS, Raphael AP, et al. Nanopatch-targeted skin vaccination against West Nile virus and chikungunya virus in mice. Small. 2010;6: 1776–1784. 10.1002/smll.201000331
    1. Hallengard D, Lum FM, Kummerer BM, Lulla A, Lulla V, Garcia-Arriaza J, et al. Prime-boost immunization strategies against Chikungunya virus. J Virol. 2014;88: 13333–13343. 10.1128/JVI.01926-14
    1. Chen W, Foo SS, Taylor A, Lulla A, Merits A, Hueston L, et al. Bindarit, an inhibitor of monocyte chemotactic protein synthesis, protects against bone loss induced by chikungunya virus infection. J Virol. 2015;89: 581–593. 10.1128/JVI.02034-14
    1. Kam YW, Lum FM, Teo TH, Lee WW, Simarmata D, Harjanto S, et al. Early neutralizing IgG response to chikungunya virus in infected patients targets a dominant linear epitope on the E2 glycoprotein. EMBO Mol Med. 2012;4: 330–343. 10.1002/emmm.201200213
    1. Suhrbier A, Mahalingam S. The immunobiology of viral arthritides. Pharmacol Ther. 2009;124: 301–308. 10.1016/j.pharmthera.2009.09.005
    1. Zaid A, Rulli NE, Rolph MS, Suhrbier A, Mahalingam S. Disease exacerbation by etanercept in a mouse model of alphaviral arthritis and myositis. Arthritis Rheum. 2011;63: 488–491. 10.1002/art.30112
    1. Poo YS, Nakaya H, Gardner J, Larcher T, Schroder WA, Le TT, et al. CCR2 deficiency promotes exacerbated chronic erosive neutrophil-dominated chikungunya virus arthritis. J Virol. 2014;88: 6862–6872. 10.1128/JVI.03364-13
    1. Ekchariyawat P, Hamel R, Bernard E, Wichit S, Surasombatpattana P, Talignani L, et al. Inflammasome signaling pathways exert antiviral effect against chikungunya virus in human dermal fibroblasts. Infect Genet Evol. 2015;32: 401–408. 10.1016/j.meegid.2015.03.025
    1. Hoarau JJ, Gay F, Pelle O, Samri A, Jaffar-Bandjee MC, Gasque P, et al. Identical strength of the T cell responses against E2, nsP1 and capsid CHIKV proteins in recovered and chronic patients after the epidemics of 2005–2006 in La Reunion Island. PLoS ONE. 2013;8: e84695 10.1371/journal.pone.0084695
    1. Lohachanakul J, Phuklia W, Thannagith M, Thonsakulprasert T, Ubol S. High concentrations of circulating interleukin-6 and monocyte chemotactic protein-1 with low concentrations of interleukin-8 were associated with severe chikungunya fever during the 2009–2010 outbreak in Thailand. Microbiol Immunol. 2012;56: 134–138. 10.1111/j.1348-0421.2011.00417.x
    1. Chirathaworn C, Rianthavorn P, Wuttirattanakowit N, Poovorawan Y. Serum IL-18 and IL-18BP levels in patients with Chikungunya virus infection. Viral Immunol. 2010;23: 113–117. 10.1089/vim.2009.0077
    1. Chopra A, Anuradha V, Ghorpade R, Saluja M. Acute chikungunya and persistent musculoskeletal pain following the 2006 Indian epidemic: a 2-year prospective rural community study. Epidemiol Infect. 2012;140: 842–850. 10.1017/S0950268811001300
    1. Foo SS, Chen W, Taylor A, Sheng KC, Yu X, Teng TS, et al. Role of pentraxin 3 in shaping arthritogenic alphaviral disease: from enhanced viral replication to immunomodulation. PLoS Pathog. 2015;11: e1004649 10.1371/journal.ppat.1004649
    1. Rusinova I, Forster S, Yu S, Kannan A, Masse M, Cumming H, et al. Interferome v2.0: an updated database of annotated interferon-regulated genes. Nucleic Acids Res. 2013;41: D1040–1046. 10.1093/nar/gks1215
    1. Landis H, Simon-Jodicke A, Kloti A, Di Paolo C, Schnorr JJ, Schneider-Schaulies S, et al. Human MxA protein confers resistance to Semliki Forest virus and inhibits the amplification of a Semliki Forest virus-based replicon in the absence of viral structural proteins. J Virol. 1998;72: 1516–1522.
    1. Teng TS, Foo SS, Simamarta D, Lum FM, Teo TH, Lulla A, et al. Viperin restricts chikungunya virus replication and pathology. J Clin Invest. 2012;122: 4447–4460. 10.1172/JCI63120
    1. Werneke SW, Schilte C, Rohatgi A, Monte KJ, Michault A, Arenzana-Seisdedos F, et al. ISG15 is critical in the control of chikungunya virus infection independent of UbE1L mediated conjugation. PLoS Pathog. 2011;7: e1002322 10.1371/journal.ppat.1002322
    1. Reynaud JM, Kim DY, Atasheva S, Rasalouskaya A, White JP, Diamond MS, et al. IFIT1 differentially interferes with translation and replication of alphavirus genomes and promotes induction of type I interferon. PLoS Pathog. 2015;11: e1004863 10.1371/journal.ppat.1004863
    1. Mostafavi S, Yoshida H, Moodley D, LeBoite H, Rothamel K, Raj T, et al. Parsing the interferon transcriptional network and its disease associations. Cell. 2016;164: 564–578. 10.1016/j.cell.2015.12.032
    1. Sali TM, Pryke KM, Abraham J, Liu A, Archer I, Broeckel R, et al. Characterization of a novel human-specific STING agonist that elicits antiviral activity against emerging alphaviruses. PLoS Pathog. 2015;11: e1005324 10.1371/journal.ppat.1005324
    1. Grieves JL, Fye JM, Harvey S, Grayson JM, Hollis T, Perrino FW. Exonuclease TREX1 degrades double-stranded DNA to prevent spontaneous lupus-like inflammatory disease. Proc Natl Acad Sci U S A. 2015;112: 5117–5122. 10.1073/pnas.1423804112
    1. Holm CK, Rahbek SH, Gad HH, Bak RO, Jakobsen MR, Jiang Z, et al. Influenza A virus targets a cGAS-independent STING pathway that controls enveloped RNA viruses. Nat Commun. 2016;7: 10680 10.1038/ncomms10680
    1. Ahn J, Ruiz P, Barber GN. Intrinsic self-DNA triggers inflammatory disease dependent on STING. J Immunol. 2014;193: 4634–4642. 10.4049/jimmunol.1401337
    1. Hasan M, Koch J, Rakheja D, Pattnaik AK, Brugarolas J, Dozmorov I, et al. Trex1 regulates lysosomal biogenesis and interferon-independent activation of antiviral genes. Nat Immunol. 2013;14: 61–71. 10.1038/ni.2475
    1. Morrison TE, Simmons JD, Heise MT. Complement receptor 3 promotes severe Ross River virus-induced disease. J Virol. 2008;82: 11263–11272. 10.1128/JVI.01352-08
    1. Teo TH, Her Z, Tan JJ, Lum FM, Lee WW, Chan YH, et al. Caribbean and La Reunion chikungunya virus isolates differ in their capacity to induce proinflammatory Th1 and NK cell responses and acute joint pathology. J Virol. 2015;89: 7955–7969. 10.1128/JVI.00909-15
    1. Linn ML, Mateo L, Gardner J, Suhrbier A. Alphavirus-specific cytotoxic T lymphocytes recognize a cross-reactive epitope from the capsid protein and can eliminate virus from persistently infected macrophages. J Virol. 1998;72: 5146–5153.
    1. Meunier E, Broz P. Interferon-inducible GTPases in cell autonomous and innate immunity. Cell Microbiol. 2016;18: 168–180. 10.1111/cmi.12546
    1. Carnec X, Meertens L, Dejarnac O, Perera-Lecoin M, Hafirassou ML, Kitaura J, et al. The phosphatidylserine and phosphatidylethanolamine receptor CD300a binds dengue virus and enhances infection. J Virol. 2015;90: 92–102. 10.1128/JVI.01849-15
    1. Heng TS, Painter MW, Immunological Genome Project C. The Immunological Genome Project: networks of gene expression in immune cells. Nat Immunol. 2008;9: 1091–1094. 10.1038/ni1008-1091
    1. Hulsen T, de Vlieg J, Alkema W. BioVenn—a web application for the comparison and visualization of biological lists using area-proportional Venn diagrams. BMC Genomics. 2008;9: 488 10.1186/1471-2164-9-488
    1. Fros JJ, Major LD, Scholte FE, Gardner J, van Hemert MJ, Suhrbier A, et al. Chikungunya virus non-structural protein 2-mediated host shut-off disables the unfolded protein response. J Gen Virol. 2015;96: 580–589. 10.1099/vir.0.071845-0
    1. Hansen KD, Brenner SE, Dudoit S. Biases in Illumina transcriptome sequencing caused by random hexamer priming. Nucleic Acids Res. 2010;38: e131 10.1093/nar/gkq224
    1. Coffey LL, Beeharry Y, Borderia AV, Blanc H, Vignuzzi M. Arbovirus high fidelity variant loses fitness in mosquitoes and mice. Proc Natl Acad Sci U S A. 2011;108: 16038–16043. 10.1073/pnas.1111650108
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15: 550 10.1186/s13059-014-0550-8
    1. Hart T, Komori HK, LaMere S, Podshivalova K, Salomon DR. Finding the active genes in deep RNA-seq gene expression studies. BMC Genomics. 2013;14: 778 10.1186/1471-2164-14-778
    1. Kumagai Y, Takeuchi O, Kato H, Kumar H, Matsui K, Morii E, et al. Alveolar macrophages are the primary interferon-alpha producer in pulmonary infection with RNA viruses. Immunity. 2007;27: 240–252. 10.1016/j.immuni.2007.07.013
    1. Wilson J The role of type I interferon in the immunobiology of chikungunya virus. PhD Thesis. The University of Queensland 2015.: University of Queensland.
    1. Olagnier D, Scholte FE, Chiang C, Albulescu IC, Nichols C, He Z, et al. Inhibition of dengue and chikungunya virus infections by RIG-I-mediated type I interferon-independent stimulation of the innate antiviral response. J Virol. 2014;88: 4180–4194. 10.1128/JVI.03114-13
    1. Sarkar SN, Sen GC. Novel functions of proteins encoded by viral stress-inducible genes. Pharmacol Ther. 2004;103: 245–259. 10.1016/j.pharmthera.2004.07.007
    1. Lazear HM, Lancaster A, Wilkins C, Suthar MS, Huang A, Vick SC, et al. IRF-3, IRF-5, and IRF-7 coordinately regulate the type I IFN response in myeloid dendritic cells downstream of MAVS signaling. PLoS Pathog. 2013;9: e1003118 10.1371/journal.ppat.1003118
    1. Chmielewski S, Piaszyk-Borychowska A, Wesoly J, Bluyssen HA. STAT1 and IRF8 in vascular inflammation and cardiovascular disease: Diagnostic and therapeutic potential. Int Rev Immunol. 2015: 1–21.
    1. Kuchipudi SV. The Complex Role of STAT3 in Viral Infections. J Immunol Res. 2015;2015: 272359 10.1155/2015/272359
    1. Yarilina A, Park-Min KH, Antoniv T, Hu X, Ivashkiv LB. TNF activates an IRF1-dependent autocrine loop leading to sustained expression of chemokines and STAT1-dependent type I interferon-response genes. Nat Immunol. 2008;9: 378–387. 10.1038/ni1576
    1. Harman AN, Lai J, Turville S, Samarajiwa S, Gray L, Marsden V, et al. HIV infection of dendritic cells subverts the IFN induction pathway via IRF-1 and inhibits type 1 IFN production. Blood. 2011;118: 298–308. 10.1182/blood-2010-07-297721
    1. Ren G, Cui K, Zhang Z, Zhao K. Division of labor between IRF1 and IRF2 in regulating different stages of transcriptional activation in cellular antiviral activities. Cell Biosci. 2015;5: 17 10.1186/s13578-015-0007-0
    1. Cheon H, Holvey-Bates EG, Schoggins JW, Forster S, Hertzog P, Imanaka N, et al. IFNbeta-dependent increases in STAT1, STAT2, and IRF9 mediate resistance to viruses and DNA damage. EMBO J. 2013;32: 2751–2763. 10.1038/emboj.2013.203
    1. Honda K, Yanai H, Negishi H, Asagiri M, Sato M, Mizutani T, et al. IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature. 2005;434: 772–777. 10.1038/nature03464
    1. Burdeinick-Kerr R, Govindarajan D, Griffin DE. Noncytolytic clearance of sindbis virus infection from neurons by gamma interferon is dependent on Jak/STAT signaling. J Virol. 2009;83: 3429–3435. 10.1128/JVI.02381-08
    1. Lee EY, Schultz KL, Griffin DE. Mice deficient in interferon-gamma or interferon-gamma receptor 1 have distinct inflammatory responses to acute viral encephalomyelitis. PLoS ONE. 2013;8: e76412 10.1371/journal.pone.0076412
    1. Pinto AJ, Morahan PS, Brinton M, Stewart D, Gavin E. Comparative therapeutic efficacy of recombinant interferons-alpha, -beta, and -gamma against alphatogavirus, bunyavirus, flavivirus, and herpesvirus infections. J Interferon Res. 1990;10: 293–298. 10.1089/jir.1990.10.293
    1. Gardner CL, Burke CW, Higgs ST, Klimstra WB, Ryman KD. Interferon-alpha/beta deficiency greatly exacerbates arthritogenic disease in mice infected with wild-type chikungunya virus but not with the cell culture-adapted live-attenuated 181/25 vaccine candidate. Virology. 2012;425: 103–112. 10.1016/j.virol.2011.12.020
    1. Voskoboinik I, Whisstock JC, Trapani JA. Perforin and granzymes: function, dysfunction and human pathology. Nat Rev Immunol. 2015;15: 388–400. 10.1038/nri3839
    1. Takeuchi A, Badr Mel S, Miyauchi K, Ishihara C, Onishi R, Guo Z, et al. CRTAM determines the CD4+ cytotoxic T lymphocyte lineage. J Exp Med. 2016;213: 123–138. 10.1084/jem.20150519
    1. Joeckel LT, Bird PI. Are all granzymes cytotoxic in vivo? Biol Chem. 2014;395: 181–202. 10.1515/hsz-2013-0238
    1. Suhrbier A, Fernan A, Burrows SR, Saul A, Moss DJ. BLT esterase activity as an alternative to chromium release in cytotoxic T cell assays. J Immunol Methods. 1991;145: 43–53.
    1. Wensink AC, Hack CE, Bovenschen N. Granzymes regulate proinflammatory cytokine responses. J Immunol. 2015;194: 491–497. 10.4049/jimmunol.1401214
    1. Hildebrand D, Bode KA, Riess D, Cerny D, Waldhuber A, Rommler F, et al. Granzyme A produces bioactive IL-1beta through a nonapoptotic inflammasome-independent pathway. Cell Rep. 2014;9: 910–917. 10.1016/j.celrep.2014.10.003
    1. Kagi D, Seiler P, Pavlovic J, Ledermann B, Burki K, Zinkernagel RM, et al. The roles of perforin- and Fas-dependent cytotoxicity in protection against cytopathic and noncytopathic viruses. Eur J Immunol. 1995;25: 3256–3262. 10.1002/eji.1830251209
    1. Mullbacher A, Ebnet K, Blanden RV, Hla RT, Stehle T, Museteanu C, et al. Granzyme A is critical for recovery of mice from infection with the natural cytopathic viral pathogen, ectromelia. Proc Natl Acad Sci U S A. 1996;93: 5783–5787.
    1. Pereira RA, Simon MM, Simmons A. Granzyme A, a noncytolytic component of CD8(+) cell granules, restricts the spread of herpes simplex virus in the peripheral nervous systems of experimentally infected mice. J Virol. 2000;74: 1029–1032.
    1. Burrack KS, Montgomery SA, Homann D, Morrison TE. CD8+ T cells control Ross River virus infection in musculoskeletal tissues of infected mice. J Immunol. 2015;194: 678–689. 10.4049/jimmunol.1401833
    1. Hartmann W, Marsland BJ, Otto B, Urny J, Fleischer B, Korten S. A novel and divergent role of granzyme A and B in resistance to helminth infection. J Immunol. 2011;186: 2472–2481. 10.4049/jimmunol.0902157
    1. Froelich CJ, Pardo J, Simon MM. Granule-associated serine proteases: granzymes might not just be killer proteases. Trends Immunol. 2009;30: 117–123. 10.1016/j.it.2009.01.002
    1. Sower LE, Froelich CJ, Allegretto N, Rose PM, Hanna WD, Klimpel GR. Extracellular activities of human granzyme A. Monocyte activation by granzyme A versus alpha-thrombin. J Immunol. 1996;156: 2585–2590.
    1. Spaeny-Dekking EH, Kamp AM, Froelich CJ, Hack CE. Extracellular granzyme A, complexed to proteoglycans, is protected against inactivation by protease inhibitors. Blood. 2000;95: 1465–1472.
    1. Tremblay GM, Wolbink AM, Cormier Y, Hack CE. Granzyme activity in the inflamed lung is not controlled by endogenous serine proteinase inhibitors. J Immunol. 2000;165: 3966–3969.
    1. Kaiserman D, Stewart SE, Plasman K, Gevaert K, Van Damme P, Bird PI. Identification of Serpinb6b as a species-specific mouse granzyme A inhibitor suggests functional divergence between human and mouse granzyme A. J Biol Chem. 2014;289: 9408–9417. 10.1074/jbc.M113.525808
    1. Spaeny-Dekking EH, Hanna WL, Wolbink AM, Wever PC, Kummer JA, Swaak AJ, et al. Extracellular granzymes A and B in humans: detection of native species during CTL responses in vitro and in vivo. J Immunol. 1998;160: 3610–3616.
    1. ten Berge IJ, Wever PC, Wolbink AM, Surachno J, Wertheim PM, Spaeny LH, et al. Increased systemic levels of soluble granzymes A and B during primary cytomegalovirus infection after renal transplantation. Transplant Proc. 1998;30: 3972–3974.
    1. Tak PP, Spaeny-Dekking L, Kraan MC, Breedveld FC, Froelich CJ, Hack CE. The levels of soluble granzyme A and B are elevated in plasma and synovial fluid of patients with rheumatoid arthritis (RA). Clin Exp Immunol. 1999;116: 366–370. 10.1046/j.1365-2249.1999.00881.x
    1. Bade B, Lohrmann J, ten Brinke A, Wolbink AM, Wolbink GJ, ten Berge IJ, et al. Detection of soluble human granzyme K in vitro and in vivo. Eur J Immunol. 2005;35: 2940–2948. 10.1002/eji.200526249
    1. Charrel RN, de Lamballerie X, Raoult D. Chikungunya outbreaks-the globalization of vectorborne diseases. N Engl J Med. 2007;356: 769–771. 10.1056/NEJMp078013
    1. Forrester NL, Guerbois M, Adams AP, Liang X, Weaver SC. Analysis of intrahost variation in Venezuelan equine encephalitis virus reveals repeated deletions in the 6-kilodalton protein gene. J Virol. 2011;85: 8709–8717. 10.1128/JVI.00165-11
    1. Folco EJ, Rocha VZ, Lopez-Ilasaca M, Libby P. Adiponectin inhibits pro-inflammatory signaling in human macrophages independent of interleukin-10. J Biol Chem. 2009;284: 25569–25575. 10.1074/jbc.M109.019786
    1. Steichen AL, Binstock BJ, Mishra BB, Sharma J. C-type lectin receptor Clec4d plays a protective role in resolution of Gram-negative pneumonia. J Leukoc Biol. 2013;94: 393–398. 10.1189/jlb.1212622
    1. McNab F, Mayer-Barber K, Sher A, Wack A, O'Garra A. Type I interferons in infectious disease. Nat Rev Immunol. 2015;15: 87–103. 10.1038/nri3787
    1. Joeckel LT, Wallich R, Martin P, Sanchez-Martinez D, Weber FC, Martin SF, et al. Mouse granzyme K has pro-inflammatory potential. Cell Death Differ. 2011;18: 1112–1119. 10.1038/cdd.2011.5
    1. Sharma M, Merkulova Y, Raithatha S, Parkinson LG, Shen Y, Cooper D, et al. Extracellular granzyme K mediates endothelial activation through the cleavage of protease-activated receptor-1. FEBS J. 2016;283: 1734–1747. 10.1111/febs.13699
    1. Wensink AC, Kemp V, Fermie J, Garcia Laorden MI, van der Poll T, Hack CE, et al. Granzyme K synergistically potentiates LPS-induced cytokine responses in human monocytes. Proc Natl Acad Sci U S A. 2014;111: 5974–5979. 10.1073/pnas.1317347111
    1. Ahmad J, Bird PI, Kaiserman D. Analysis of the evolution of granule associated serine proteases of immune defence (GASPIDs) suggests a revised nomenclature. Biol Chem. 2014;395: 1253–1262. 10.1515/hsz-2014-0174
    1. Plasman K, Demol H, Bird PI, Gevaert K, Van Damme P. Substrate specificities of the granzyme tryptases A and K. J Proteome Res. 2014;13: 6067–6077. 10.1021/pr500968d
    1. Arias MA, Jimenez de Bagues MP, Aguilo N, Menao S, Hervas-Stubbs S, de Martino A, et al. Elucidating sources and roles of granzymes A and B during bacterial infection and sepsis. Cell Rep. 2014;8: 420–429. 10.1016/j.celrep.2014.06.012
    1. Metkar SS, Menaa C, Pardo J, Wang B, Wallich R, Freudenberg M, et al. Human and mouse granzyme A induce a proinflammatory cytokine response. Immunity. 2008;29: 720–733. 10.1016/j.immuni.2008.08.014
    1. Santiago L, Menaa C, Arias M, Martin P, Jaime-Sanchez P, Metkar S, et al. Granzyme A contributes to inflammatory arthritis through stimulation of osteoclastogenesis. Arthritis Rheumatol. 2016.
    1. Le TT, Skak K, Schroder K, Schroder WA, Boyle GM, Pierce CJ, et al. IL-1 contributes to the anti-cancer efficacy of ingenol mebutate. PLoS ONE. 2016;11: e0153975 10.1371/journal.pone.0153975
    1. Petitdemange C, Becquart P, Wauquier N, Beziat V, Debre P, Leroy EM, et al. Unconventional repertoire profile is imprinted during acute chikungunya infection for natural killer cells polarization toward cytotoxicity. PLoS Pathog. 2011;7: e1002268 10.1371/journal.ppat.1002268
    1. Assuncao-Miranda I, Cruz-Oliveira C, Da Poian AT. Molecular mechanisms involved in the pathogenesis of alphavirus-induced arthritis. Biomed Res Int. 2013;2013: 973516 10.1155/2013/973516
    1. Vrazo AC, Hontz AE, Figueira SK, Butler BL, Ferrell JM, Binkowski BF, et al. Live cell evaluation of granzyme delivery and death receptor signaling in tumor cells targeted by human natural killer cells. Blood. 2015;126: e1–e10. 10.1182/blood-2015-03-632273
    1. Hidalgo LG, Einecke G, Allanach K, Halloran PF. The transcriptome of human cytotoxic T cells: similarities and disparities among allostimulated CD4(+) CTL, CD8(+) CTL and NK cells. Am J Transplant. 2008;8: 627–636. 10.1111/j.1600-6143.2007.02128.x
    1. Weiskopf D, Bangs DJ, Sidney J, Kolla RV, De Silva AD, de Silva AM, et al. Dengue virus infection elicits highly polarized CX3CR1+ cytotoxic CD4+ T cells associated with protective immunity. Proc Natl Acad Sci U S A. 2015;112: E4256–4263. 10.1073/pnas.1505956112
    1. Simon MM, Kramer MD, Prester M, Gay S. Mouse T-cell associated serine proteinase 1 degrades collagen type IV: a structural basis for the migration of lymphocytes through vascular basement membranes. Immunology. 1991;73: 117–119.
    1. Prakash MD, Munoz MA, Jain R, Tong PL, Koskinen A, Regner M, et al. Granzyme B promotes cytotoxic lymphocyte transmigration via basement membrane remodeling. Immunity. 2014;41: 960–972. 10.1016/j.immuni.2014.11.012
    1. Suidan HS, Bouvier J, Schaerer E, Stone SR, Monard D, Tschopp J. Granzyme A released upon stimulation of cytotoxic T lymphocytes activates the thrombin receptor on neuronal cells and astrocytes. Proc Natl Acad Sci U S A. 1994;91: 8112–8116.
    1. Gutierrez-Rodriguez M, Herranz R. From Multiple PAR1 Receptor/Protein Interactions to their Multiple Therapeutic Implications. Curr Top Med Chem. 2015;15: 2080–2114.
    1. Ramachandran R, Altier C, Oikonomopoulou K, Hollenberg MD. Proteinases, Their Extracellular Targets, and Inflammatory Signaling. Pharmacol Rev. 2016;68: 1110–1142. 10.1124/pr.115.010991
    1. Sambrano GR, Coughlin SR. The carboxyl tail of protease-activated receptor-1 is required for chemotaxis. Correlation of signal termination and directional migration. J Biol Chem. 1999;274: 20178–20184.
    1. d'Audigier C, Cochain C, Rossi E, Guerin CL, Bieche I, Blandinieres A, et al. Thrombin receptor PAR-1 activation on endothelial progenitor cells enhances chemotaxis-associated genes expression and leukocyte recruitment by a COX-2-dependent mechanism. Angiogenesis. 2015;18: 347–359. 10.1007/s10456-015-9471-8
    1. Chen D, Carpenter A, Abrahams J, Chambers RC, Lechler RI, McVey JH, et al. Protease-activated receptor 1 activation is necessary for monocyte chemoattractant protein 1-dependent leukocyte recruitment in vivo. J Exp Med. 2008;205: 1739–1746. 10.1084/jem.20071427
    1. Aerts L, Hamelin ME, Rheaume C, Lavigne S, Couture C, Kim W, et al. Modulation of protease activated receptor 1 influences human metapneumovirus disease severity in a mouse model. PLoS ONE. 2013;8: e72529 10.1371/journal.pone.0072529
    1. Mosnier LO, Sinha RK, Burnier L, Bouwens EA, Griffin JH. Biased agonism of protease-activated receptor 1 by activated protein C caused by noncanonical cleavage at Arg46. Blood. 2012;120: 5237–5246. 10.1182/blood-2012-08-452169
    1. Petitdemange C, Wauquier N, Devilliers H, Yssel H, Mombo I, Caron M, et al. Longitudinal Analysis of Natural Killer Cells in Dengue Virus-Infected Patients in Comparison to Chikungunya and Chikungunya/Dengue Virus-Infected Patients. PLoS Negl Trop Dis. 2016;10: e0004499 10.1371/journal.pntd.0004499
    1. Hermsen CC, Konijnenberg Y, Mulder L, Loe C, van Deuren M, van der Meer JW, et al. Circulating concentrations of soluble granzyme A and B increase during natural and experimental Plasmodium falciparum infections. Clin Exp Immunol. 2003;132: 467–472. 10.1046/j.1365-2249.2003.02160.x
    1. Lauw FN, Simpson AJ, Hack CE, Prins JM, Wolbink AM, van Deventer SJ, et al. Soluble granzymes are released during human endotoxemia and in patients with severe infection due to gram-negative bacteria. J Infect Dis. 2000;182: 206–213. 10.1086/315642
    1. Gardner J, Rudd PA, Prow NA, Belarbi E, Roques P, Larcher T, et al. Infectious chikungunya virus in the saliva of mice, monkeys and humans. PLoS ONE. 2015;10: e0139481 10.1371/journal.pone.0139481
    1. Mullbacher A, Waring P, Tha Hla R, Tran T, Chin S, Stehle T, et al. Granzymes are the essential downstream effector molecules for the control of primary virus infections by cytolytic leukocytes. Proc Natl Acad Sci U S A. 1999;96: 13950–13955.
    1. Anthony DA, Andrews DM, Chow M, Watt SV, House C, Akira S, et al. A role for granzyme M in TLR4-driven inflammation and endotoxicosis. J Immunol. 2010;185: 1794–1803. 10.4049/jimmunol.1000430
    1. Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics. 2009;25: 1105–1111. 10.1093/bioinformatics/btp120
    1. Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR, et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc. 2012;7: 562–578. 10.1038/nprot.2012.016
    1. Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ, et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 2010;28: 511–515. 10.1038/nbt.1621
    1. Huang da W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009;37: 1–13. 10.1093/nar/gkn923
    1. Thorvaldsdottir H, Robinson JT, Mesirov JP. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform. 2013;14: 178–192. 10.1093/bib/bbs017
    1. Kearse M, Moir R, Wilson A, Stones-Havas S, Cheung M, Sturrock S, et al. Geneious Basic: an integrated and extendable desktop software platform for the organization and analysis of sequence data. Bioinformatics. 2012;28: 1647–1649. 10.1093/bioinformatics/bts199
    1. Kel AE, Gossling E, Reuter I, Cheremushkin E, Kel-Margoulis OV, Wingender E. MATCH: A tool for searching transcription factor binding sites in DNA sequences. Nucleic Acids Res. 2003;31: 3576–3579.
    1. Frith MC, Fu Y, Yu L, Chen JF, Hansen U, Weng Z. Detection of functional DNA motifs via statistical over-representation. Nucleic Acids Res. 2004;32: 1372–1381. 10.1093/nar/gkh299
    1. Sun J, Coughlin P, Salem HH, Bird P. Production and characterization of recombinant human proteinase inhibitor 6 expressed in Pichia pastoris. Biochim Biophys Acta. 1995;1252: 28–34.
    1. Risso D, Schwartz K, Sherlock G, Dudoit S. GC-content normalization for RNA-Seq data. BMC Bioinformatics. 2011;12: 480 10.1186/1471-2105-12-480

Source: PubMed

3
Suscribir