Multiorgan proteomic analysis of infected animal models predict potential host factors for chikungunya virus

Dongdong Lin , Cong Tang , Junbin Wang , Yun Yang , Hao Yang , Yanan Zhou , Wenhai Yu , Bai Li , Qing Huang , Haixuan Wang , Ran An , Xiaoming Liang , Yuhuan Yan , Longhai Yuan , Xuena Du , Yuxia Yuan , Yanwen Li , Shuaiyao Lu

MedComm ›› 2025, Vol. 6 ›› Issue (1) : e70013

PDF
MedComm ›› 2025, Vol. 6 ›› Issue (1) : e70013 DOI: 10.1002/mco2.70013
ORIGINAL ARTICLE

Multiorgan proteomic analysis of infected animal models predict potential host factors for chikungunya virus

Author information +
History +
PDF

Abstract

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that is primarily known for causing severe joint and muscle symptoms, but its pathological effects have extended beyond these tissues. In this study, we conducted a comprehensive proteomic analysis across various organs in rodent and nonhuman primate models to investigate CHIKV’s impact on organs beyond joints and muscles and to identify key host factors involved in its pathogenesis. Our findings reveal significant species-specific similarities and differences in immune responses and metabolic regulation, with proteins like Interferon-Stimulated Gene 15 (ISG15) and Retinoic Acid-Inducible Gene I (RIG-I) playing crucial roles in the anti-CHIKV defense. We observed upregulated and downregulated metabolic status in CHIKV-infected rhesus monkeys and mice, respectively. Additionally, we identified host factors such as S100 Calcium-Binding Protein A8/A9 (S100A8/A9), Voltage-Dependent Anion Channel 1/2 (VDAC1/2), Complement Component 3 (C3), Apoptosis-Inducing Factor Mitochondria-Associated 1 (AIFM1), Endothelial Cell-Specific Chemotaxis Regulator (ECSCR), and Kininogen 1 (KNG1) that may contribute to CHIKV-induced inflammation and hemorrhage. These insights put emphases on the importance of understanding CHIKV’s impact on organs beyond joints and muscles, providing potential therapeutic targets and enhancing our understanding of CHIKV pathogenesis. This research underscores the need for appropriate animal models in CHIKV studies and informs the development of targeted therapies to address its systemic effects.

Keywords

animal models / chikungunya virus / host factors / multiorgan proteomics / pathogenesis / targeted therapy

Cite this article

Download citation ▾
Dongdong Lin, Cong Tang, Junbin Wang, Yun Yang, Hao Yang, Yanan Zhou, Wenhai Yu, Bai Li, Qing Huang, Haixuan Wang, Ran An, Xiaoming Liang, Yuhuan Yan, Longhai Yuan, Xuena Du, Yuxia Yuan, Yanwen Li, Shuaiyao Lu. Multiorgan proteomic analysis of infected animal models predict potential host factors for chikungunya virus. MedComm, 2025, 6(1): e70013 DOI:10.1002/mco2.70013

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Kumar R, Ahmed S, Parray HA, Das S. Chikungunya and arthritis: an overview. Travel Med Infect Dis. 2021; 44: 102168.

[2]

Kim AS, Diamond MS. A molecular understanding of alphavirus entry and antibody protection. Nat Rev Microbiol. 2023; 21(6): 396-407.

[3]

Lum F-M, Ng LFP. Cellular and molecular mechanisms of chikungunya pathogenesis. Antiviral Research. 2015; 120: 165-174.

[4]

Bala Murugan S, Sathishkumar R. Chikungunya infection: a potential re-emerging global threat. Asian Pacific Journal of Tropical Medicine. 2016; 9(10): 933-937.

[5]

Song H, Zhao Z, Chai Y, et al. Molecular basis of arthritogenic alphavirus receptor MXRA8 binding to chikungunya virus envelope protein. Cell. 2019; 177(7): 1714-1724.

[6]

Alonso-Palomares Luis A, Moreno-García M, Lanz-Mendoza H, Salazar Ma I. Molecular basis for arbovirus transmission by aedes aegypti mosquitoes. Intervirology. 2019; 61(6): 255-264.

[7]

Khongwichit S, Chansaenroj J, Chirathaworn C, Poovorawan Y. Chikungunya virus infection: molecular biology, clinical characteristics, and epidemiology in Asian countries. J Biomed Sci. 2021; 28(1): 84.

[8]

Kharwadkar S, Herath N. Clinical manifestations of dengue, Zika and chikungunya in the Pacific Islands: a systematic review and meta-analysis. Reviews in Medical Virology. 2024; 34(2): e2521.

[9]

Bartholomeeusen K, Daniel M, LaBeaud DA, et al. Chikungunya fever. Nature Reviews Disease Primers. 2023; 9(1): 17.

[10]

Torales M, Beeson A, Grau L, et al. Notes from the field: chikungunya outbreak—Paraguay, 2022–2023. MMWR Morb Mortal Wkly Rep. 2023; 72(23): 636-638.

[11]

Suhrbier A. Rheumatic manifestations of chikungunya: emerging concepts and interventions. Nature Reviews Rheumatology. 2019; 15(10): 597-611.

[12]

Nsoesie EO, Kraemer MU, Golding N, et al. Global distribution and environmental suitability for chikungunya virus, 1952 to 2015. Euro Surveill. 2016; 21(20): 10.

[13]

de Souza WM, Ribeiro GS, de Lima STS, et al. Chikungunya: a decade of burden in the Americas. Lancet Reg Health Am. 2024; 30: 100673.

[14]

Tjaden NB, Suk JE, Fischer D, Thomas SM, Beierkuhnlein C, Semenza JC. Modelling the effects of global climate change on Chikungunya transmission in the 21st century. Scientific Reports. 2017; 7(1): 3813.

[15]

Musso D, Rodriguez-Morales AJ, Levi JE, Cao-Lormeau V-M, Gubler DJ. Unexpected outbreaks of arbovirus infections: lessons learned from the Pacific and tropical America. The Lancet Infectious Diseases. 2018; 18(11): e355-e361.

[16]

Wang M, Wang L, Leng P, Guo J, Zhou H. Drugs targeting structural and nonstructural proteins of the chikungunya virus: a review. International Journal of Biological Macromolecules. 2024; 262: 129949.

[17]

August A, Attarwala HZ, Himansu S, et al. A phase 1 trial of lipid-encapsulated mRNA encoding a monoclonal antibody with neutralizing activity against Chikungunya virus. Nature Medicine. 2021; 27(12): 2224-2233.

[18]

Shi J, Su Z, Fan Z, et al. Extensive evolution analysis of the global chikungunya virus strains revealed the origination of CHIKV epidemics in Pakistan in 2016. Virologica Sinica. 2017; 32(6): 520-532.

[19]

Phadungsombat J, Imad HA, Nakayama EE, et al. Spread of a novel Indian Ocean lineage carrying E1-K211E/E2-V264A of chikungunya virus east/central/south African genotype across the Indian Subcontinent, Southeast Asia, and Eastern Africa. Microorganisms. 2022; 10(2): 354.

[20]

Winkler ES, Shrihari S. The intestinal microbiome restricts alphavirus infection and dissemination through a bile acid-type I IFN signaling axis. Cell. 2020; 182(4): 901-918.

[21]

Wilson JA, Prow NA, Schroder WA, et al. RNA-Seq analysis of chikungunya virus infection and identification of granzyme A as a major promoter of arthritic inflammation. PLoS Pathog. 2017; 13(2): e1006155.

[22]

Nath P, Chauhan NR, Jena KK, et al. Inhibition of IRGM establishes a robust antiviral immune state to restrict pathogenic viruses. EMBO reports. 2021; 22(11): e52948.

[23]

McNab F, Mayer-Barber K, Sher A, Wack A, O’Garra A. Type I interferons in infectious disease. Nat Rev Immunol. 2015; 15(2): 87-103.

[24]

Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010; 140(6): 805-820.

[25]

Her Z, Teng TS, Tan JJ, et al. Loss of TLR3 aggravates CHIKV replication and pathology due to an altered virus-specific neutralizing antibody response. EMBO Mol Med. 2015; 7(1): 24-41.

[26]

Garcia G Jr., Irudayam JI, Jeyachandran AV, et al. Innate immune pathway modulator screen identifies STING pathway activation as a strategy to inhibit multiple families of arbo and respiratory viruses. Cell Rep Med. 2023; 4(5): 101024.

[27]

Cardona-Ospina JA, Villamil-Gómez WE, Jimenez-Canizales CE, Castañeda-Hernández DM, Rodríguez-Morales AJ. Estimating the burden of disease and the economic cost attributable to chikungunya, Colombia, 2014. Trans R Soc Trop Med Hyg. 2015; 109(12): 793-802.

[28]

Teo TH, Chan YH, Lee WW, et al. Fingolimod treatment abrogates chikungunya virus-induced arthralgia. Sci Transl Med. 2017; 9(375): eaal1333.

[29]

Pott F, Postmus D, Brown RJP, et al. Single-cell analysis of arthritogenic alphavirus-infected human synovial fibroblasts links low abundance of viral RNA to induction of innate immunity and arthralgia-associated gene expression. Emerg Microbes Infect. 2021; 10(1): 2151-2168.

[30]

Agrawal M, Pandey N, Rastogi M, Dogra S, Singh SK. Chikungunya virus modulates the miRNA expression patterns in human synovial fibroblasts. J Med Virol. 2020; 92(2): 139-148.

[31]

Neupane B, Acharya D, Nazneen F, Gonzalez-Fernandez G, Flynt AS, Bai F. Interleukin-17A facilitates chikungunya virus infection by inhibiting IFN-α2 expression. Front Immunol. 2020; 11: 588382.

[32]

Meertens L, Hafirassou ML, Couderc T, et al. FHL1 is a major host factor for chikungunya virus infection. Nature. 2019; 574(7777): 259-263.

[33]

De Caluwé L, Ariën KK, Bartholomeeusen K. Host factors and pathways involved in the entry of mosquito-borne alphaviruses. Trends Microbiol. 2021; 29(7): 634-647.

[34]

de Souza WM, Fumagalli MJ, de Lima STS, et al. Pathophysiology of chikungunya virus infection associated with fatal outcomes. Cell Host Microbe. 2024; 32(4): 606-622.

[35]

Ganesan VK, Duan B, Reid SP. Chikungunya virus: pathophysiology, mechanism, and modeling. Viruses. 2017; 9(12): 368.

[36]

Haese NN, Broeckel RM, Hawman DW, Heise MT, Morrison TE, Streblow DN. Animal models of chikungunya virus infection and disease. The Journal of Infectious Diseases. 2016; 214(5): S482-S487.

[37]

Couderc T, Chrétien F, Schilte C, 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(2): e29.

[38]

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(2): 103-112.

[39]

Messaoudi I, Vomaske J, Totonchy T, et al. Chikungunya virus infection results in higher and persistent viral replication in aged rhesus macaques due to defects in anti-viral immunity. PLoS Negl Trop Dis. 2013; 7(7): e2343.

[40]

Chen C-I, Clark DC, Pesavento P, et al. Comparative Pathogenesis of Epidemic and Enzootic Chikungunya Viruses in a Pregnant Rhesus Macaque Model. The American Society of Tropical Medicine and Hygiene; 2010: 1249-1258.

[41]

Budayeva HG, Kirkpatrick DS. Monitoring protein communities and their responses to therapeutics. Nat Rev Drug Discov. 2020; 19(6): 414-426.

[42]

Li X, Wang W, Chen J. Recent progress in mass spectrometry proteomics for biomedical research. Sci China Life Sci. 2017; 60(10): 1093-1113.

[43]

Silva LA, Dermody TS. Chikungunya virus: epidemiology, replication, disease mechanisms, and prospective intervention strategies. J Clin Invest. 2017; 127(3): 737-749.

[44]

Traverse EM, Millsapps EM, Underwood EC, Hopkins HK, Young M, Barr KL. Chikungunya immunopathology as it presents in different organ systems. Viruses. 2022; 14(8): 1786.

[45]

Cerqueira-Silva T, Pescarini JM, Cardim LL, et al. Risk of death following chikungunya virus disease in the 100 Million Brazilian Cohort, 2015–18: a matched cohort study and self-controlled case series. Lancet Infect Dis. 2024; 24(5): 504-513.

[46]

Sharp TM, Keating MK, Shieh WJ, et al. Clinical characteristics, histopathology, and tissue immunolocalization of chikungunya virus antigen in fatal cases. Clin Infect Dis. 2021; 73(2): e345-e354.

[47]

Hoarau JJ, Jaffar Bandjee MC, Krejbich Trotot P, et al. Persistent chronic inflammation and infection by Chikungunya arthritogenic alphavirus in spite of a robust host immune response. J Immunol. 2010; 184(10): 5914-5927.

[48]

Kumar S, Jaffar-Bandjee MC, Giry C, et al. Mouse macrophage innate immune response to Chikungunya virus infection. Virol J. 2012; 9: 313.

[49]

Zuo W, He D, Liang C, et al. The persistence of SARS-CoV-2 in tissues and its association with long COVID symptoms: a cross-sectional cohort study in China. The Lancet Infectious Diseases. 2024; 24(8): 845-855.

[50]

Perng YC, Lenschow DJ. ISG15 in antiviral immunity and beyond. Nat Rev Microbiol. 2018; 16(7): 423-439.

[51]

Wu YY, Xing J, Li XF, Yang YL, Shao H, Li J. Roles of interferon induced protein with tetratricopeptide repeats (IFIT) family in autoimmune disease. Autoimmun Rev. 2023; 22(11): 103453.

[52]

Rehwinkel J, Gack MU. RIG-I-like receptors: their regulation and roles in RNA sensing. Nat Rev Immunol. 2020; 20(9): 537-551.

[53]

Yuen MF, Chen CY, Liu CJ, et al. A phase 2, open-label, randomized, multiple-dose study evaluating Inarigivir in treatment-naïve patients with chronic hepatitis B. Liver Int. 2023; 43(1): 77-89.

[54]

Zhou S, Wang L. Unraveling the structural and chemical features of biological short hydrogen bonds. Chem Sci. 2019; 10(33): 7734-7745.

[55]

Gopal R, Monin L, Torres D, et al. S100A8/A9 proteins mediate neutrophilic inflammation and lung pathology during tuberculosis. Am J Respir Crit Care Med. 2013; 188(9): 1137-1146.

[56]

Gehrig S, Duerr J, Weitnauer M, et al. Lack of neutrophil elastase reduces inflammation, mucus hypersecretion, and emphysema, but not mucus obstruction, in mice with cystic fibrosis-like lung disease. Am J Respir Crit Care Med. 2014; 189(9): 1082-1092.

[57]

Lodge KM, Vassallo A, Liu B, et al. Hypoxia increases the potential for neutrophil-mediated endothelial damage in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2022; 205(8): 903-916.

[58]

Xian H, Watari K, Sanchez-Lopez E, et al. Oxidized DNA fragments exit mitochondria via mPTP-and VDAC-dependent channels to activate NLRP3 inflammasome and interferon signaling. Immunity. 2022; 55(8): 1370-1385.

[59]

Baik SH, Ramanujan VK, Becker C, Fett S, Underhill DM, Wolf AJ. Hexokinase dissociation from mitochondria promotes oligomerization of VDAC that facilitates NLRP3 inflammasome assembly and activation. Sci Immunol. 2023; 8(84): eade7652.

[60]

Jackson DN, Panopoulos M, Neumann WL, et al. Mitochondrial dysfunction during loss of prohibitin 1 triggers Paneth cell defects and ileitis. Gut. 2020; 69(11): 1928-1938.

[61]

Carvalho MOS, Souza ALCS, Carvalho MB, et al. Evaluation of alpha-1 antitrypsin levels and SERPINA1 gene polymorphisms in sickle cell disease. Frontiers in Immunology. 2017: 8. Original Research..

[62]

Greene CM, Marciniak SJ, Teckman J, et al. α1-Antitrypsin deficiency. Nature Reviews Disease Primers. 2016; 2(1): 16051.

[63]

Sharp TM, Keating MK, Shieh W-J, et al. Clinical characteristics, histopathology, and tissue immunolocalization of chikungunya virus antigen in fatal cases. Clinical Infectious Diseases. 2021; 73(2): e345-e354.

[64]

Holze C, Michaudel C, Mackowiak C, et al. Oxeiptosis, a ROS-induced caspase-independent apoptosis-like cell-death pathway. Nat Immunol. 2018; 19(2): 130-140.

[65]

Wang T, Liu H, Itoh K, et al. C9orf72 regulates energy homeostasis by stabilizing mitochondrial complex I assembly. Cell Metabolism. 2021; 33(3): 531-546.

[66]

Matsumoto Y, Ichikawa T, Kurozumi K, et al. Annexin A2-STAT3-Oncostatin M receptor axis drives phenotypic and mesenchymal changes in glioblastoma. Acta Neuropathol Commun. 2020; 8(1): 42.

[67]

Madureira PA, Surette AP, Phipps KD, Taboski MA, Miller VA, Waisman DM. The role of the annexin A2 heterotetramer in vascular fibrinolysis. Blood. 2011; 118(18): 4789-4797.

[68]

Liao Y, Feng J, Sun W, et al. CIRP promotes the progression of non-small cell lung cancer through activation of Wnt/β-catenin signaling via CTNNB1. J Exp Clin Cancer Res. 2021; 40(1): 275.

[69]

Amini-Nik S, Cambridge E, Yu W, et al. β-Catenin-regulated myeloid cell adhesion and migration determine wound healing. J Clin Invest. 2014; 124(6): 2599-2610.

[70]

Schmitt MMN, Megens RTA, Zernecke A, et al. Endothelial junctional adhesion molecule-a guides monocytes into flow-dependent predilection sites of atherosclerosis. Circulation. 2014; 129(1): 66-76.

[71]

Dib F, Quéméner A, Bayart S, et al. Biological, clinical features and modelling of heterozygous variants of glycoprotein Ib platelet subunit alpha (GP1BA) and glycoprotein Ib platelet subunit beta (GP1BB) genes responsible for constitutional thrombocytopenia. Br J Haematol. 2022; 199(5): 744-753.

[72]

Brown TJ, Barrett N, Meng H, et al. Nonsteroidal anti-inflammatory drugs as a targeted therapy for bone marrow failure in Ghosal hematodiaphyseal dysplasia. Blood. 2023; 141(13): 1553-1559.

[73]

Ikeda K, Nakano R, Uraoka M, et al. Identification of ARIA regulating endothelial apoptosis and angiogenesis by modulating proteasomal degradation of cIAP-1 and cIAP-2. Proceedings of the National Academy of Sciences. 2009; 106(20): 8227-8232.

[74]

Akakabe Y, Koide M, Kitamura Y, et al. Ecscr regulates insulin sensitivity and predisposition to obesity by modulating endothelial cell functions. Nature Communications. 2013; 4(1): 2389.

[75]

Marceau F. Drugs of the Kallikrein–Kinin system: an overview. Drugs and Drug Candidates. 2023; 2(3): 538-553.

[76]

Mohammadi K, Shafie D, Ghomashi N, Abdolizadeh A, Sadeghpour M. Kinin-kallikrein system: new perspectives in heart failure. Heart Fail Rev. 2024; 29(3): 729-737.

[77]

Silva LM, Doyle AD, Greenwell-Wild T, et al. Fibrin is a critical regulator of neutrophil effector function at the oral mucosal barrier. Science. 2021; 374(6575): eabl5450.

[78]

Liu X, Zhao R, Ding Q, Yao X, Tsang SY. TRPC7 regulates the electrophysiological functions of embryonic stem cell-derived cardiomyocytes. Stem Cell Research & Therapy. 2021; 12(1): 262.

[79]

Roques P, Ljungberg K, Kümmerer BM, et al. Attenuated and vectored vaccines protect nonhuman primates against Chikungunya virus. JCI Insight. 2017; 2(6): e83527.

[80]

Weber C, Büchner SM, Schnierle BS. A small antigenic determinant of the Chikungunya virus E2 protein is sufficient to induce neutralizing antibodies which are partially protective in mice. PLoS Negl Trop Dis. 2015; 9(4): e0003684.

[81]

Roy CJ, Adams AP, Wang E, et al. Chikungunya vaccine candidate is highly attenuated and protects nonhuman primates against telemetrically monitored disease following a single dose. J Infect Dis. 2014; 209(12): 1891-1899.

[82]

Kam Y-W, Lee WWL, Simarmata D, et al. Unique epitopes recognized by antibodies induced in chikungunya virus-infected non-human primates: implications for the study of immunopathology and vaccine development. PLOS ONE. 2014; 9(4): e95647.

[83]

Chakradhar S. Disease in three dimensions: tissue engineering takes on infectious disease. Nature Medicine. 2017; 23(1): 2-4.

[84]

Mills M, Estes MK. Physiologically relevant human tissue models for infectious diseases. Drug Discov Today. 2016; 21(9): 1540-1552.

[85]

Fontaine Krystal A, Sanchez Erica L, Camarda R, Lagunoff M. Dengue virus induces and requires glycolysis for optimal replication. Journal of Virology. 2015; 89(4): 2358-2366.

[86]

Thaker SK, Chapa T. Differential metabolic reprogramming by zika virus promotes cell death in human versus mosquito cells. Cell Metabolism. 2019; 29(5): 1206-1216.

[87]

Smallwood HS, Duan S, Morfouace M, et al. Targeting metabolic reprogramming by influenza infection for therapeutic intervention. Cell Reports. 2017; 19(8): 1640-1653.

[88]

Heaton NS, Perera R, Berger KL, et al. Dengue virus nonstructural protein 3 redistributes fatty acid synthase to sites of viral replication and increases cellular fatty acid synthesis. Proceedings of the National Academy of Sciences. 2010; 107(40): 17345-17350.

[89]

Girdhar K, Powis A, Raisingani A, et al. Viruses and metabolism: the effects of viral infections and viral insulins on host metabolism. Annu Rev Virol. 2021; 8(1): 373-391.

[90]

El Safadi D, Paulo-Ramos A, Hoareau M, et al. The influence of metabolism on immune response: a journey to understand immunometabolism in the context of viral infection. Viruses. 2023; 15(12): 2399.

[91]

Zhao C, Sridharan H, Chen R, Baker DP, Wang S, Krug RM. Influenza B virus non-structural protein 1 counteracts ISG15 antiviral activity by sequestering ISGylated viral proteins. Nat Commun. 2016; 7: 12754.

[92]

Singh PK, Singh S, Farr D, Kumar A. Interferon-stimulated gene 15 (ISG15) restricts Zika virus replication in primary human corneal epithelial cells. Ocul Surf. 2019; 17(3): 551-559.

[93]

Sarkar L, Liu G, Gack MU. ISG15: its roles in SARS-CoV-2 and other viral infections. Trends in Microbiology. 2023; 31(12): 1262-1275.

[94]

Noval MG, Spector SN, Bartnicki E, et al. MAVS signaling is required for preventing persistent chikungunya heart infection and chronic vascular tissue inflammation. Nat Commun. 2023; 14(1): 4668.

[95]

Onomoto K, Onoguchi K, Yoneyama M. Regulation of RIG-I-like receptor-mediated signaling: interaction between host and viral factors. Cell Mol Immunol. 2021; 18(3): 539-555.

[96]

Allen Irving C, Moore Chris B, Schneider M, et al. NLRX1 protein attenuates inflammatory responses to infection by interfering with the RIG-I-MAVS and TRAF6-NF-κB signaling pathways. Immunity. 2011; 34(6): 854-865.

[97]

Moore CB, Bergstralh DT, Duncan JA, et al. NLRX1 is a regulator of mitochondrial antiviral immunity. Nature. 2008; 451(7178): 573-577.

[98]

Xu S, Xu L-G, Yuan C, et al. FKBP8 inhibits virus-induced RLR-VISA signaling. Journal of Medical Virology. 2018; 91: 482-492.

[99]

Lei Y, Wen H, Yu Y, et al. The mitochondrial proteins NLRX1 and TUFM form a complex that regulates type I interferon and autophagy. Immunity. 2012; 36(6): 933-946.

[100]

Li XD, Sun L, Seth RB, Pineda G, Chen ZJ. Hepatitis C virus protease NS3/4A cleaves mitochondrial antiviral signaling protein off the mitochondria to evade innate immunity. Proc Natl Acad Sci U S A. 2005; 102(49): 17717-17722.

[101]

Qian S, Fan W, Liu T, et al. Seneca valley virus suppresses host type i interferon production by targeting adaptor proteins MAVS, TRIF, and TANK for cleavage. Journal of Virology. 2017; 91(16): e00823-17.10.1128/jvi.00823-17

[102]

Choi HJ, Park A, Kang S, et al. Human cytomegalovirus-encoded US9 targets MAVS and STING signaling to evade type I interferon immune responses. Nat Commun. 2018; 9(1): 125.

[103]

Mukherjee A, Morosky SA, Delorme-Axford E, et al. The coxsackievirus B 3Cpro protease cleaves MAVS and TRIF to attenuate host type i interferon and apoptotic signaling. PLOS Pathogens. 2011; 7(3): e1001311.

[104]

Gack MU, Shin YC, Joo CH, et al. TRIM25 RING-finger E3 ubiquitin ligase is essential for RIG-I-mediated antiviral activity. Nature. 2007; 446(7138): 916-920.

[105]

Zeng W, Sun L, Jiang X, et al. Reconstitution of the RIG-I pathway reveals a signaling role of unanchored polyubiquitin chains in innate immunity. Cell. 2010; 141(2): 315-330.

[106]

Zhu J, Zhang Y, Ghosh A, et al. Antiviral activity of human OASL protein is mediated by enhancing signaling of the RIG-I RNA sensor. Immunity. 2014; 40(6): 936-948.

[107]

Ng LFP, Chow A, Sun Y-J, et al. IL-1β IL-6, and RANTES as biomarkers of chikungunya severity. PLOS ONE. 2009; 4(1): e4261.

[108]

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(1): 115-123.

[109]

Lum F-M, Chan Y-H, Teo T-H, et al. Crosstalk between CD64+MHCII+macrophages and CD4+T cells drives joint pathology during chikungunya. EMBO Molecular Medicine. 2024; 16(3): 641-663663.

[110]

Zhou W, Cao X, Xu Q, Qu J, Sun Y. The double-edged role of neutrophil heterogeneity in inflammatory diseases and cancers. MedComm. 2023; 4(4): e325. (2020).

[111]

Mellor LF, Gago-Lopez N, Bakiri L, et al. Keratinocyte-derived S100A9 modulates neutrophil infiltration and affects psoriasis-like skin and joint disease. Ann Rheum Dis. 2022; 81(10): 1400-1408.

[112]

Inciarte-Mundo J, Frade-Sosa B, Sanmartí R. From bench to bedside: calprotectin (S100A8/S100A9) as a biomarker in rheumatoid arthritis. Front Immunol. 2022; 13: 1001025.

[113]

Trzybulska D, Olewicz-Gawlik A, Graniczna K, et al. Quantitative analysis of elastase and cathepsin G mRNA levels in peripheral blood CD14(+) cells from patients with rheumatoid arthritis. Cell Immunol. 2014; 292(1-2): 40-44.

[114]

Schober P, Boer C, Schwarte LA. Correlation coefficients: appropriate use and interpretation. Anesth Analg. 2018; 126(5): 1763-1768.

[115]

Akoglu H. User’s guide to correlation coefficients. Turkish Journal of Emergency Medicine. 2018; 18(3): 91-93.

RIGHTS & PERMISSIONS

2025 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

140

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/