A balanced game: chicken macrophage response to ALV-J infection

Min Feng, Tingting Xie, Yuanfang Li, Nan Zhang, Qiuyuan Lu, Yaohong Zhou, Meiqing Shi, Jingchen Sun, Xiquan Zhang, Min Feng, Tingting Xie, Yuanfang Li, Nan Zhang, Qiuyuan Lu, Yaohong Zhou, Meiqing Shi, Jingchen Sun, Xiquan Zhang

Abstract

Avian leukosis virus subgroup J (ALV-J) infection can cause tumors and immunosuppression in infected chickens. Macrophages play a central role in host defense against invading pathogens. In this study, we discovered an interesting phenomenon: ALV-J replication is weakened from 3 hours post-infection (hpi) to 36 hpi, which was verified using Western blotting and RT-PCR. To further investigate the interaction between ALV-J and macrophages, transcriptome analysis was performed to analyze the host genes' function in chicken primary monocyte-derived macrophages (MDM). Compared to the uninfected control, 624 up-regulated differentially expressed genes (DEG) and 341 down-regulated DEG at 3 hpi, and 174 up-regulated DEG and 87 down-regulated DEG at 36 hpi were identified in chicken MDM, respectively. ALV-J infection induced strong innate immune responses in chicken MDM at 3 hpi, instead of 36 hpi, according to the analysis results of Gene Ontology and KEGG pathway. Importantly, the host factors, such as up-regulated MIP-3α, IL-1β, iNOS, K60, IRG1, CH25H, NFKBIZ, lysozyme and OASL were involved in the host defense response during the course of ALV-J infection. On the contrary, up-regulated EX-FABP, IL4I1, COX-2, NFKBIA, TNFAIP3 and the Jak STAT pathway inhibitors including CISH, SOCS1 and SOCS3 are beneficial to ALV-J survival in chicken macrophages. We speculated that ALV-J tropism for macrophages helps to establish a latent infection in chicken MDM from 6 to 36 hpi. The present study provides a comprehensive view of the interactions between macrophages and ALV-J. It suggests the mechanisms of defense of chicken macrophages against ALV-J invasion and how ALV-J escape the host innate immune responses.

Figures

Figure 1
Figure 1
Detection of ALV-J replication in chicken MDM from 3 hpi to 36 hpi. A PCR detection with DNA template and ALV-J specific primer. All samples extracted from 3 hpi to 36 hpi produced specific 545 bp fragment. B RT-PCR detection with cDNA template and ALV-J specific primer. C Western blotting analysis shows that samples extracted from 3 hpi produced obvious specific ALV-J envelope protein blots, but the protein blots became weaker at 6 hpi and tended to disappear at 12 to 36 hpi.
Figure 2
Figure 2
DEG in chicken MDM infected with ALV-J at 3 hpi and 36 hpi. Venn diagrams of up-regulated DEG (A) and down-regulated DEG (B) at 3 hpi and 36 hpi from chicken MDM. C Venn diagrams of up-regulated DEG at 3 hpi and down-regulated DEG at 36 hpi in chicken MDM. D Venn diagrams of down-regulated DEG at 3 hpi compared up-regulated DEG at 36 hpi in chicken MDM.
Figure 3
Figure 3
Gene ontology (GO) terms analysis of DEG expressed in ALV-J-infected MDM. Representative GO terms of up-regulated DEG in the ALV-J-infected MDM at A 3 hpi and B 36 hpi. Down-regulated DEG enriched in the representative GO terms of ALV-J-infected MDM at C 3 hpi and D 36 hpi.
Figure 4
Figure 4
KEGG pathways in ALV-J-infected MDM. KEGG pathways of up-regulated DEG expressed in ALV-J-infected MDM at A 3 hpi and B 36 hpi. C Down-regulated DEG enriched in KEGG pathways of ALV-J-infected MDM at 3 hpi.
Figure 5
Figure 5
Heatmap of immune-related DEG at different time points after ALV-J infection. DEG with similar expressed patterns were clustered and are displayed in a heatmap format. Color intensity corresponds to relative expression level normalized according to log2 fold change. A Selected immune-related DEG from 3 hpi and 36 hpi. Red, up-regulated DEG; blue, down-regulated DEG. B ISG expression in MDM at 3 hpi and 36 hpi. Red, up-regulated ISG; purple, down-regulated ISG.
Figure 6
Figure 6
Validation of RNA-Seq data by qPCR. DEG were selected at (A) 3 hpi and (B) 36 hpi. qPCR results were represented using relative expression value. RNA-seq value is log2(foldchange) values of DEG. *p < 0.05, **p < 0.01, ***p < 0.001.
Figure 7
Figure 7
Overexpression ofK60, IRG1, OASLandCH25Hcould inhibit ALV-J replication. MDM cells were transfected with pCMV-K60, pCMV-IRG1, pCMV-OASL, pCMV-CH25H and infected with ALV-J strain SCAU-HN06 at 24 post-transfection. MDM cells transfected with pCMV-EGFP as a control. A SCAU-HN06 strain envelop protein was measured by Western blot at 3 hpi. B The level of SCAU-HN06 strain envelop protein was analyzed by Image Studio Ver 5.2 (Odyssey Fc). C The expression of SCAU-HN06 env gene was measured by qPCR at 3 hpi. *p < 0.05, **p < 0.01.
Figure 8
Figure 8
Overexpression ofCISH, EX-FABP, IL4I1andSOCS3could enhance ALV-J replication. MDM cells were transfected with plasmids including EGFP, CISH, EX-FABP, IL4I1 and SOCS3 and infected with SCAU-HN06 at 24 post-transfection. A, B The level of SCAU-HN06 strain envelop protein was detected by Western blot at 3 hpi and analyzed by Image Studio Ver 5.2 (Odyssey Fc). (C) The expression of SCAU-HN06 env gene was detected by qPCR at 3 hpi. *p < 0.05, **p < 0.01.

References

    1. Feng M, Zhang X. Immunity to avian leukosis virus: where are we now and what should we do? Front Immunol. 2016;7:624. doi: 10.3389/fimmu.2016.00624.
    1. Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013;496:445–455. doi: 10.1038/nature12034.
    1. Campbell JH, Hearps AC, Martin GE, Williams KC, Crowe SM. The importance of monocytes and macrophages in HIV pathogenesis, treatment, and cure. AIDS. 2014;28:2175–2187. doi: 10.1097/QAD.0000000000000408.
    1. Chen YC, Wang SY. Activation of terminally differentiated human monocytes/macrophages by dengue virus: productive infection, hierarchical production of innate cytokines and chemokines, and the synergistic effect of lipopolysaccharide. J Virol. 2002;76:9877–9887. doi: 10.1128/JVI.76.19.9877-9887.2002.
    1. Shi C, Liu Y, Ding Y, Zhang Y, Zhang J. PRRSV receptors and their roles in virus infection. Arch Microbiol. 2015;197:503–512. doi: 10.1007/s00203-015-1088-1.
    1. Lee CC, Wu CC, Lin TL. Role of chicken melanoma differentiation-associated gene 5 in induction and activation of innate and adaptive immune responses to infectious bursal disease virus in cultured macrophages. Arch Virol. 2015;160:3021–3035. doi: 10.1007/s00705-015-2612-y.
    1. Shojadoost B, Kulkarni RR, Brisbin JT, Quinteiro-Filho W, Alkie TN, Sharif S (2017) Interactions between lactobacilli and chicken macrophages induce antiviral responses against avian influenza virus. Res Vet Sci, in press
    1. Zhang P, Ding Z, Liu X, Chen Y, Li J, Tao Z, Fei Y, Xue C, Qian J, Wang X, Li Q, Stoeger T, Chen J, Bi Y, Yin R. Enhanced replication of virulent Newcastle disease virus in chicken macrophages is due to polarized activation of cells by inhibition of TLR7. Front Immunol. 2018;9:366. doi: 10.3389/fimmu.2018.00366.
    1. Han X, Tian Y, Guan R, Gao W, Yang X, Zhou L, Wang H. Infectious bronchitis virus infection induces apoptosis during replication in chicken macrophage HD11 cells. Viruses. 2017;9:E198. doi: 10.3390/v9080198.
    1. Feng M, Dai M, Cao W, Tan Y, Li Z, Shi M, Zhang X. ALV-J strain SCAU-HN06 induces innate immune responses in chicken primary monocyte-derived macrophages. Poult Sci. 2017;96:42–50. doi: 10.3382/ps/pew229.
    1. Dawes ME, Griggs LM, Collisson EW, Briles WE, Drechsler Y. Dramatic differences in the response of macrophages from B2 and B19 MHC-defined haplotypes to interferon gamma and polyinosinic:polycytidylic acid stimulation. Poult Sci. 2014;93:830–838. doi: 10.3382/ps.2013-03511.
    1. Smith LM, Brown SR, Howes K, McLeod S, Arshad SS, Barron GS, Venugopal K, McKay JC, Payne LN. Development and application of polymerase chain reaction (PCR) tests for the detection of subgroup J avian leukosis virus. Virus Res. 1998;54:87–98. doi: 10.1016/S0168-1702(98)00022-7.
    1. Dai M, Feng M, Ye Y, Wu X, Liu D, Liao M, Cao W. Exogenous avian leukosis virus-induced activation of the ERK/AP1 pathway is required for virus replication and correlates with virus-induced tumorigenesis. Sci Rep. 2016;6:19226. doi: 10.1038/srep19226.
    1. Gallus_gallus-4.0. . Accessed Apr 2013
    1. da Huang W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4:44–57. doi: 10.1038/nprot.2008.211.
    1. Ye J, Coulouris G, Zaretskaya I, Cutcutache I, Rozen S, Madden T. Primer-BLAST: a tool to design target-specific primers for polymerase chain reaction. BMC Bioinf. 2012;13:14. doi: 10.1186/1471-2105-13-134.
    1. Kint J, Fernandez-Gutierrez M, Maier HJ, Britton P, Langereis MA, Koumans J, Wiegertjes GF, Forlenza M. Activation of the chicken type I interferon response by infectious bronchitis coronavirus. J Virol. 2015;89:1156–1167. doi: 10.1128/JVI.02671-14.
    1. Liu XL, Shan WJ, Jia LJ, Yang X, Zhang JJ, Wu YR, Xu FZ, Li JN. Avian leukosis virus subgroup J triggers caspase-1-mediated inflammatory response in chick livers. Virus Res. 2016;215:65–71. doi: 10.1016/j.virusres.2016.01.011.
    1. Tao ZY, Zhu CH, Shi ZH, Song C, Xu WJ, Song WT, Zou JM, Qin AJ. Molecular characterization, expression, and functional analysis of NOD1 in Qingyuan partridge chicken. Genet Mol Res. 2015;14:2691–2701. doi: 10.4238/2015.March.30.29.
    1. Zhou Z, Wang Z, Cao L, Hu S, Zhang Z, Qin B, Guo Z, Nie K. Upregulation of chicken TLR4, TLR15 and MyD88 in heterophils and monocyte-derived macrophages stimulated with Eimeria tenella in vitro. Exp Parasitol. 2013;133:427–433. doi: 10.1016/j.exppara.2013.01.002.
    1. Giotis ES, Robey RC, Skinner NG, Tomlinson CD, Goodbourn S, Skinner MA. Chicken interferome: avian interferon-stimulated genes identified by microarray and RNA-seq of primary chick embryo fibroblasts treated with a chicken type I interferon (IFN-alpha) Vet Res. 2016;47:75. doi: 10.1186/s13567-016-0363-8.
    1. Liu SY, Sanchez DJ, Aliyari R, Lu S, Cheng G. Systematic identification of type I and type II interferon-induced antiviral factors. Proc Natl Acad Sci U S A. 2012;109:4239–4244. doi: 10.1073/pnas.1114981109.
    1. Schoggins JW, Wilson SJ, Panis M, Murphy MY, Jones CT, Bieniasz P, Rice CM. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature. 2011;472:481–485. doi: 10.1038/nature09907.
    1. Zhu M, Ma X, Cui X, Zhou J, Li C, Huang L, Shang Y, Cheng Z. Inhibition of avian tumor virus replication by CCCH-type zinc finger antiviral protein. Oncotarget. 2017;8:58865–58871.
    1. Dai M, Wu S, Feng M, Feng S, Sun C, Bai D, Gu M, Liao M, Cao W. Recombinant chicken interferon-alpha inhibits the replication of exogenous avian leukosis virus (ALV) in DF-1 cells. Mol Immunol. 2016;76:62–69. doi: 10.1016/j.molimm.2016.06.012.
    1. Schoggins JW, Rice CM. Interferon-stimulated genes and their antiviral effector functions. Curr Opin Virol. 2011;1:519–525. doi: 10.1016/j.coviro.2011.10.008.
    1. Liu SY, Aliyari R, Chikere K, Li G, Marsden MD, Smith JK, Pernet O, Guo H, Nusbaum R, Zack JA, Freiberg AN, Su L, Lee B, Cheng G. Interferon-inducible cholesterol-25-hydroxylase broadly inhibits viral entry by production of 25-hydroxycholesterol. Immunity. 2013;38:92–105. doi: 10.1016/j.immuni.2012.11.005.
    1. Lafferty MK, Sun L, DeMasi L, Lu W, Garzino-Demo A. CCR6 ligands inhibit HIV by inducing APOBEC3G. Blood. 2010;115:1564–1571. doi: 10.1182/blood-2009-06-226423.
    1. Ghosh M, Shen Z, Schaefer TM, Fahey JV, Gupta P, Wira CR. CCL20/MIP3alpha is a novel anti-HIV-1 molecule of the human female reproductive tract. Am J Reprod Immunol. 2009;62:60–71. doi: 10.1111/j.1600-0897.2009.00713.x.
    1. Yang D, Chen Q, Hoover DM, Staley P, Tucker KD, Lubkowski J, Oppenheim JJ. Many chemokines including CCL20/MIP-3alpha display antimicrobial activity. J Leukoc Biol. 2003;74:448–455. doi: 10.1189/jlb.0103024.
    1. Dinarello CA. Interleukin-1 and the pathogenesis of the acute-phase response. N Engl J Med. 1984;311:1413–1418. doi: 10.1056/NEJM198411293112205.
    1. Smirnova MG, Kiselev SL, Gnuchev NV, Birchall JP, Pearson JP. Role of the pro-inflammatory cytokines tumor necrosis factor-alpha, interleukin-1 beta, interleukin-6 and interleukin-8 in the pathogenesis of the otitis media with effusion. Eur Cytokine Netw. 2002;13:161–172.
    1. Torre D, Pugliese A, Speranza F. Role of nitric oxide in HIV-1 infection: friend or foe? Lancet Infect Dis. 2002;2:273–280. doi: 10.1016/S1473-3099(02)00262-1.
    1. Kaiser P, Hughes S, Bumstead N. The chicken 9E3/CEF4 CXC chemokine is the avian orthologue of IL8 and maps to chicken chromosome 4 syntenic with genes flanking the mammalian chemokine cluster. Immunogenetics. 1999;49:673–684. doi: 10.1007/s002510050664.
    1. Sick C, Schneider K, Staeheli P, Weining KC. Novel chicken CXC and CC chemokines. Cytokine. 2000;12:181–186. doi: 10.1006/cyto.1999.0543.
    1. Larson CL, Shah DH, Dhillon AS, Call DR, Ahn S, Haldorson GJ, Davitt C, Konkel ME. Campylobacter jejuni invade chicken LMH cells inefficiently and stimulate differential expression of the chicken CXCLi1 and CXCLi2 cytokines. Microbiology. 2008;154:3835–3847. doi: 10.1099/mic.0.2008/021279-0.
    1. Kim JJ, Nottingham LK, Sin JI, Tsai A, Morrison L, Oh J, Dang K, Hu Y, Kazahaya K, Bennett M, Dentchev T, Wilson DM, Chalian AA, Boyer JD, Agadjanyan MG, Weiner DB. CD8 positive T cells influence antigen-specific immune responses through the expression of chemokines. J Clin Invest. 1998;102:1112–1124. doi: 10.1172/JCI3986.
    1. Wilkinson PC, Newman I. Identification of IL-8 as a locomotor attractant for activated human lymphocytes in mononuclear cell cultures with anti-CD3 or purified protein derivative of Mycobacterium tuberculosis. J Immunol. 1992;149:2689–2694.
    1. Lee CG, Jenkins NA, Gilbert DJ, Copeland NG, O’Brien WE. Cloning and analysis of gene regulation of a novel LPS-inducible cDNA. Immunogenetics. 1995;41:263–270. doi: 10.1007/BF00172150.
    1. Cho H, Proll SC, Szretter KJ, Katze MG, Gale M, Jr, Diamond MS. Differential innate immune response programs in neuronal subtypes determine susceptibility to infection in the brain by positive-stranded RNA viruses. Nat Med. 2013;19:458–464. doi: 10.1038/nm.3108.
    1. Li LF, Yu J, Zhang Y, Yang Q, Li Y, Zhang L, Wang J, Li S, Luo Y, Sun Y, Qiu HJ. Interferon-inducible oligoadenylate synthetase-like protein acts as an antiviral effector against classical swine fever virus via the MDA5-mediated type i interferon-signaling pathway. J Virol. 2017;91:e01514–e01516.
    1. Dhar J, Cuevas RA, Goswami R, Zhu J, Sarkar SN, Barik S. 2′-5′-oligoadenylate synthetase-like protein inhibits respiratory syncytial virus replication and is targeted by the viral nonstructural protein 1. J Virol. 2015;89:10115–10119. doi: 10.1128/JVI.01076-15.
    1. Ishibashi M, Wakita T, Esumi M. 2′,5′-Oligoadenylate synthetase-like gene highly induced by hepatitis C virus infection in human liver is inhibitory to viral replication in vitro. Biochem Biophys Res Commun. 2010;392:397–402. doi: 10.1016/j.bbrc.2010.01.034.
    1. Steiner C, Muller M, Baniahmad A, Renkawitz R. Lysozyme gene activity in chicken macrophages is controlled by positive and negative regulatory elements. Nucleic Acids Res. 1987;15:4163–4178. doi: 10.1093/nar/15.10.4163.
    1. Ragland SA, Criss AK. From bacterial killing to immune modulation: recent insights into the functions of lysozyme. PLoS Pathog. 2017;13:e1006512. doi: 10.1371/journal.ppat.1006512.
    1. Lee-Huang S, Huang PL, Sun Y, Huang PL, Kung HF, Blithe DL, Chen HC. Lysozyme and RNases as anti-HIV components in beta-core preparations of human chorionic gonadotropin. Proc Natl Acad Sci U S A. 1999;96:2678–2681. doi: 10.1073/pnas.96.6.2678.
    1. Lee-Huang S, Maiorov V, Huang PL, Ng A, Lee HC, Chang YT, Kallenbach N, Huang PL, Chen HC. Structural and functional modeling of human lysozyme reveals a unique nonapeptide, HL9, with anti-HIV activity. Biochemistry. 2005;44:4648–4655. doi: 10.1021/bi0477081.
    1. Sundaram K, Rahman MA, Mitra S, Knoell DL, Woodiga SA, King SJ, Wewers MD. IκBζ regulates human monocyte pro-inflammatory responses induced by Streptococcus pneumoniae. PLoS One. 2016;11:e0161931. doi: 10.1371/journal.pone.0161931.
    1. Tsang J, Chain BM, Miller RF, Webb BL, Barclay W, Towers GJ, Katz DR, Noursadeghi M. HIV-1 infection of macrophages is dependent on evasion of innate immune cellular activation. AIDS. 2009;23:2255–2263. doi: 10.1097/QAD.0b013e328331a4ce.
    1. Heim MH. The Jak-STAT pathway: specific signal transduction from the cell membrane to the nucleus. Eur J Clin Invest. 1996;26:1–12. doi: 10.1046/j.1365-2362.1996.103248.x.
    1. Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol. 2007;7:454–465. doi: 10.1038/nri2093.
    1. Kubo M, Hanada T, Yoshimura A. Suppressors of cytokine signaling and immunity. Nat Immunol. 2003;4:1169–1176. doi: 10.1038/ni1012.
    1. Akhtar LN, Qin H, Muldowney MT, Yanagisawa LL, Kutsch O, Clements JE, Benveniste EN. Suppressor of cytokine signaling 3 inhibits antiviral IFN-beta signaling to enhance HIV-1 replication in macrophages. J Immunol. 2010;185:2393–2404. doi: 10.4049/jimmunol.0903563.
    1. Kedzierski L, Tate MD, Hsu AC, Kolesnik TB, Linossi EM, Dagley L, Dong Z, Freeman S, Infusini G, Starkey MR, Bird NL, Chatfield SM, Babon JJ, Huntington N, Belz G, Webb A, Wark PA, Nicola NA, Xu J, Kedzierska K, Hansbro PM, Nicholson SE. Suppressor of cytokine signaling (SOCS)5 ameliorates influenza infection via inhibition of EGFR signaling. eLife. 2017;6:e20444. doi: 10.7554/eLife.20444.
    1. Chiba T, Inoko H, Kimura M, Sato T. Role of nuclear IκBs in inflammation regulation. Biomol Concepts. 2013;4:187–196. doi: 10.1515/bmc-2012-0039.
    1. Parvatiyar K, Harhaj EW. Regulation of inflammatory and antiviral signaling by A20. Microbes Infect. 2011;13:209–215. doi: 10.1016/j.micinf.2010.11.003.
    1. Shembade N, Ma A, Harhaj EW. Inhibition of NF-kappaB signaling by A20 through disruption of ubiquitin enzyme complexes. Science. 2010;327:1135–1139. doi: 10.1126/science.1182364.
    1. Maelfait J, Roose K, Bogaert P, Sze M, Saelens X, Pasparakis M, Carpentier I, van Loo G, Beyaert R. A20 (Tnfaip3) deficiency in myeloid cells protects against influenza A virus infection. PLoS Pathog. 2012;8:e1002570. doi: 10.1371/journal.ppat.1002570.
    1. Maelfait J, Roose K, Vereecke L, Mc Guire C, Sze M, Schuijs MJ, Willart M, Ibanez LI, Hammad H, Lambrecht BN, Beyaert R, Saelens X, van Loo G. A20 deficiency in lung epithelial cells protects against influenza A virus infection. PLoS Pathog. 2016;12:e1005410. doi: 10.1371/journal.ppat.1005410.
    1. Carbonnelle-Puscian A, Copie-Bergman C, Baia M, Martin-Garcia N, Allory Y, Haioun C, Cremades A, Abd-Alsamad I, Farcet JP, Gaulard P, Castellano F, Molinier-Frenkel V. The novel immunosuppressive enzyme IL4I1 is expressed by neoplastic cells of several B-cell lymphomas and by tumor-associated macrophages. Leukemia. 2009;23:952–960. doi: 10.1038/leu.2008.380.
    1. Yue Y, Huang W, Liang J, Guo J, Ji J, Yao Y, Zheng M, Cai Z, Lu L, Wang J. IL4I1 is a novel regulator of M2 macrophage polarization that can inhibit T cell activation via l-tryptophan and arginine depletion and IL-10 production. PLoS One. 2015;10:e0142979. doi: 10.1371/journal.pone.0142979.
    1. Lin YT, Wu YH, Tseng CK, Lin CK, Chen WC, Hsu YC, Lee JC. Green tea phenolic epicatechins inhibit hepatitis C virus replication via cycloxygenase-2 and attenuate virus-induced inflammation. PLoS One. 2013;8:e54466. doi: 10.1371/journal.pone.0054466.
    1. Yue X, Yang F, Yang Y, Mu Y, Sun W, Li W, Xu D, Wu J, Zhu Y. Induction of cyclooxygenase-2 expression by hepatitis B virus depends on demethylation-associated recruitment of transcription factors to the promoter. Virol J. 2011;8:118. doi: 10.1186/1743-422X-8-118.
    1. Lin CK, Tseng CK, Wu YH, Liaw CC, Lin CY, Huang CH, Chen YH, Lee JC. Cyclooxygenase-2 facilitates dengue virus replication and serves as a potential target for developing antiviral agents. Sci Rep. 2017;7:44701. doi: 10.1038/srep44701.
    1. Zhu H, Cong JP, Yu D, Bresnahan WA, Shenk TE. Inhibition of cyclooxygenase 2 blocks human cytomegalovirus replication. Proc Natl Acad Sci U S A. 2002;99:3932–3937. doi: 10.1073/pnas.052713799.
    1. Matulova M, Rajova J, Vlasatikova L, Volf J, Stepanova H, Havlickova H, Sisak F, Rychlik I. Characterization of chicken spleen transcriptome after infection with Salmonella enterica serovar Enteritidis. PLoS One. 2012;7:e48101. doi: 10.1371/journal.pone.0048101.
    1. Di Marco E, Sessarego N, Zerega B, Cancedda R, Cancedda FD. Inhibition of cell proliferation and induction of apoptosis by ExFABP gene targeting. J Cell Physiol. 2003;196:464–473. doi: 10.1002/jcp.10310.
    1. Smith AJ, Li Q, Wietgrefe SW, Schacker TW, Reilly CS, Haase AT. Host genes associated with HIV-1 replication in lymphatic tissue. J Immunol. 2010;185:5417–5424. doi: 10.4049/jimmunol.1002197.
    1. Kumar A, Abbas W, Herbein G. HIV-1 latency in monocytes/macrophages. Viruses. 2014;6:1837–1860. doi: 10.3390/v6041837.

Source: PubMed

3
订阅