Acessibilidade / Reportar erro

Influence of host genetic polymorphisms involved in immune response and their role in the development of Chikungunya disease: a review

Abstract

Chikungunya virus (CHIKV) is transmitted by the bite of infected mosquitoes and can cause significant pathogenicity in humans. Moreover, its importance has increased in the Americas since 2013. The primary vectors for viral delivery are the mosquito species Aedes aegypti and Aedes albopictus. Several factors, including host genetic variations and immune response against CHIKV, influence the outcomes of Chikungunya disease. This work aimed to gather information about different single nucleotide polymorphisms (SNPs) in genes that influence the host immune response during an infection by CHIKV. The viral characteristics, disease epidemiology, clinical manifestations, and immune response against CHIKV are also addressed. The main immune molecules related to this arboviral disease elucidated in this review are TLR3/7/8, DC-SIGN, HLA-DRB1/HLA-DQB1, TNF, IL1RN, OAS2/3, and CRP. Advances in knowledge about the genetic basis of the immune response during CHIKV infection are essential for expanding the understanding of disease pathophysiology, providing new genetic markers for prognosis, and identifying molecular targets for the development of new drug treatments.

Single nucleotide polymorphisms (SNPs); Chikungunya virus (CHIKV); Chikungunya disease; Immune response


Introduction

The chikungunya virus (CHIKV) is a member of the Alphavirus genus, belonging to the Togaviridae family, whose genetic material is constituted by a single strand of RNA of approximately 12 kb with positive polarity. This virus is the etiologic agent of Chikungunya disease (CHIK) and was first isolated in 1952-1953 during an epidemic in East Africa (Makonde Plateau-Tanzania) (11. Naresh Kumar CVM, Sai Gopal DVR. Reemergence of Chikungunya virus in Indian subcontinent. Indian J Virol 2010; 21: 8-17, doi: 10.1007/s13337-010-0012-1.
https://doi.org/10.1007/s13337-010-0012-...
,22. Chaaithanya IK, Muruganandam N, Surya P, Anwesh M, Alagarasu K, Vijayachari P. Association of oligoadenylate synthetase gene cluster and DC-SIGN (CD209) gene polymorphisms with clinical symptoms in Chikungunya virus infection. DNA Cell Biol 2016; 35: 44-50, doi: 10.1089/dna.2015.2819.
https://doi.org/10.1089/dna.2015.2819...
). Between 1960 and 1990, CHIK outbreaks occurred sporadically in Africa and Asia, reappearing more frequently from 2000 onwards and spreading to other countries (33. Burt FJ, Chen W, Miner JJ, Lenschow DJ, Merits A, Schnettler E, et al. Chikungunya virus: an update on the biology and pathogenesis of this emerging pathogen. Lancet Infect Dis 2017; 17: e107-e117, doi: 10.1016/S1473-3099(16)30385-1.
https://doi.org/10.1016/S1473-3099(16)30...
). In 2014, CHIKV was responsible for causing a major epidemic in Brazil, mainly affecting the Northeast region (44. Weaver SC, Lecuit M. Chikungunya virus and the global spread of a mosquito-borne disease. N Engl J Med 2015; 372: 1231-1239, doi: 10.1056/NEJMra1406035.
https://doi.org/10.1056/NEJMra1406035...
). In the Americas alone, about 693,489 suspected cases and 37,480 confirmed cases of CHIK were reported in 2015 (55. Giménez-Richarte Á, de Salazar MO, Arbona C, Giménez-Richarte MP, Collado M, Fernández PL, et al. Prevalence of Chikungunya, Dengue and Zika viruses in blood donors: a systematic literature review and meta-analysis. Blood Transfus 2022; 20: 267-280, doi: 10.2450/2021.0106-21.
https://doi.org/10.2450/2021.0106-21...
,66. Ferreira JM, Santos LDS, Oliveira SP, Dos Santos BRC, Dos Santos ACM, de Moura EL, et al. Chikungunya virus infection outcome: a systematic review of host genetics. Immunol Invest 2021; 50: 58-79, doi: 10.1080/08820139.2020.1733011.
https://doi.org/10.1080/08820139.2020.17...
). In 2022, from January until November 23, 2022, a total of 362,021 cases and 77 deaths were reported. The majority of cases have been reported in Brazil (247,537) (77. ECDC (European Centre for Disease Prevention and Control). Chikungunya worldwide overview. Available from https://www.ecdc.eur opa.eu/en/chikungunya-monthly. Accessed Dec 05, 2022.
https://www.ecdc.eur opa.eu/en/chikungun...
). CHIKV infection may have different clinical presentations. The acute phase of Chikungunya is associated with various symptoms and characterized by the onset of high fever, polyarthritis, and maculopapular rash. The chronic phase is marked by arthritogenic manifestations lasting >3 months and the onset of rheumatic symptoms (88. Gasque P, Bandjee MCJ, Reyes MM, Viasus D. Chikungunya pathogenesis: from the clinics to the bench. J Infect Dis 2016; 214: S446-S448, doi: 10.1093/infdis/jiw362.
https://doi.org/10.1093/infdis/jiw362...
). After the bite of an infected mosquito, CHIKV is inoculated into the blood capillaries and dermis, where it infects resident skin cells such as keratinocytes and dermal fibroblasts, initiating the first cycle of viral replication (99. 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, doi: 10.1093/infdis/jiv049.
https://doi.org/10.1093/infdis/jiv049...
). The main sites of secondary infection are the muscles and joints, causing polyarthralgia/polyarthritis. The incubation period for CHIKV is usually 2 to 7 days in most cases (1010. Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines 2012; 11: 1087-1101, doi: 10.1586/erv.12.84.
https://doi.org/10.1586/erv.12.84...
). Numerous risk factors for developing severe disease in adult patients have been reported, including age and gender. Moreover, host genetic variation has a significant influence on the immune response and outcome of CHIKV infections. It modulates the disease, influences viral entry into cells, symptom onset and progression, and individual susceptibility or resistance to infection (1111. Dutta SK, Tripathi A. Association of toll-like receptor polymorphisms with susceptibility to Chikungunya virus infection. Virology 2017; 511: 207-213, doi: 10.1016/j.virol.2017.08.009.
https://doi.org/10.1016/j.virol.2017.08....
,1212. Mickienė A, Pakalnienė J, Nordgren J, Carlsson B, Hagbom M, Svensson L, et al. Polymorphisms in chemokine receptor 5 and Toll-like receptor 3 genes are risk factors for clinical tick-borne encephalitis in the Lithuanian population. PLoS One 2014; 9: e106798, doi: 10.1371/journal.pone.0106798.
https://doi.org/10.1371/journal.pone.010...
). Thus, the knowledge of the host profile of genes that influence or contribute to the outcome and susceptibility of CHIKV infection is an essential tool to elucidate the pathophysiology of this arboviral disease and its progression (22. Chaaithanya IK, Muruganandam N, Surya P, Anwesh M, Alagarasu K, Vijayachari P. Association of oligoadenylate synthetase gene cluster and DC-SIGN (CD209) gene polymorphisms with clinical symptoms in Chikungunya virus infection. DNA Cell Biol 2016; 35: 44-50, doi: 10.1089/dna.2015.2819.
https://doi.org/10.1089/dna.2015.2819...
,1313. Bucardo F, Reyes Y, Morales M, Briceão R, González F, Lundkvist Å, et al. Association of genetic polymorphisms in DC-SIGN, toll-like receptor 3, and tumor necrosis factor α genes and the Lewis-negative phenotype with Chikungunya infection and disease in Nicaragua. J Infect Dis 2021; 223: 278-286, doi: 10.1093/infdis/jiaa364.
https://doi.org/10.1093/infdis/jiaa364...
). Therefore, the present review aimed to identify in the literature the influence of host genetic variations on the immune response during CHIKV infection and the role of genetic polymorphisms on the outcome of the disease.

General characteristics of Chikungunya virus

CHIKV is an enveloped alphavirus with 60-70 nm in diameter. It belongs to the Togaviridae family, and the main vectors in the transmission of CHIKV are female mosquitoes of the Aedes genus, more specifically the Aedes aegypti and Aedes albopictus, which are widely distributed in endemic regions (44. Weaver SC, Lecuit M. Chikungunya virus and the global spread of a mosquito-borne disease. N Engl J Med 2015; 372: 1231-1239, doi: 10.1056/NEJMra1406035.
https://doi.org/10.1056/NEJMra1406035...
,55. Giménez-Richarte Á, de Salazar MO, Arbona C, Giménez-Richarte MP, Collado M, Fernández PL, et al. Prevalence of Chikungunya, Dengue and Zika viruses in blood donors: a systematic literature review and meta-analysis. Blood Transfus 2022; 20: 267-280, doi: 10.2450/2021.0106-21.
https://doi.org/10.2450/2021.0106-21...
).

CHIKV has an 11.8 kb-long single-stranded positive-sense RNA (1414. Metz SW, Geertsema C, Martina BE, Andrade P, Heldens JG, van Oers MM, et al. Functional processing and secretion of Chikungunya virus E1 and E2 glycoproteins in insect cells. Virol J 2011; 8: 1-12, doi: 10.1186/1743-422X-8-353.
https://doi.org/10.1186/1743-422X-8-353...
). The CHIKV genome contains two reading frames (open reading frame, ORF): the 5' ORF, translated from genomic RNA (49S) and coding for four nonstructural proteins (NSP1, NSP2, NSP3, and NSP4), and the 3' ORF, translated from a subgenomic RNA (26S) and coding for a polyprotein later cleaved into the capsid protein, two envelope proteins (E1 and E2), and two small peptides, E3 and 6K (1515. Jose J, Snyder JE, Kuhn RJ. A structural and functional perspective of alphavirus replication and assembly. Future Microbiol 2009; 4: 837-856, doi: 10.2217/fmb.09.59.
https://doi.org/10.2217/fmb.09.59...
,1616. Thiberville SD, Moyen N, Dupuis-Maguiraga L, Nougairede A, Gould EA, Roques P, et al. Chikungunya fever: epidemiology, clinical syndrome, pathogenesis and therapy. Antiviral Res 2013; 99: 345-370, doi: 10.1016/j.antiviral.2013.06.009.
https://doi.org/10.1016/j.antiviral.2013...
). E1 and E2 are surface glycoproteins, 439 and 423 amino acid-long, respectively (1717. Khan AH, Morita K, Parquet MDC, Hasebe F, Mathenge EGM, Igarashi A. Complete nucleotide sequence of Chikungunya virus and evidence for an internal polyadenylation site. J Gen Virol 2002; 83: 3075-3084, doi: 10.1099/0022-1317-83-12-3075.
https://doi.org/10.1099/0022-1317-83-12-...
,1818. Voss JE, Vaney MC, Duquerroy S, Vonrhein C, Girard-Blanc C, Crublet E, et al. Glycoprotein organization of Chikungunya virus particles revealed by X-ray crystallography. Nature 2010; 468: 709-712, doi: 10.1038/nature09555.
https://doi.org/10.1038/nature09555...
). E1 is responsible for the fusion of the viral membrane and the endosomal membrane, and E2 is involved in receptor binding and subsequent receptor-mediated endocytosis (1515. Jose J, Snyder JE, Kuhn RJ. A structural and functional perspective of alphavirus replication and assembly. Future Microbiol 2009; 4: 837-856, doi: 10.2217/fmb.09.59.
https://doi.org/10.2217/fmb.09.59...
,1919. Carvalho CAM, Silva JL, Oliveira AC, Gomes AMO. On the entry of an emerging arbovirus into host cells: Mayaro virus takes the highway to the cytoplasm through fusion with early endosomes and caveolae-derived vesicles. PeerJ 2017; 5: e3245, doi: 10.7717/peerj.3245.
https://doi.org/10.7717/peerj.3245...
). The 61-amino acid 6K protein increases cell permeability to monovalent cations and virion budding during infection (1515. Jose J, Snyder JE, Kuhn RJ. A structural and functional perspective of alphavirus replication and assembly. Future Microbiol 2009; 4: 837-856, doi: 10.2217/fmb.09.59.
https://doi.org/10.2217/fmb.09.59...
,1818. Voss JE, Vaney MC, Duquerroy S, Vonrhein C, Girard-Blanc C, Crublet E, et al. Glycoprotein organization of Chikungunya virus particles revealed by X-ray crystallography. Nature 2010; 468: 709-712, doi: 10.1038/nature09555.
https://doi.org/10.1038/nature09555...
). Transframe protein (TF) is produced as a result of the C-terminal extension of 6K protein (2020. Ramsey J, Mukhopadhyay S. Disentangling the frames, the state of research on the alphavirus 6K and TF proteins. Viruses 2017; 9: 228, doi: 10.3390/v9080228.
https://doi.org/10.3390/v9080228...
). Nonstructural proteins (NSP1-4) are responsible for viral replication machinery (2121. Tomar S, Hardy RW, Smith JL, Kuhn RJ. Catalytic core of alphavirus nonstructural protein nsP4 possesses terminal adenylyltransferase activity. J Virol 2006; 80: 9962-9969, doi: 10.1128/JVI.01067-06.
https://doi.org/10.1128/JVI.01067-06...
).

Four distinct CHIKV genotypes (or lineages) have already been identified and named based on their geographical distribution: i) the West African; ii) the East/Central/South African (ECSA); iii) the Asian, and iv) the Indian Ocean Lineage (IOL) (2222. Fabri AA, Rodrigues CDS, dos Santos CC, Chalhoub FLL, Sampaio SA, Faria NRC, et al. Co-circulation of two independent clades and persistence of chikv-ecsa genotype during epidemic waves in Rio de Janeiro, Southeast Brazil. Pathogens 2020; 9: 984, doi: 10.3390/pathogens9120984.
https://doi.org/10.3390/pathogens9120984...
,2323. Agarwal A, Dash PK, Singh AK, Sharma S, Gopalan N, Rao PVL, et al. Evidence of experimental vertical transmission of emerging novel ECSA genotype of Chikungunya virus in Aedes aegypti. PLoS Negl Trop Dis 2014; 8: e2990, doi: 10.1371/journal.pntd.0002990.
https://doi.org/10.1371/journal.pntd.000...
). Since 2014, the ECSA genotype has been detected in several Brazilian states in the northeastern, southeastern, and northern regions, seriously threatening public health (2222. Fabri AA, Rodrigues CDS, dos Santos CC, Chalhoub FLL, Sampaio SA, Faria NRC, et al. Co-circulation of two independent clades and persistence of chikv-ecsa genotype during epidemic waves in Rio de Janeiro, Southeast Brazil. Pathogens 2020; 9: 984, doi: 10.3390/pathogens9120984.
https://doi.org/10.3390/pathogens9120984...
).

The female Aedes mosquito introduces the virions at the intradermal level through the bite, and they enter the subcutaneous capillaries. There, a local viral replication occurs in susceptible cells such as fibroblasts, endothelial cells, and macrophages (2424. Chow A, Her Z, Ong EKS, Chen J, Dimatatac F, Kwek DJC, 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, doi: 10.1093/infdis/jiq042.
https://doi.org/10.1093/infdis/jiq042...
). Couderc et al. (2525. Couderc T, Chrétien 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, doi: 10.1371/journal.ppat.0040029.
https://doi.org/10.1371/journal.ppat.004...
) established the development of an animal model and observed that CHIKV replicates first in the liver and target muscles, joints, and skin, resembling the tissue/cell tropism seen on biopsy samples from humans infected with CHIKV. They also observed the spread of the virus to the choroid plexuses and leptomeninges in the central nervous system in severe infections (2525. Couderc T, Chrétien 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, doi: 10.1371/journal.ppat.0040029.
https://doi.org/10.1371/journal.ppat.004...
).

Epidemiology of Chikungunya

CHIKV is a zoonotic virus that uses several non-human primates and possibly other vertebrates as amplification hosts (1010. Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines 2012; 11: 1087-1101, doi: 10.1586/erv.12.84.
https://doi.org/10.1586/erv.12.84...
). Phylogenetic studies of CHIKV suggest that the virus has been circulating enzootically in Africa for centuries or longer (1010. Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines 2012; 11: 1087-1101, doi: 10.1586/erv.12.84.
https://doi.org/10.1586/erv.12.84...
,2626. Tsetsarkin KA, Chen R, Weaver SC. Interspecies transmission and Chikungunya virus emergence. Curr Opin Virol 2016; 16: 143-150, doi: 10.1016/j.coviro.2016.02.007.
https://doi.org/10.1016/j.coviro.2016.02...
). In addition to evidence of Chikungunya outbreaks in the 18th century, there are descriptions of serial epidemics consistent with the disease in India, dating from the 1820s, 1853, 1871, and 1923, especially in Calcutta but also extending to Burma (1010. Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines 2012; 11: 1087-1101, doi: 10.1586/erv.12.84.
https://doi.org/10.1586/erv.12.84...
). In 2005, a large outbreak of Chikungunya occurred on islands in the Indian Ocean, reaching several countries in Asia in the following years, causing the infection of more than 1.9 million people (2727. 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: 84, doi: 10.1186/s12929-021-00778-8.
https://doi.org/10.1186/s12929-021-00778...
,2828. Gratz NG. Critical review of the vector status of Aedes albopictus. Med Vet Entomol 2004; 18: 215-227, doi: 10.1111/j.0269-283X.2004.00513.x.
https://doi.org/10.1111/j.0269-283X.2004...
).

CHIKV arrived in the Americas for the first time in 2013. In October of that year, the first autochthonous cases were diagnosed on Saint Martin island in the French Caribbean (2929. Leparc-Goffart I, Nougairede A, Cassadou S, Prat C, De Lamballerie X. Chikungunya in the Americas. Lancet 2014; 383: 514, doi: 10.1016/S0140-6736(14)60185-9.
https://doi.org/10.1016/S0140-6736(14)60...
). A major epidemic began that spread to more than 43 American countries, with 1,300,000 cases and 191 deaths in 2014 (44. Weaver SC, Lecuit M. Chikungunya virus and the global spread of a mosquito-borne disease. N Engl J Med 2015; 372: 1231-1239, doi: 10.1056/NEJMra1406035.
https://doi.org/10.1056/NEJMra1406035...
).

In Brazil, the virus was first identified in 2014. As of June 2021, 41,673 probable cases of Chikungunya were notified in Brazil. Of these, 21,127 occurred in the Northeast region, 18,663 in the Southeast region, 605 in the North region, 655 in the South region, and 623 in the Center-West region (3030. Brasil. Monitoramento dos casos de arboviroses urbanas causados por vírus transmitidos pelo mosquito Aedes (Dengue, Chikungunya E Zika), semanas epidemiológicas 1 a 42, 2021. Bol Epidemiológico Arboviroses 2021; 52: 1-20. Accessed May 29, 2022.). Globally, as of 23 November 2022, 362,021 cases and 77 deaths were reported. The majority of cases have been reported in Brazil (247,537), India (108,957), Guatemala (1,615), Thailand (842), and Malaysia (688). Deaths have been reported in Brazil (75) and Kenya (1) (7).

Clinical manifestations of Chikungunya

In most reports, almost all of the infected patients became symptomatic. Acute clinical symptoms usually include high fever (>38.5°C), chills, severe joint and muscle pain, rash, weakness, and headache. Fever is the main symptom in children. Atypical and severe cases were frequently observed in this group of patients, with the disease leading to hyperpigmentation and erythema (3131. Contopoulos-Ioannidis D, Newman-Lindsay S, Chow C, LaBeaud AD. Mother-to-child transmission of Chikungunya virus: A systematic review and meta-analysis. PLoS Negl Trop Dis 2018; 12: e0006510, doi: 10.1371/journal.pntd.0006510.
https://doi.org/10.1371/journal.pntd.000...
,3232. Torres JR, Falleiros-Arlant LH, Dueãas L, Pleitez-Navarrete J, Salgado DM, Brea-Del Castillo J. Congenital and perinatal complications of Chikungunya fever: a Latin American experience. Int J Infect Dis 2016; 51: 85-88, doi: 10.1016/j.ijid.2016.09.009.
https://doi.org/10.1016/j.ijid.2016.09.0...
). CHIKV can cause persistent infection in macrophages and replicate at low levels for several days, spreading to joint tissues and causing cellular infiltrates, persistent inflammation, and arthritic pain (1010. Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines 2012; 11: 1087-1101, doi: 10.1586/erv.12.84.
https://doi.org/10.1586/erv.12.84...
). Studies have detected CHIKV RNA in the affected joints, being able to induce arthritis (3333. Narendra SC, Chalise JP, Höök N, Magnusson M. Dendritic cells activated by double‐stranded RNA induce arthritis via autocrine type I IFN signaling. J Leukoc Biol 2014; 95: 661-666, doi: 10.1189/jlb.0613320.
https://doi.org/10.1189/jlb.0613320...
).

Studies show that more than 40% of patients progress to the chronic form of the disease. The pathophysiological mechanisms of musculoskeletal pain and chronic arthritis after CHIKV infection are only partially understood (3434. Javelle E, Ribera A, Degasne I, Gaüzàre BA, Marimoutou C, Simon F. Specific management of post-chikungunya rheumatic disorders: a retrospective study of 159 cases in Reunion Island from 2006-2012. PLoS Negl Trop Dis 2015; 9: e0003603, doi: 10.1371/journal.pntd.0003603.
https://doi.org/10.1371/journal.pntd.000...
).

Studies carried out after CHIKV outbreaks in Réunion Island in 2006 and Italy in 2007 showed persistence of myalgia, asthenia, and arthralgia in 60-67% of cases 36 and 12 months after infection, respectively (3535. Noret M, Herrero L, Rulli N, Rolph M, Smith PN, Li RW, et al. Interleukin 6, RANKL, and osteoprotegerin expression by Chikungunya virus-infected human osteoblasts. J Infect Dis 2012; 206: 455-457, doi: 10.1093/infdis/jis368.
https://doi.org/10.1093/infdis/jis368...
). Chronic manifestations may include monoarthritis, oligoarthritis, undifferentiated polyarthritis, and joint involvement mimicking rheumatoid arthritis (3636. Madariaga M, Ticona E, Resurrecion C. Chikungunya: bending over the Americas and the rest of the world. Braz J Infect Dis 2016; 20: 91-98, doi: 10.1016/j.bjid.2015.10.004.
https://doi.org/10.1016/j.bjid.2015.10.0...
). Fatigue, mood disorders, and sleep disturbances were also common chronic symptoms (3737. Paixão ES, Rodrigues LC, Costa MCN, Itaparica M, Barreto F, Gérardin P, et al. Chikungunya chronic disease: a systematic review and meta-analysis. Trans R Soc Trop Med Hyg 2018; 112: 301-316, doi: 10.1093/trstmh/try063.
https://doi.org/10.1093/trstmh/try063...
). Long-term arthralgia was typically polyarthralgia (70%), which was generally symmetrical (90%) and highly disabling (77%). These manifestations were often associated with local swelling (63%) or depression (56%) (3535. Noret M, Herrero L, Rulli N, Rolph M, Smith PN, Li RW, et al. Interleukin 6, RANKL, and osteoprotegerin expression by Chikungunya virus-infected human osteoblasts. J Infect Dis 2012; 206: 455-457, doi: 10.1093/infdis/jis368.
https://doi.org/10.1093/infdis/jis368...
).

CHIKV causes an exaggerated inflammatory response and infection of osteoblasts, which resembles rheumatoid arthritis by the similar pattern of leukocyte infiltration, cytokine production, and complement system activation (3838. Burt F, Chen W, Mahalingam S. Chikungunya virus and arthritic disease. Lancet Infect Dis 2014; 14: 789-790, doi: 10.1016/S1473-3099(14)70869-2.
https://doi.org/10.1016/S1473-3099(14)70...
). Neurological complications, including encephalitis, optic neuritis, facial paralysis, sensorineural deafness, and Guillain-Barré syndrome, may occur in a variable proportion of patients. Less frequently, CHIKV causes myocarditis, cardiac arrhythmia, severe sepsis, septic shock, and renal failure (3939. Amaral JK, Taylor PC, Teixeira MM, Morrison TET, Schoen RT. The clinical features, pathogenesis and methotrexate therapy of chronic Chikungunya arthritis. Viruses 2019; 11: 289, doi: 10.3390/v11030289.
https://doi.org/10.3390/v11030289...
).

Immune responses to CHIKV

Innate immune response

The mechanism by which CHIKV initiates the immune response is similar to that observed in other RNA viruses. The innate immune response begins with the recognition of viral RNA by pattern recognition receptors (PRRs) (4040. Thompson CD, Matta B, Barnes BJ. Therapeutic targeting of IRFs: pathway-dependence or structure-based? Front Immunol 2018; 9: 2622, doi: 10.3389/fimmu.2018.02622.
https://doi.org/10.3389/fimmu.2018.02622...
,4141. He X, Jia H, Jing Z, Liu D. Recognition of pathogen-associated nucleic acids by endosomal nucleic acid-sensing toll-like receptors. Acta Biochim Biophys Sin (Shangai) 2013; 45: 241-258, doi: 10.1093/abbs/gms122.
https://doi.org/10.1093/abbs/gms122...
). In the case of CHIKV, viral RNA activates the Toll-like receptors (TLR-3 and TLR-7) and retinoic acid-inducible gene-like receptors (RIG-I) and melanoma differentiation-associated protein 5 (MDA5) (4141. He X, Jia H, Jing Z, Liu D. Recognition of pathogen-associated nucleic acids by endosomal nucleic acid-sensing toll-like receptors. Acta Biochim Biophys Sin (Shangai) 2013; 45: 241-258, doi: 10.1093/abbs/gms122.
https://doi.org/10.1093/abbs/gms122...
,4242. Blasius AL, Beutler B. Intracellular toll-like receptors. Immunity 2010; 32: 305-315, doi: 10.1016/j.immuni.2010.03.012.
https://doi.org/10.1016/j.immuni.2010.03...
). The signal emitted through the ectodomain of these receptors allows the recruitment of adapter molecules that activate different transcription factors such as nuclear factor (NF)-κB and the interferon regulatory factors (IRFs), inducing the transcription of antiviral interferons (type I IFNs: IFN-α and IFN-β) and proinflammatory cytokines (4141. He X, Jia H, Jing Z, Liu D. Recognition of pathogen-associated nucleic acids by endosomal nucleic acid-sensing toll-like receptors. Acta Biochim Biophys Sin (Shangai) 2013; 45: 241-258, doi: 10.1093/abbs/gms122.
https://doi.org/10.1093/abbs/gms122...
,4242. Blasius AL, Beutler B. Intracellular toll-like receptors. Immunity 2010; 32: 305-315, doi: 10.1016/j.immuni.2010.03.012.
https://doi.org/10.1016/j.immuni.2010.03...
).

The release of type I IFNs controls viral replication in cells and viral progression during acute infection. Studies show that inefficient type I IFNs signaling is a risk factor for developing severe forms of CHIKV-associated disease (2525. Couderc T, Chrétien 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, doi: 10.1371/journal.ppat.0040029.
https://doi.org/10.1371/journal.ppat.004...
,3333. Narendra SC, Chalise JP, Höök N, Magnusson M. Dendritic cells activated by double‐stranded RNA induce arthritis via autocrine type I IFN signaling. J Leukoc Biol 2014; 95: 661-666, doi: 10.1189/jlb.0613320.
https://doi.org/10.1189/jlb.0613320...
). In humans, IFN-α is released early and is detected on the first day of infection. Moreover, its concentration correlates with plasma viral load (1010. Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines 2012; 11: 1087-1101, doi: 10.1586/erv.12.84.
https://doi.org/10.1586/erv.12.84...
,4343. 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, doi: 10.1084/jem.20090851.
https://doi.org/10.1084/jem.20090851...
). Experimental models show an early induction of IFN-α, MCP-1/CCL-2, and IL-6, followed by the detection of MIP-1α/CCL-3 and MIP-1β/CCL-4, IFN-γ, and TNF (4444. 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, doi: 10.1172/JCI40104.
https://doi.org/10.1172/JCI40104...
,4545. 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, doi: 10.1128/JVI.03364-13.
https://doi.org/10.1128/JVI.03364-13...
).

Teo et al. (4646. Teo TH, Her Z, Tan JJL, Lum FM, Lee WWL, Chan YH, et al. Caribbean and La Réunion 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, doi: 10.1128/JVI.00909-15.
https://doi.org/10.1128/JVI.00909-15...
) demonstrated the importance of NK cells during acute joint pathology in CHIKV-infected mice and revealed that reduced NK cells activity could underlie the reduced pathology in the host. Different studies showed that in the chronic phase of Chikungunya, the cellular infiltrate in the affected joints is mainly composed of macrophages, NK cells, monocytes, and macrophage-derived osteoclasts (1010. Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines 2012; 11: 1087-1101, doi: 10.1586/erv.12.84.
https://doi.org/10.1586/erv.12.84...
,4747. Dupuis-Maguiraga L, Noret M, Brun S, Le Grand R, Gras G, Roques P. Chikungunya disease: infection-associated markers from the acute to the chronic phase of arbovirus-induced arthralgia. PLoS Negl Trop Dis 2012; 6: e1446, doi: 10.1371/journal.pntd.0001446.
https://doi.org/10.1371/journal.pntd.000...
). Osteoclasts and IL-6 may be associated with bone destruction. In most cases, post-CHIKV arthritis is usually non-erosive (3535. Noret M, Herrero L, Rulli N, Rolph M, Smith PN, Li RW, et al. Interleukin 6, RANKL, and osteoprotegerin expression by Chikungunya virus-infected human osteoblasts. J Infect Dis 2012; 206: 455-457, doi: 10.1093/infdis/jis368.
https://doi.org/10.1093/infdis/jis368...
).

Persistent arthralgia was associated with higher levels of IL-6 and granulocyte-macrophage colony-stimulating factor (GM-CSF) (2424. Chow A, Her Z, Ong EKS, Chen J, Dimatatac F, Kwek DJC, 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, doi: 10.1093/infdis/jiq042.
https://doi.org/10.1093/infdis/jiq042...
). A study suggests that γδ T cells play a protective role by limiting the inflammatory response induced by CHIKV, and mice with γδ T cell deficiency showed exacerbated CHIKV infection (4848. Long KM, Ferris MT, Whitmore AC, Montgomery SA, Thurlow LR, McGee CE, et al. γδ T cells play a protective role in Chikungunya virus-induced disease. J Virol 2016; 90: 433-443, doi: 10.1128/JVI.02159-15.
https://doi.org/10.1128/JVI.02159-15...
).

Studies show that CHIKV presents different mechanisms to counteract the host's immune response. CHIKV inhibits IFNs signaling through its nonstructural protein nsP2 by blocking the JAK/STAT (signal transducer and activator of transcription) pathway (4949. Lucas-Hourani M, Lupan A, Despràs P, Thoret S, Pamlard O, Dubois J, et al. A phenotypic assay to identify Chikungunya virus inhibitors targeting the nonstructural protein nsP2. J Biomol Screen 2013; 18: 172-179, doi: 10.1177/1087057112460091.
https://doi.org/10.1177/1087057112460091...
,5050. Fros JJ, Liu WJ, Prow NA, Geertsema C, Ligtenberg M, Vanlandingham DL, et al. Chikungunya virus nonstructural protein 2 inhibits type I/II interferon-stimulated JAK-STAT signaling. J Virol 2010; 84: 10877-10887, doi: 10.1128/JVI.00949-10.
https://doi.org/10.1128/JVI.00949-10...
). Moreover, mutations within nsP2 alter the production of IFN-β, and the nsP2 viral protein can inhibit RNA polymerase II by inducing the degradation of its catalytic subunit RpB1, blocking the expression of cellular genes (5151. Meshram CD, Lukash T, Phillips AT, Akhrymuk I, Frolova EI, Frolov I. Lack of nsP2-specific nuclear functions attenuates Chikungunya virus replication both in vitro and in vivo. Virology 2019; 534: 14-24, doi: 10.1016/j.virol.2019.05.016.
https://doi.org/10.1016/j.virol.2019.05....
,5252. Akhrymuk I, Kulemzin S V, Frolova EI. Evasion of the innate immune response: the Old World alphavirus nsP2 protein induces rapid degradation of Rpb1, a catalytic subunit of RNA polymerase II. J Virol 2012; 86: 7180-7191, doi: 10.1128/JVI.00541-12.
https://doi.org/10.1128/JVI.00541-12...
).

Adaptive immune response

Symptoms of acute CHIK are caused by direct cell damage and local inflammation. However, the specific contributions of viral replication and host adaptive immune response to CHIKV infection remain to be fully elucidated (5353. Dhanwani R, Khan M, Alam SI, Rao PVL, Parida M. Differential proteome analysis of Chikungunya virus‐infected new‐born mice tissues reveal implication of stress, inflammatory and apoptotic pathways in disease pathogenesis. Proteomics 2011; 11: 1936-1951, doi: 10.1002/pmic.201000500.
https://doi.org/10.1002/pmic.201000500...
). The acute phase of the human disease leads to the activation and proliferation of CD8+ T cells, whereas the CD4+ T cell response is dominant during the chronic phase of Chikungunya (5454. 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, doi: 10.1093/infdis/jiq006.
https://doi.org/10.1093/infdis/jiq006...
,5555. Poh CM, Chan YH, Ng LFP. Role of T cells in Chikungunya virus infection and utilizing their potential in anti-viral immunity. Front Immunol 2020; 11: 287, doi: 10.3389/fimmu.2020.00287.
https://doi.org/10.3389/fimmu.2020.00287...
).

T and B cells play an essential role in the immune response against CHIKV and the pathogenesis of CHIKV-associated joint disease. Hawman et al. (5656. Hawman DW, Stoermer KA, Montgomery SA, Pal P, Oko L, Diamond MS, et al. Chronic joint disease caused by persistent Chikungunya virus infection is controlled by the adaptive immune response. J Virol 2013; 87: 13878-13888, doi: 10.1128/JVI.02666-13.
https://doi.org/10.1128/JVI.02666-13...
) studied a chronic infection model of Chikungunya using Rag1−/− mice devoid of mature T and B cells. It was demonstrated that CHIKV RNA persisted in joint-associated tissues. In contrast, CHIKV RNA was cleared from visceral and muscle tissues of wild-type mice, suggesting that an acquired immune system is required for CHIKV clearance (5656. Hawman DW, Stoermer KA, Montgomery SA, Pal P, Oko L, Diamond MS, et al. Chronic joint disease caused by persistent Chikungunya virus infection is controlled by the adaptive immune response. J Virol 2013; 87: 13878-13888, doi: 10.1128/JVI.02666-13.
https://doi.org/10.1128/JVI.02666-13...
).

CD4+ T cells were shown to contribute to joint inflammation in the course of CHIKV infection in mice. Carissimo et al. (5757. Carissimo G, Teo TH, Chan YH, Lee CYP, Lee B, Torres-Ruesta A, et al. Viperin controls Chikungunya virus-specific pathogenic T cell IFNγ Th1 stimulation in mice. Life Sci Alliance 2019; 2: e201900298, doi: 10.26508/lsa.201900298.
https://doi.org/10.26508/lsa.201900298...
) showed that interferon-stimulated Viperin protein expression correlated with viral load. An animal model of CHIKV infection using Viperin−/− mice has shown that the absence of this protein significantly contributed to promote an increase of the Th1 profile, with increased INFγ secretion in the evaluated tissues, resulting in high-intensity joint inflammation compared to wild-type mice. Regulatory T cells (Tregs) may also be involved in CHIKV infection immunopathogenesis, as Tregs expansion reduces CHIKV disease by selectively inhibiting CHIKV-specific CD4+ effector T cells during the chronic phase (5858. Lee WWL, Teo TH, Her Z, Lum FM, Kam YW, Haase D, et al. Expanding regulatory T cells alleviates Chikungunya virus-induced pathology in mice. J Virol 2015; 89: 7893-7904, doi: 10.1128/JVI.00998-15.
https://doi.org/10.1128/JVI.00998-15...
).

In mouse models, CD8+ T cell numbers in the joints increase during acute CHIKV infection. However, they do not protect against CHIKV-associated pathologies, as mice deficient in CD8+ T cells still develop joint inflammation (5757. Carissimo G, Teo TH, Chan YH, Lee CYP, Lee B, Torres-Ruesta A, et al. Viperin controls Chikungunya virus-specific pathogenic T cell IFNγ Th1 stimulation in mice. Life Sci Alliance 2019; 2: e201900298, doi: 10.26508/lsa.201900298.
https://doi.org/10.26508/lsa.201900298...
).

The development of anti-CHIKV neutralizing antibodies is essential to control viremia. In humans and mice, the antibody-mediated immune response seems to target the envelope glycoprotein E2 of CHIKV (5959. Chua CL, Sam IC, Chiam CW, Chan YF. The neutralizing role of IgM during early Chikungunya virus infection. PLoS One 2017; 12: e0171989, doi: 10.1371/journal.pone.0171989.
https://doi.org/10.1371/journal.pone.017...
,6060. 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: e0003684, doi: 10.1371/journal.pntd.0003684.
https://doi.org/10.1371/journal.pntd.000...
). Kam et al. (6161. Kam YW, Simarmata D, Chow A, Her Z, Teng TS, Ong EKS, et al. Early appearance of neutralizing immunoglobulin G3 antibodies is associated with Chikungunya virus clearance and long-term clinical protection. J Infect Dis 2012; 205: 1147-1154, doi: 10.1093/infdis/jis033.
https://doi.org/10.1093/infdis/jis033...
) found that the early release of IgG3 isotype antibodies in vivo dominates the humoral response against CHIKV. The neutralizing effect of IgG3 antibodies was also evident in in vitro infection assays. Exposure of CHIKV to patient plasma without IgG3 prevented its inhibitory effect on viral infection of HEK 293T cells. Elderly people have a weaker adaptive response, are more prone to CHIKV infection, and the disease is more severe in these patients (6262. Amdekar S, Parashar D, Alagarasu K. Chikungunya virus-induced arthritis: role of host and viral factors in the pathogenesis. Viral Immunol 2017; 30: 691-702, doi: 10.1089/vim.2017.0052.
https://doi.org/10.1089/vim.2017.0052...
).

The role of polymorphisms in genes involved in host immune response during CHIKV infection

Host factors, such as innate and adaptive immune responses, and also genetic background, play an essential role in Chikungunya disease development and progression. Different studies have shown the association of several host genetic polymorphisms that influence the immune response against CHIKV, resulting in susceptibility or resistance to infection. Here, we addressed the main findings about TLR3/7/8, DC-SIGN, HLA-DRB1/HLA-DQB1, TNF, IL-1RN, OAS2/3, and CRP genetic polymorphisms in Chikungunya (Figure 1 and Table 1).

Figure 1
Summary of host genetic polymorphisms involved in the immune response and their association with Chikungunya virus (CHIKV) infection and clinical symptoms of Chikungunya.
Table 1
Influence of genetic polymorphisms in genes involved in the immune response against Chikungunya virus (CHIKV) infection and disease outcomes.

Toll-like receptors (TLR-3, TLR-7, and TLR-8)

Toll-like receptors (TLRs) are proteins that detect invading or endogenous pathogens, emitting signals that initiate the innate and adaptive immune responses (4242. Blasius AL, Beutler B. Intracellular toll-like receptors. Immunity 2010; 32: 305-315, doi: 10.1016/j.immuni.2010.03.012.
https://doi.org/10.1016/j.immuni.2010.03...
). They are prototypic pattern recognition receptors (PRRs), which recognize conserved microbial signature molecules known as pathogen-associated molecular patterns (PAMPs) or microbe-associated molecular patterns (MAMPs), which recognize any microbe regardless of their degree of pathogenicity (4141. He X, Jia H, Jing Z, Liu D. Recognition of pathogen-associated nucleic acids by endosomal nucleic acid-sensing toll-like receptors. Acta Biochim Biophys Sin (Shangai) 2013; 45: 241-258, doi: 10.1093/abbs/gms122.
https://doi.org/10.1093/abbs/gms122...
,4242. Blasius AL, Beutler B. Intracellular toll-like receptors. Immunity 2010; 32: 305-315, doi: 10.1016/j.immuni.2010.03.012.
https://doi.org/10.1016/j.immuni.2010.03...
).

TLRs that recognize bacterial and fungal components are located on the cell surface. In contrast, TLRs that recognize viral or microbial nucleic acids are located in intracellular membranes, such as endosomes or phagosomes (6363. Hennessy EJ, Parker AE, O'neill LAJ. Targeting Toll-like receptors: emerging therapeutics? Nat Rev Drug Discov 2010; 9: 293-307, doi: 10.1038/nrd3203.
https://doi.org/10.1038/nrd3203...
). Intracellular TLRs (TLR-3, 7, 8, and 9) play crucial roles in response to intracellular pathogens; TLR-3 recognizes double-stranded RNA, TLR-7 and TLR-8 are activated by single-stranded RNA, and TLR-9 senses CpG sequences in DNA molecules (6464. Diebold SS, Brencicova E. Nucleic acids and endosomal pattern recognition: how to tell friend from foe? Front Cell Infect Microbiol 2013; 3: 37, doi: 10.3389/fcimb.2013.00037.
https://doi.org/10.3389/fcimb.2013.00037...
).

TLR-3 is expressed within many cell types, including microglial cells, mast cells, eosinophils, macrophages, NK cells, and dendritic cells, but is less expressed in B and T lymphocytes (6565. Dunlevy F, McElvaney NG, Greene CM. TLR3 sensing of viral infection. Open Infect Dis J 2010; 4: 1-10, doi: 10.2174/1874279301004010001.
https://doi.org/10.2174/1874279301004010...
). The TLR3 L412F (rs3775291) is considered the main SNP in the LRR domain of the TLR3 gene and is associated with the alteration of the TLR-3 function (6666. Ranjith-Kumar CT, Miller W, Sun J, Xiong J, Santos J, Yarbrough I, et al. Effects of single nucleotide polymorphisms on Toll-like receptor 3 activity and expression in cultured cells. J Biol Chem 2007; 282: 17696-17705, doi: 10.1074/jbc.M700209200.
https://doi.org/10.1074/jbc.M700209200...
,6767. Blasius AL, Beutler B. Intracellular toll-like receptors. Immunity 2010; 32: 305-315, doi: 10.1016/j.immuni.2010.03.012.
https://doi.org/10.1016/j.immuni.2010.03...
).

Bucardo et al. (1313. Bucardo F, Reyes Y, Morales M, Briceão R, González F, Lundkvist Å, et al. Association of genetic polymorphisms in DC-SIGN, toll-like receptor 3, and tumor necrosis factor α genes and the Lewis-negative phenotype with Chikungunya infection and disease in Nicaragua. J Infect Dis 2021; 223: 278-286, doi: 10.1093/infdis/jiaa364.
https://doi.org/10.1093/infdis/jiaa364...
) evaluated the role of TLR3 polymorphisms (rs3775291 A/G) in susceptibility to CHIKV infection and its association with clinical symptoms. Multivariate logistic regression analysis showed that Chikungunya disease cases were significantly more likely to be carriers of the AA genotype than the GG genotype (OR=3.26, 95%CI: 1.15, 9.21), with no association observed for the heterozygous AG genotype.

Sengupta et al. (6868. Sengupta S, Mukherjee S, Bhattacharya N, Tripathi A. Differential genotypic signatures of Toll-like receptor polymorphisms among dengue-chikungunya mono-and co-infected Eastern Indian patients. Eur J Clin Microbiol Infect Dis 2021; 1-13, doi: 10.1007/s10096-020-04125-x.
https://doi.org/10.1007/s10096-020-04125...
) performed a study with co-infected patients (dengue virus and CHIKV) and CHIKV mono-infected patients, and they reported that patients with TT genotype of TLR3 (rs3775290) exhibited significantly higher susceptibility towards co-infection. The rs3775290 polymorphism of the TLR3 gene was also evaluated by Dutta et al. (1111. Dutta SK, Tripathi A. Association of toll-like receptor polymorphisms with susceptibility to Chikungunya virus infection. Virology 2017; 511: 207-213, doi: 10.1016/j.virol.2017.08.009.
https://doi.org/10.1016/j.virol.2017.08....
). However, this genetic variant was not associated with CHIKV susceptibility, cytokines secretion, or clinical symptoms.

TLR7/8 are tandem duplicated genes on the X-chromosome located in the endosome membrane and recognize ssRNA and synthetic oligoribonucleotides (6969. Khanmohammadi S, Rezaei N. Role of Toll‐like receptors in the pathogenesis of COVID‐19. J Med Virol 2021; 93: 2735-2739, doi: 10.1002/jmv.26826.
https://doi.org/10.1002/jmv.26826...
,7070. Rauseo DM, Fernández-Mestre M. X-linked Toll-like receptor 7 polymorphism associated with susceptibility to Chikungunya Fever. Asian Pac J Trop Med 2019; 12: 137, doi: 10.4103/1995-7645.254940.
https://doi.org/10.4103/1995-7645.254940...
). TLR-7 is highly expressed in plasmacytoid dendritic cells, which are a cell subset that have a plasmacytoid morphology and primarily secrete vast amounts of type I IFNs in response to viral infection (6969. Khanmohammadi S, Rezaei N. Role of Toll‐like receptors in the pathogenesis of COVID‐19. J Med Virol 2021; 93: 2735-2739, doi: 10.1002/jmv.26826.
https://doi.org/10.1002/jmv.26826...
,7070. Rauseo DM, Fernández-Mestre M. X-linked Toll-like receptor 7 polymorphism associated with susceptibility to Chikungunya Fever. Asian Pac J Trop Med 2019; 12: 137, doi: 10.4103/1995-7645.254940.
https://doi.org/10.4103/1995-7645.254940...
).

The rs3853839 C/G polymorphism of the TLR7 gene shows a significant difference in GC genotype and its distribution among the group of patients infected with CHIKV compared to the control group (OR=3.44, 95%CI: 2.127-5.57; P<0.00001) (1111. Dutta SK, Tripathi A. Association of toll-like receptor polymorphisms with susceptibility to Chikungunya virus infection. Virology 2017; 511: 207-213, doi: 10.1016/j.virol.2017.08.009.
https://doi.org/10.1016/j.virol.2017.08....
). In this same study, they also examined the SNP rs3775290 from the TLR3 gene, and it was associated with the presence of joint pain. The SNPs rs179010, rs5741880, and rs38533839 of the TLR7 gene were associated with increased fever, joint pain, and rash, respectively. Interestingly, the SNP in the TLR7 gene associated with fever (rs179010) was also present in patients with CHIKV infection and increased levels of IFN-α. Finally, fever was also associated with an SNP (rs3764880) in the TLR8 gene (1111. Dutta SK, Tripathi A. Association of toll-like receptor polymorphisms with susceptibility to Chikungunya virus infection. Virology 2017; 511: 207-213, doi: 10.1016/j.virol.2017.08.009.
https://doi.org/10.1016/j.virol.2017.08....
). The exact function of SNP rs179010 of TLR7 is not yet fully elucidated, although some studies suggest that this variant may decrease the expression of TLR-7, downregulating the production of autoantibodies or cytokines (7171. Xiao W, Liu Z, Lin J, Li J, Wu K, Ma Y, et al. Association of Toll‐like receptor 7 and 8 gene polymorphisms with Graves' disease in Chinese Cantonese population. Tissue Antigens 2015; 85: 29-34, doi: 10.1111/tan.12479.
https://doi.org/10.1111/tan.12479...
).

Other SNPs in TLR7 (rs179008 C/A) and TLR8 (rs3764879 G/C) genes were significantly related to increased susceptibility to CHIKV infection (6868. Sengupta S, Mukherjee S, Bhattacharya N, Tripathi A. Differential genotypic signatures of Toll-like receptor polymorphisms among dengue-chikungunya mono-and co-infected Eastern Indian patients. Eur J Clin Microbiol Infect Dis 2021; 1-13, doi: 10.1007/s10096-020-04125-x.
https://doi.org/10.1007/s10096-020-04125...
). Moreover, in the same study, patients with GC genotypes of TLR7 (rs3853839) and TLR8 (rs3764879) and AG genotypes of TLR8 (rs3764880) were more susceptible to CHIKV mono-infection (6868. Sengupta S, Mukherjee S, Bhattacharya N, Tripathi A. Differential genotypic signatures of Toll-like receptor polymorphisms among dengue-chikungunya mono-and co-infected Eastern Indian patients. Eur J Clin Microbiol Infect Dis 2021; 1-13, doi: 10.1007/s10096-020-04125-x.
https://doi.org/10.1007/s10096-020-04125...
). Other studies have demonstrated that TLR8 polymorphisms could produce a truncated TLR-8 receptor with a shorter signal peptide, which results in a more rapid decay of its expression or may affect the in vitro protein function, impairing NF-κB activation (7272. Gantier MP, Irving AT, Kaparakis‐Liaskos M, Xu D, Evans VA, Cameron PU, et al. Genetic modulation of TLR8 response following bacterial phagocytosis. Hum Mutat 2010; 31: 1069-1079, doi: 10.1002/humu.21321.
https://doi.org/10.1002/humu.21321...
).

In conclusion, most of the polymorphisms previously described in the different studies for the TLR3/7/8 genes increase the risk of Chikungunya infection and may influence susceptibility, disease progression, and clinical outcome among infected people. Those genetic variations or SNPs in the TLR genes could induce differential innate immune responses towards the same pathogen.

DC-SIGN (CD209)

DC-SIGN (dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin) is a type C lectin expressed in macrophages and dendritic cells, with fundamental importance in the immune response due to its possible interaction with viral and bacterial pathogens (22. Chaaithanya IK, Muruganandam N, Surya P, Anwesh M, Alagarasu K, Vijayachari P. Association of oligoadenylate synthetase gene cluster and DC-SIGN (CD209) gene polymorphisms with clinical symptoms in Chikungunya virus infection. DNA Cell Biol 2016; 35: 44-50, doi: 10.1089/dna.2015.2819.
https://doi.org/10.1089/dna.2015.2819...
). Like TLRs, DC-SIGN acts as a pattern recognition receptor, promoting phagocytosis in macrophages and dendritic cells (7373. Ojeda N, Salazar C, Cárdenas C, Marshall SH. Expression of DC-SIGN-like C-Type lectin receptors in Salmo salar. Dev Comp Immunol 2020; 113: 103806, doi: 10.1016/j.dci.2020.103806.
https://doi.org/10.1016/j.dci.2020.10380...
). This molecule is encoded by the CD209 gene, and it is located on chromosome 19 and is highly polymorphic (7474. Fang X, Hu Z, Shang W, Zhu J, Xu C, Rao X. Genetic polymorphisms of molecules involved in host immune response to dengue virus infection. FEMS Immunol Med Microbiol 2012; 66: 134-146, doi: 10.1111/j.1574-695X.2012.00995.x.
https://doi.org/10.1111/j.1574-695X.2012...
). Polymorphisms in the CD209 gene have been associated with increased susceptibility to CHIKV infection. This gene has SNPs that affect the expression of DC-SIGN at position −336 (rs4804803) (22. Chaaithanya IK, Muruganandam N, Surya P, Anwesh M, Alagarasu K, Vijayachari P. Association of oligoadenylate synthetase gene cluster and DC-SIGN (CD209) gene polymorphisms with clinical symptoms in Chikungunya virus infection. DNA Cell Biol 2016; 35: 44-50, doi: 10.1089/dna.2015.2819.
https://doi.org/10.1089/dna.2015.2819...
,7575. Alagarasu K, Damle IM, Bachal R V, Mulay AP, Shah PS, Dayaraj C. Association of promoter region polymorphisms of CD209 gene with clinical outcomes of dengue virus infection in Western India. Infect Genet Evol 2013; 17: 239-242, doi: 10.1016/j.meegid.2013.04.024.
https://doi.org/10.1016/j.meegid.2013.04...
).

The rs4804803 variant represents a transition from guanine to adenine at position −336 promoter region of the CD209 gene, and this SNP can regulate the expression of DC-SIGN (7676. Pabalan N, Chaisri S, Tabunhan S, Phumyen A, Jarjanazi H, Steiner TS. Associations of DC-SIGN (CD209) promoter-336G/A polymorphism (rs4804803) with dengue infection: A systematic review and meta-analysis. Acta Trop 2018; 177: 186-193, doi: 10.1016/j.actatropica.2017.10.017.
https://doi.org/10.1016/j.actatropica.20...
). Bucardo et al. (1313. Bucardo F, Reyes Y, Morales M, Briceão R, González F, Lundkvist Å, et al. Association of genetic polymorphisms in DC-SIGN, toll-like receptor 3, and tumor necrosis factor α genes and the Lewis-negative phenotype with Chikungunya infection and disease in Nicaragua. J Infect Dis 2021; 223: 278-286, doi: 10.1093/infdis/jiaa364.
https://doi.org/10.1093/infdis/jiaa364...
) analyzed the polymorphic variant rs4804803 and after a multivariate logistic regression analysis showed that Chikungunya patients were significantly more likely to be carriers of the AG genotype than carriers of the AA wild-type genotype (OR=5.17; 95%CI: 1.6-16.7). The authors also showed that the proportion of individuals carrying the AG genotype of the DC-SIGN-encoding gene was higher in the group seropositive for Chikungunya-specific IgG than in the seronegative group (OR=2.79, 95%CI: 0.88-8.9) (1313. Bucardo F, Reyes Y, Morales M, Briceão R, González F, Lundkvist Å, et al. Association of genetic polymorphisms in DC-SIGN, toll-like receptor 3, and tumor necrosis factor α genes and the Lewis-negative phenotype with Chikungunya infection and disease in Nicaragua. J Infect Dis 2021; 223: 278-286, doi: 10.1093/infdis/jiaa364.
https://doi.org/10.1093/infdis/jiaa364...
).

Another study conducted by Chaaithanya et al. (22. Chaaithanya IK, Muruganandam N, Surya P, Anwesh M, Alagarasu K, Vijayachari P. Association of oligoadenylate synthetase gene cluster and DC-SIGN (CD209) gene polymorphisms with clinical symptoms in Chikungunya virus infection. DNA Cell Biol 2016; 35: 44-50, doi: 10.1089/dna.2015.2819.
https://doi.org/10.1089/dna.2015.2819...
) evaluated the same variant A/G (rs4804803) of the CD209 gene in patients with Chikungunya and healthy controls. The results showed that the frequency of the GG genotype was significantly higher in CHIKV patients compared to healthy controls (P=0.046). The authors suggested that the rs4804803 G/G genotype might influence the expression of DC-SIGN and affect the innate and the development of adaptive immune responses mediated through DCs and hence the possible association with susceptibility to CHIKV infection (22. Chaaithanya IK, Muruganandam N, Surya P, Anwesh M, Alagarasu K, Vijayachari P. Association of oligoadenylate synthetase gene cluster and DC-SIGN (CD209) gene polymorphisms with clinical symptoms in Chikungunya virus infection. DNA Cell Biol 2016; 35: 44-50, doi: 10.1089/dna.2015.2819.
https://doi.org/10.1089/dna.2015.2819...
). In summary, the rs4804803 variant of the DC-SIGN gene contributes to the disease and the infection by the virus. The results of the studies provide new insight into how host genetics influence susceptibility to Chikungunya infection.

Human leukocyte antigens

Human leukocyte antigens (HLA) are coded on the short arm of chromosome 6, occupying a large portion of the DNA (∼3,500 kilobases). HLA are the most polymorphic genes in the human genome. These polymorphisms allow the immune system to increase the repertoire of peptides presented by HLA molecules, which will affect the susceptibility to infectious diseases (7777. Chaaithanya IK, Muruganandam N, Anwesh M, Rajesh R, Ghosal SR, Kartick C, et al. HLA class II allele polymorphism in an outbreak of Chikungunya fever in middle Andaman, India. Immunology 2013; 140: 202-210, doi: 10.1111/imm.12128.
https://doi.org/10.1111/imm.12128...
). There are two main classes of HLA (HLA class I and HLA class II). T cells that express CD8 molecules react with HLA class I molecules on the cell surface of all nucleated cells. HLA class II molecules are usually present only on antigen-presenting cells such as B cells, macrophages, dendritic cells, and Langerhans cells and interact with T CD4+ lymphocytes (7878. Thanapati S, Hande A, Das R, Gurav Y, Tripathy AS. Association of human leukocyte antigen class II allele and haplotypes in Chikungunya viral infection in a western Indian population. Trans R Soc Trop Med Hyg 2014; 108: 277-282, doi: 10.1093/trstmh/tru030.
https://doi.org/10.1093/trstmh/tru030...
). Polymorphism in the HLA genes is clustered in the peptide-binding region and affects the repertoire of peptides presented by an HLA molecule to the T cell.

Studies have demonstrated the association of HLA class II alleles with susceptibility or resistance to CHIKV infection. An Indian study by Chaaithanya et al. (7777. Chaaithanya IK, Muruganandam N, Anwesh M, Rajesh R, Ghosal SR, Kartick C, et al. HLA class II allele polymorphism in an outbreak of Chikungunya fever in middle Andaman, India. Immunology 2013; 140: 202-210, doi: 10.1111/imm.12128.
https://doi.org/10.1111/imm.12128...
) demonstrated a significantly decreased frequency of the HLA-DQB1*03: 03 allele in CHIKV patients compared with healthy subjects, suggesting these alleles might be associated with reduced risk of developing CHIKV symptoms, or severe disease.

In the two-locus haplotypes analysis by Thanapati et al. (7878. Thanapati S, Hande A, Das R, Gurav Y, Tripathy AS. Association of human leukocyte antigen class II allele and haplotypes in Chikungunya viral infection in a western Indian population. Trans R Soc Trop Med Hyg 2014; 108: 277-282, doi: 10.1093/trstmh/tru030.
https://doi.org/10.1093/trstmh/tru030...
), the authors showed that DRB1*11/DQB1*03 and DRB1*04/DQB1*03 haplotypes revealed statistically significant associations with resistance and susceptibility to CHIKV infection, respectively.

A prospective follow-up study of 21 Chikungunya patients from Réunion Island suggested the role of CHIKV infection in rheumatoid arthritis initiation. The study showed the prevalence of HLA-DRB1*01 and DRB1*04 alleles in patients who had rheumatoid arthritis after the infection (7979. Bouquillard E, Combe B. A report of 21 cases of rheumatoid arthritis following Chikungunya fever. A mean follow-up of two years. Joint Bone Spine 2009; 76: 654-657, doi: 10.1016/j.jbspin.2009.08.005.
https://doi.org/10.1016/j.jbspin.2009.08...
). Thus, the studies showed an association of DRB1*11/DQB1*03 and DRB1*04/DQB1*03 as resistant and susceptible haplotypes, respectively, to CHIKV infection. These results suggest that genetic susceptibility and/or resistance to Chikungunya infection may be modulated by HLA class II alleles.

Tumor necrosis factor

Tumor necrosis factor (TNF), an important proinflammatory cytokine, plays a role in regulating cell differentiation, proliferation, and death, as well as in innate and adaptive immune response. The TNF gene is located on chromosome 6 in the class III region of the MHC locus (8080. Qidwai T, Khan F. Tumour necrosis factor gene polymorphism and disease prevalence. Scand J Immunol 2011; 74: 522-547, doi: 10.1111/j.1365-3083.2011.02602.x.
https://doi.org/10.1111/j.1365-3083.2011...
). Several SNPs found in the regulatory region influence the transcription of the TNF gene and the circulating level of TNF (7878. Thanapati S, Hande A, Das R, Gurav Y, Tripathy AS. Association of human leukocyte antigen class II allele and haplotypes in Chikungunya viral infection in a western Indian population. Trans R Soc Trop Med Hyg 2014; 108: 277-282, doi: 10.1093/trstmh/tru030.
https://doi.org/10.1093/trstmh/tru030...
).

A study by Bucardo et al. (1313. Bucardo F, Reyes Y, Morales M, Briceão R, González F, Lundkvist Å, et al. Association of genetic polymorphisms in DC-SIGN, toll-like receptor 3, and tumor necrosis factor α genes and the Lewis-negative phenotype with Chikungunya infection and disease in Nicaragua. J Infect Dis 2021; 223: 278-286, doi: 10.1093/infdis/jiaa364.
https://doi.org/10.1093/infdis/jiaa364...
) evaluated the variant rs1800629 A/G of the TNF gene in patients with Chikungunya. TNF genotype distribution was not associated with the risk of clinical Chikungunya disease nor with Chikungunya seropositivity. However, Chikungunya patients with persistent joint pain were significantly less likely to be carriers of the AG genotype than the GG genotype (OR=0.24; 95%CI: 0.074-0.76). The authors suggested that carriers of the AG genotype, expressing higher levels of TNF-α, might be better able to control infection and clear the infection early, leading to lower prevalence of persistent joint pain (1313. Bucardo F, Reyes Y, Morales M, Briceão R, González F, Lundkvist Å, et al. Association of genetic polymorphisms in DC-SIGN, toll-like receptor 3, and tumor necrosis factor α genes and the Lewis-negative phenotype with Chikungunya infection and disease in Nicaragua. J Infect Dis 2021; 223: 278-286, doi: 10.1093/infdis/jiaa364.
https://doi.org/10.1093/infdis/jiaa364...
).

Overall, the rs1800629 polymorphism of the TNF gene was not associated with susceptibility to Chikungunya disease or seropositivity by the study presented. However, the AG genotype was associated with a higher concentration of TNF-α that helps in the inhibition of viral replication and early control of the infection leading to a decrease in joint pain in individuals with chronic Chikungunya.

Interleukin-1 receptor antagonist (IL-1RN)

The IL-1 family consists of IL-1α and IL-1β. These cytokines bind the IL-1 receptor (IL-1R) and initiate an inflammation cascade to induce vascular dilation and fever (8181. Nicklin MJH, Weith A, Duff GW. A physical map of the region encompassing the human interleukin-1α, interleukin-1β, and interleukin-1 receptor antagonist genes. Genomics 1994; 19: 382-384, doi: 10.1006/geno.1994.1076.
https://doi.org/10.1006/geno.1994.1076...
). The human interleukin-1 receptor antagonist (IL1RN) gene is located on the long arm of chromosome 2, and the IL-1RN molecule is a potent endogenous competitive inhibitor of IL-1α and IL-1β and has an anti-inflammatory role (8181. Nicklin MJH, Weith A, Duff GW. A physical map of the region encompassing the human interleukin-1α, interleukin-1β, and interleukin-1 receptor antagonist genes. Genomics 1994; 19: 382-384, doi: 10.1006/geno.1994.1076.
https://doi.org/10.1006/geno.1994.1076...
). IL1RN gene polymorphism results in quantitative differences in both IL-1RN and IL-1β production. The polymorphic region within intron 2 of the IL1RN* gene contains variable numbers of tandem repeats (VNTR) of 86 bp, resulting in five alleles of the IL1RN* (*1-*5), corresponding to 2, 3, 4, 5, and 6 copies of the 86-bp sequence, respectively (8181. Nicklin MJH, Weith A, Duff GW. A physical map of the region encompassing the human interleukin-1α, interleukin-1β, and interleukin-1 receptor antagonist genes. Genomics 1994; 19: 382-384, doi: 10.1006/geno.1994.1076.
https://doi.org/10.1006/geno.1994.1076...
,8282. Tripathy AS, Ganu MA, Sonam L, Alagarasu K, Walimbe AM, Thanapati S, et al. Association of IL1RN VNTR polymorphism with Chikungunya infection: a study from Western India. J Med Virol 2019; 91: 1901-1908, doi: 10.1002/jmv.25546.
https://doi.org/10.1002/jmv.25546...
).

The study conducted by Tripathy et al. (8282. Tripathy AS, Ganu MA, Sonam L, Alagarasu K, Walimbe AM, Thanapati S, et al. Association of IL1RN VNTR polymorphism with Chikungunya infection: a study from Western India. J Med Virol 2019; 91: 1901-1908, doi: 10.1002/jmv.25546.
https://doi.org/10.1002/jmv.25546...
) revealed a significant association of the IL1RN *1/*1 genotype under a recessive genetic model with the risk of developing CHIKV infection (OR=1.57, 95%CI: 1.06-2.34, P=0.02). Other findings also indicated that IL1RN*2 allele under the dominant model was associated with protection against chronic Chikungunya (OR=0.54, 95%CI: 0.34-0.85, P=0.009). These findings also suggest a role of higher IL-1RN production in the pathogenesis of chronic Chikungunya (8282. Tripathy AS, Ganu MA, Sonam L, Alagarasu K, Walimbe AM, Thanapati S, et al. Association of IL1RN VNTR polymorphism with Chikungunya infection: a study from Western India. J Med Virol 2019; 91: 1901-1908, doi: 10.1002/jmv.25546.
https://doi.org/10.1002/jmv.25546...
). Ultimately, the results suggest the association of ILRN VNTR polymorphism with Chikungunya. Individuals with the IL1RN*1/*1 genotype showed an increased risk of CHIKV infection, whereas the IL1RN*2 allele could have a protective effect against the development of chronic disease.

Oligoadenylate synthetase (OAS)

Human oligoadenylate synthetase (OAS) is a family of enzymes encoded by three closely linked genes on chromosome 12q24.2 in the following order: small (OAS1, p40/p46), medium (OAS2, p69/71), and large (OAS3, p100) (8383. Hovanessian AG, Justesen J. The human 2′-5′ oligoadenylate synthetase family: unique interferon-inducible enzymes catalyzing 2′-5′ instead of 3′-5′ phosphodiester bond formation. Biochimie 2007; 89: 779-788, doi: 10.1016/j.biochi.2007.02.003.
https://doi.org/10.1016/j.biochi.2007.02...
,8484. Bréhin AC, Casadémont I, Frenkiel MP, Julier C, Sakuntabhai A, Despràs P. The large form of human 2′, 5′-Oligoadenylate Synthetase (OAS3) exerts antiviral effect against Chikungunya virus. Virology 2009; 384: 216-222, doi: 10.1016/j.virol.2008.10.021.
https://doi.org/10.1016/j.virol.2008.10....
). OAS3 efficiently inhibits the growth of CHIKV. The antiviral effect was a blockade in the early stages of virus replication (8484. Bréhin AC, Casadémont I, Frenkiel MP, Julier C, Sakuntabhai A, Despràs P. The large form of human 2′, 5′-Oligoadenylate Synthetase (OAS3) exerts antiviral effect against Chikungunya virus. Virology 2009; 384: 216-222, doi: 10.1016/j.virol.2008.10.021.
https://doi.org/10.1016/j.virol.2008.10....
). OAS families are formed by homologous enzymes that are encoded by IFN-stimulated genes. The 2'-5'-oligoadenylate synthetase (OAS)/ribonuclease L (RNase L) system is an innate immunity pathway that responds to pathogens and is associated with the induction of viral and cellular RNAs degradation, thereby blocking viral infections (8585. Kristiansen H, Gad HH, Eskildsen-Larsen S, Despres P, Hartmann R. The oligoadenylate synthetase family: an ancient protein family with multiple antiviral activities. J Interferon Cytokine Res 2011; 31: 41-47, doi: 10.1089/jir.2010.0107.
https://doi.org/10.1089/jir.2010.0107...
). Polymorphisms in the genes encoding OAS enzymes are known to influence susceptibility and severity of viral diseases (22. Chaaithanya IK, Muruganandam N, Surya P, Anwesh M, Alagarasu K, Vijayachari P. Association of oligoadenylate synthetase gene cluster and DC-SIGN (CD209) gene polymorphisms with clinical symptoms in Chikungunya virus infection. DNA Cell Biol 2016; 35: 44-50, doi: 10.1089/dna.2015.2819.
https://doi.org/10.1089/dna.2015.2819...
,8686. Sadler AJ, Williams BRG. Interferon-inducible antiviral effectors. Nat Rev Immunol 2008; 8: 559-568, doi: 10.1038/nri2314.
https://doi.org/10.1038/nri2314...
).

Chaaithanya et al. (22. Chaaithanya IK, Muruganandam N, Surya P, Anwesh M, Alagarasu K, Vijayachari P. Association of oligoadenylate synthetase gene cluster and DC-SIGN (CD209) gene polymorphisms with clinical symptoms in Chikungunya virus infection. DNA Cell Biol 2016; 35: 44-50, doi: 10.1089/dna.2015.2819.
https://doi.org/10.1089/dna.2015.2819...
) aimed to investigate the association of OAS polymorphisms with susceptibility to CHIKV infection, evaluating samples from 101 CHIKV-infected patients and 101 samples obtained from healthy subjects. The OAS2 rs1732778 A/A genotype, under a recessive genetic model (A/A vs G/G+G/A), was significantly associated with a reduced risk of nausea [OR with 95%CI: 0.16 (0.05-0.57), P-value=0.0028] (22. Chaaithanya IK, Muruganandam N, Surya P, Anwesh M, Alagarasu K, Vijayachari P. Association of oligoadenylate synthetase gene cluster and DC-SIGN (CD209) gene polymorphisms with clinical symptoms in Chikungunya virus infection. DNA Cell Biol 2016; 35: 44-50, doi: 10.1089/dna.2015.2819.
https://doi.org/10.1089/dna.2015.2819...
). The OAS2 rs15895 G/A genotype and OAS3 rs2285932 C/T genotype in an overdominant model were significantly associated with a reduced risk of chills. The OAS3 rs2285932 T allele in a dominant model (C/T+T/T vs C/C) and OAS1 rs1131434 G/G genotype under a recessive genetic model were associated with a reduced risk of edema. OAS2 rs15895 G/A in an overdominant model was associated with a reduced risk of conjunctival redness. Thus, the authors suggested that OAS gene cluster polymorphisms were associated with a reduced risk of developing nausea, chills, edema, and conjunctival redness in CHIKV infection (22. Chaaithanya IK, Muruganandam N, Surya P, Anwesh M, Alagarasu K, Vijayachari P. Association of oligoadenylate synthetase gene cluster and DC-SIGN (CD209) gene polymorphisms with clinical symptoms in Chikungunya virus infection. DNA Cell Biol 2016; 35: 44-50, doi: 10.1089/dna.2015.2819.
https://doi.org/10.1089/dna.2015.2819...
).

Finally, this study suggested that SNPs in the OAS gene cluster are associated with the risk of developing clinical symptoms in patients infected with CHIKV. Further follow-up studies are needed to evaluate the role of these genes in association with clinical symptoms in CHIKV patients.

C-reactive protein

C-reactive protein (CRP), an acute phase protein, is genetically located in the region of chromosome 1q23.2 and has been previously used as an inflammatory and infectious biomarker in clinical contexts (8787. Sengupta S, Bhattacharya N, Tripathi A. Increased CRP, anti-CCP antibody, IL-2R, COMP levels in prognosis of post-chikungunya chronic arthritis and protective role of their specific genotypes against arthritic manifestation. Virus Res 2022; 23: 198998, doi: 10.1016/j.virusres.2022.198998.
https://doi.org/10.1016/j.virusres.2022....
). The main stimulus factor for the liver production of CRP is the IL-6 cytokine. Moreover, TNF and IL-1β, among others, act synergistically with IL-6, exacerbating the stimulus for CRP production (8888. Volanakis JE. Human C-reactive protein: expression, structure, and function. Mol Immunol 2001; 38: 189-197, doi: 10.1016/S0161-5890(01)00042-6.
https://doi.org/10.1016/S0161-5890(01)00...
,8989. Verma S, Li SH, Badiwala MV, Weisel RD, Fedak PWM, Li RK, et al. Endothelin antagonism and interleukin-6 inhibition attenuate the proatherogenic effects of C-reactive protein. Circulation 2002; 105: 1890-1896, doi: 10.1161/01.CIR.0000015126.83143.B4.
https://doi.org/10.1161/01.CIR.000001512...
).

Sengupta et al. (9090. Sengupta S, Bhattacharya N, Tripathi A. Association of C-reactive protein polymorphisms with serum-CRP concentration and viral load among dengue-chikungunya mono/co-infected patients. Antiviral Res 2022; 197: 105225, doi: 10.1016/j.antiviral.2021.105225.
https://doi.org/10.1016/j.antiviral.2021...
) investigated the role of polymorphisms in the gene encoding CRP and whether these SNPs influence the level of serum CRP concentration in patients with mono-infection (dengue virus infection/n=201 or CHIKV infection/n=167) and patients co-infected with both viruses (n=128). Patients with CT genotype of rs3093059 and with TT genotype of rs3091244 were more susceptible to co-infection (OR=1.951/P=0.0378 and OR=4.381/P=0.0138). Patients with the CT genotype of the rs3091244 variant exhibited protection against CHIKV infection (OR=0.3611; P=0.278) and increased CRP levels compared to those with the CC genotype. Moreover, the same study showed that individuals with the CC genotype (rs3091244) presented a higher viral load for CHIKV (OR=2.643; P=0.0313). Thus, CRP genotype might play a significant role in determining serum-CRP concentration, viral load, and dengue-CHIKV mono/co-infection (9090. Sengupta S, Bhattacharya N, Tripathi A. Association of C-reactive protein polymorphisms with serum-CRP concentration and viral load among dengue-chikungunya mono/co-infected patients. Antiviral Res 2022; 197: 105225, doi: 10.1016/j.antiviral.2021.105225.
https://doi.org/10.1016/j.antiviral.2021...
).

The same research group published a recent article showing the influence of CRP rs3093059 and rs3091244 polymorphisms in the prognosis of post-Chikungunya chronic arthritis (8787. Sengupta S, Bhattacharya N, Tripathi A. Increased CRP, anti-CCP antibody, IL-2R, COMP levels in prognosis of post-chikungunya chronic arthritis and protective role of their specific genotypes against arthritic manifestation. Virus Res 2022; 23: 198998, doi: 10.1016/j.virusres.2022.198998.
https://doi.org/10.1016/j.virusres.2022....
). The study was conducted with 167 Chikungunya patients and 102 healthy subjects. Statistical analysis revealed that individuals with the TC genotype of rs3091244 were less susceptible to CHIKV infection (P=0.0126). Moreover, CT (rs3093059) and TT (rs3091244) genotypes were significantly associated with patients without any arthralgic manifestations (P<0.0001, P=0.0004, respectively) (8787. Sengupta S, Bhattacharya N, Tripathi A. Increased CRP, anti-CCP antibody, IL-2R, COMP levels in prognosis of post-chikungunya chronic arthritis and protective role of their specific genotypes against arthritic manifestation. Virus Res 2022; 23: 198998, doi: 10.1016/j.virusres.2022.198998.
https://doi.org/10.1016/j.virusres.2022....
).

Thus, the findings shown in these studies suggested that both serum biomarker levels and genetic factors played an essential role in the development of post-Chikungunya chronic arthritis among infected patients. These markers could be used to assess patients and identify their potential to develop chronic arthritis.

Conclusion

This review presented several polymorphisms in genes responsible for encoding transmembrane proteins, enzymes, cell mediators, recognition molecules, and cytokines essential to host immune response against CHIKV infection. SNPs in these genes can alter an individual's gene profile and influence the pathogenesis and progression of Chikungunya disease. These polymorphic variants can increase the risk of infection in the host or even decrease the risk of infection, having a protective effect. The analysis of genetic variations in different genes that influence the immune response can serve as a valuable strategy to identify patients at increased risk of infection by a specific arbovirus, which may lead to more aggressive treatment to prevent the development of severe clinical manifestations.

Acknowledgments

This work was financially supported by CAPES (Coordenação de Aperfeiçoamento de Pessoal de Nível Superior; Finance Code 001) and FUNCAP (Fundação Cearense de Apoio ao Desenvolvimento Científico e Tecnológico - SUS/PPSUS-CE FUNCAP-PP3-0118-00049.01.00/17).

References

  • 1
    Naresh Kumar CVM, Sai Gopal DVR. Reemergence of Chikungunya virus in Indian subcontinent. Indian J Virol 2010; 21: 8-17, doi: 10.1007/s13337-010-0012-1.
    » https://doi.org/10.1007/s13337-010-0012-1
  • 2
    Chaaithanya IK, Muruganandam N, Surya P, Anwesh M, Alagarasu K, Vijayachari P. Association of oligoadenylate synthetase gene cluster and DC-SIGN (CD209) gene polymorphisms with clinical symptoms in Chikungunya virus infection. DNA Cell Biol 2016; 35: 44-50, doi: 10.1089/dna.2015.2819.
    » https://doi.org/10.1089/dna.2015.2819
  • 3
    Burt FJ, Chen W, Miner JJ, Lenschow DJ, Merits A, Schnettler E, et al. Chikungunya virus: an update on the biology and pathogenesis of this emerging pathogen. Lancet Infect Dis 2017; 17: e107-e117, doi: 10.1016/S1473-3099(16)30385-1.
    » https://doi.org/10.1016/S1473-3099(16)30385-1
  • 4
    Weaver SC, Lecuit M. Chikungunya virus and the global spread of a mosquito-borne disease. N Engl J Med 2015; 372: 1231-1239, doi: 10.1056/NEJMra1406035.
    » https://doi.org/10.1056/NEJMra1406035
  • 5
    Giménez-Richarte Á, de Salazar MO, Arbona C, Giménez-Richarte MP, Collado M, Fernández PL, et al. Prevalence of Chikungunya, Dengue and Zika viruses in blood donors: a systematic literature review and meta-analysis. Blood Transfus 2022; 20: 267-280, doi: 10.2450/2021.0106-21.
    » https://doi.org/10.2450/2021.0106-21
  • 6
    Ferreira JM, Santos LDS, Oliveira SP, Dos Santos BRC, Dos Santos ACM, de Moura EL, et al. Chikungunya virus infection outcome: a systematic review of host genetics. Immunol Invest 2021; 50: 58-79, doi: 10.1080/08820139.2020.1733011.
    » https://doi.org/10.1080/08820139.2020.1733011
  • 7
    ECDC (European Centre for Disease Prevention and Control). Chikungunya worldwide overview. Available from https://www.ecdc.eur opa.eu/en/chikungunya-monthly Accessed Dec 05, 2022.
    » https://www.ecdc.eur opa.eu/en/chikungunya-monthly
  • 8
    Gasque P, Bandjee MCJ, Reyes MM, Viasus D. Chikungunya pathogenesis: from the clinics to the bench. J Infect Dis 2016; 214: S446-S448, doi: 10.1093/infdis/jiw362.
    » https://doi.org/10.1093/infdis/jiw362
  • 9
    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, doi: 10.1093/infdis/jiv049.
    » https://doi.org/10.1093/infdis/jiv049
  • 10
    Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines 2012; 11: 1087-1101, doi: 10.1586/erv.12.84.
    » https://doi.org/10.1586/erv.12.84
  • 11
    Dutta SK, Tripathi A. Association of toll-like receptor polymorphisms with susceptibility to Chikungunya virus infection. Virology 2017; 511: 207-213, doi: 10.1016/j.virol.2017.08.009.
    » https://doi.org/10.1016/j.virol.2017.08.009
  • 12
    Mickienė A, Pakalnienė J, Nordgren J, Carlsson B, Hagbom M, Svensson L, et al. Polymorphisms in chemokine receptor 5 and Toll-like receptor 3 genes are risk factors for clinical tick-borne encephalitis in the Lithuanian population. PLoS One 2014; 9: e106798, doi: 10.1371/journal.pone.0106798.
    » https://doi.org/10.1371/journal.pone.0106798
  • 13
    Bucardo F, Reyes Y, Morales M, Briceão R, González F, Lundkvist Å, et al. Association of genetic polymorphisms in DC-SIGN, toll-like receptor 3, and tumor necrosis factor α genes and the Lewis-negative phenotype with Chikungunya infection and disease in Nicaragua. J Infect Dis 2021; 223: 278-286, doi: 10.1093/infdis/jiaa364.
    » https://doi.org/10.1093/infdis/jiaa364
  • 14
    Metz SW, Geertsema C, Martina BE, Andrade P, Heldens JG, van Oers MM, et al. Functional processing and secretion of Chikungunya virus E1 and E2 glycoproteins in insect cells. Virol J 2011; 8: 1-12, doi: 10.1186/1743-422X-8-353.
    » https://doi.org/10.1186/1743-422X-8-353
  • 15
    Jose J, Snyder JE, Kuhn RJ. A structural and functional perspective of alphavirus replication and assembly. Future Microbiol 2009; 4: 837-856, doi: 10.2217/fmb.09.59.
    » https://doi.org/10.2217/fmb.09.59
  • 16
    Thiberville SD, Moyen N, Dupuis-Maguiraga L, Nougairede A, Gould EA, Roques P, et al. Chikungunya fever: epidemiology, clinical syndrome, pathogenesis and therapy. Antiviral Res 2013; 99: 345-370, doi: 10.1016/j.antiviral.2013.06.009.
    » https://doi.org/10.1016/j.antiviral.2013.06.009
  • 17
    Khan AH, Morita K, Parquet MDC, Hasebe F, Mathenge EGM, Igarashi A. Complete nucleotide sequence of Chikungunya virus and evidence for an internal polyadenylation site. J Gen Virol 2002; 83: 3075-3084, doi: 10.1099/0022-1317-83-12-3075.
    » https://doi.org/10.1099/0022-1317-83-12-3075
  • 18
    Voss JE, Vaney MC, Duquerroy S, Vonrhein C, Girard-Blanc C, Crublet E, et al. Glycoprotein organization of Chikungunya virus particles revealed by X-ray crystallography. Nature 2010; 468: 709-712, doi: 10.1038/nature09555.
    » https://doi.org/10.1038/nature09555
  • 19
    Carvalho CAM, Silva JL, Oliveira AC, Gomes AMO. On the entry of an emerging arbovirus into host cells: Mayaro virus takes the highway to the cytoplasm through fusion with early endosomes and caveolae-derived vesicles. PeerJ 2017; 5: e3245, doi: 10.7717/peerj.3245.
    » https://doi.org/10.7717/peerj.3245
  • 20
    Ramsey J, Mukhopadhyay S. Disentangling the frames, the state of research on the alphavirus 6K and TF proteins. Viruses 2017; 9: 228, doi: 10.3390/v9080228.
    » https://doi.org/10.3390/v9080228
  • 21
    Tomar S, Hardy RW, Smith JL, Kuhn RJ. Catalytic core of alphavirus nonstructural protein nsP4 possesses terminal adenylyltransferase activity. J Virol 2006; 80: 9962-9969, doi: 10.1128/JVI.01067-06.
    » https://doi.org/10.1128/JVI.01067-06
  • 22
    Fabri AA, Rodrigues CDS, dos Santos CC, Chalhoub FLL, Sampaio SA, Faria NRC, et al. Co-circulation of two independent clades and persistence of chikv-ecsa genotype during epidemic waves in Rio de Janeiro, Southeast Brazil. Pathogens 2020; 9: 984, doi: 10.3390/pathogens9120984.
    » https://doi.org/10.3390/pathogens9120984
  • 23
    Agarwal A, Dash PK, Singh AK, Sharma S, Gopalan N, Rao PVL, et al. Evidence of experimental vertical transmission of emerging novel ECSA genotype of Chikungunya virus in Aedes aegypti. PLoS Negl Trop Dis 2014; 8: e2990, doi: 10.1371/journal.pntd.0002990.
    » https://doi.org/10.1371/journal.pntd.0002990
  • 24
    Chow A, Her Z, Ong EKS, Chen J, Dimatatac F, Kwek DJC, 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, doi: 10.1093/infdis/jiq042.
    » https://doi.org/10.1093/infdis/jiq042
  • 25
    Couderc T, Chrétien 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, doi: 10.1371/journal.ppat.0040029.
    » https://doi.org/10.1371/journal.ppat.0040029
  • 26
    Tsetsarkin KA, Chen R, Weaver SC. Interspecies transmission and Chikungunya virus emergence. Curr Opin Virol 2016; 16: 143-150, doi: 10.1016/j.coviro.2016.02.007.
    » https://doi.org/10.1016/j.coviro.2016.02.007
  • 27
    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: 84, doi: 10.1186/s12929-021-00778-8.
    » https://doi.org/10.1186/s12929-021-00778-8
  • 28
    Gratz NG. Critical review of the vector status of Aedes albopictus Med Vet Entomol 2004; 18: 215-227, doi: 10.1111/j.0269-283X.2004.00513.x.
    » https://doi.org/10.1111/j.0269-283X.2004.00513.x
  • 29
    Leparc-Goffart I, Nougairede A, Cassadou S, Prat C, De Lamballerie X. Chikungunya in the Americas. Lancet 2014; 383: 514, doi: 10.1016/S0140-6736(14)60185-9.
    » https://doi.org/10.1016/S0140-6736(14)60185-9
  • 30
    Brasil. Monitoramento dos casos de arboviroses urbanas causados por vírus transmitidos pelo mosquito Aedes (Dengue, Chikungunya E Zika), semanas epidemiológicas 1 a 42, 2021. Bol Epidemiológico Arboviroses 2021; 52: 1-20. Accessed May 29, 2022.
  • 31
    Contopoulos-Ioannidis D, Newman-Lindsay S, Chow C, LaBeaud AD. Mother-to-child transmission of Chikungunya virus: A systematic review and meta-analysis. PLoS Negl Trop Dis 2018; 12: e0006510, doi: 10.1371/journal.pntd.0006510.
    » https://doi.org/10.1371/journal.pntd.0006510
  • 32
    Torres JR, Falleiros-Arlant LH, Dueãas L, Pleitez-Navarrete J, Salgado DM, Brea-Del Castillo J. Congenital and perinatal complications of Chikungunya fever: a Latin American experience. Int J Infect Dis 2016; 51: 85-88, doi: 10.1016/j.ijid.2016.09.009.
    » https://doi.org/10.1016/j.ijid.2016.09.009
  • 33
    Narendra SC, Chalise JP, Höök N, Magnusson M. Dendritic cells activated by double‐stranded RNA induce arthritis via autocrine type I IFN signaling. J Leukoc Biol 2014; 95: 661-666, doi: 10.1189/jlb.0613320.
    » https://doi.org/10.1189/jlb.0613320
  • 34
    Javelle E, Ribera A, Degasne I, Gaüzàre BA, Marimoutou C, Simon F. Specific management of post-chikungunya rheumatic disorders: a retrospective study of 159 cases in Reunion Island from 2006-2012. PLoS Negl Trop Dis 2015; 9: e0003603, doi: 10.1371/journal.pntd.0003603.
    » https://doi.org/10.1371/journal.pntd.0003603
  • 35
    Noret M, Herrero L, Rulli N, Rolph M, Smith PN, Li RW, et al. Interleukin 6, RANKL, and osteoprotegerin expression by Chikungunya virus-infected human osteoblasts. J Infect Dis 2012; 206: 455-457, doi: 10.1093/infdis/jis368.
    » https://doi.org/10.1093/infdis/jis368
  • 36
    Madariaga M, Ticona E, Resurrecion C. Chikungunya: bending over the Americas and the rest of the world. Braz J Infect Dis 2016; 20: 91-98, doi: 10.1016/j.bjid.2015.10.004.
    » https://doi.org/10.1016/j.bjid.2015.10.004
  • 37
    Paixão ES, Rodrigues LC, Costa MCN, Itaparica M, Barreto F, Gérardin P, et al. Chikungunya chronic disease: a systematic review and meta-analysis. Trans R Soc Trop Med Hyg 2018; 112: 301-316, doi: 10.1093/trstmh/try063.
    » https://doi.org/10.1093/trstmh/try063
  • 38
    Burt F, Chen W, Mahalingam S. Chikungunya virus and arthritic disease. Lancet Infect Dis 2014; 14: 789-790, doi: 10.1016/S1473-3099(14)70869-2.
    » https://doi.org/10.1016/S1473-3099(14)70869-2
  • 39
    Amaral JK, Taylor PC, Teixeira MM, Morrison TET, Schoen RT. The clinical features, pathogenesis and methotrexate therapy of chronic Chikungunya arthritis. Viruses 2019; 11: 289, doi: 10.3390/v11030289.
    » https://doi.org/10.3390/v11030289
  • 40
    Thompson CD, Matta B, Barnes BJ. Therapeutic targeting of IRFs: pathway-dependence or structure-based? Front Immunol 2018; 9: 2622, doi: 10.3389/fimmu.2018.02622.
    » https://doi.org/10.3389/fimmu.2018.02622
  • 41
    He X, Jia H, Jing Z, Liu D. Recognition of pathogen-associated nucleic acids by endosomal nucleic acid-sensing toll-like receptors. Acta Biochim Biophys Sin (Shangai) 2013; 45: 241-258, doi: 10.1093/abbs/gms122.
    » https://doi.org/10.1093/abbs/gms122
  • 42
    Blasius AL, Beutler B. Intracellular toll-like receptors. Immunity 2010; 32: 305-315, doi: 10.1016/j.immuni.2010.03.012.
    » https://doi.org/10.1016/j.immuni.2010.03.012
  • 43
    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, doi: 10.1084/jem.20090851.
    » https://doi.org/10.1084/jem.20090851
  • 44
    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, doi: 10.1172/JCI40104.
    » https://doi.org/10.1172/JCI40104
  • 45
    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, doi: 10.1128/JVI.03364-13.
    » https://doi.org/10.1128/JVI.03364-13
  • 46
    Teo TH, Her Z, Tan JJL, Lum FM, Lee WWL, Chan YH, et al. Caribbean and La Réunion 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, doi: 10.1128/JVI.00909-15.
    » https://doi.org/10.1128/JVI.00909-15
  • 47
    Dupuis-Maguiraga L, Noret M, Brun S, Le Grand R, Gras G, Roques P. Chikungunya disease: infection-associated markers from the acute to the chronic phase of arbovirus-induced arthralgia. PLoS Negl Trop Dis 2012; 6: e1446, doi: 10.1371/journal.pntd.0001446.
    » https://doi.org/10.1371/journal.pntd.0001446
  • 48
    Long KM, Ferris MT, Whitmore AC, Montgomery SA, Thurlow LR, McGee CE, et al. γδ T cells play a protective role in Chikungunya virus-induced disease. J Virol 2016; 90: 433-443, doi: 10.1128/JVI.02159-15.
    » https://doi.org/10.1128/JVI.02159-15
  • 49
    Lucas-Hourani M, Lupan A, Despràs P, Thoret S, Pamlard O, Dubois J, et al. A phenotypic assay to identify Chikungunya virus inhibitors targeting the nonstructural protein nsP2. J Biomol Screen 2013; 18: 172-179, doi: 10.1177/1087057112460091.
    » https://doi.org/10.1177/1087057112460091
  • 50
    Fros JJ, Liu WJ, Prow NA, Geertsema C, Ligtenberg M, Vanlandingham DL, et al. Chikungunya virus nonstructural protein 2 inhibits type I/II interferon-stimulated JAK-STAT signaling. J Virol 2010; 84: 10877-10887, doi: 10.1128/JVI.00949-10.
    » https://doi.org/10.1128/JVI.00949-10
  • 51
    Meshram CD, Lukash T, Phillips AT, Akhrymuk I, Frolova EI, Frolov I. Lack of nsP2-specific nuclear functions attenuates Chikungunya virus replication both in vitro and in vivo Virology 2019; 534: 14-24, doi: 10.1016/j.virol.2019.05.016.
    » https://doi.org/10.1016/j.virol.2019.05.016
  • 52
    Akhrymuk I, Kulemzin S V, Frolova EI. Evasion of the innate immune response: the Old World alphavirus nsP2 protein induces rapid degradation of Rpb1, a catalytic subunit of RNA polymerase II. J Virol 2012; 86: 7180-7191, doi: 10.1128/JVI.00541-12.
    » https://doi.org/10.1128/JVI.00541-12
  • 53
    Dhanwani R, Khan M, Alam SI, Rao PVL, Parida M. Differential proteome analysis of Chikungunya virus‐infected new‐born mice tissues reveal implication of stress, inflammatory and apoptotic pathways in disease pathogenesis. Proteomics 2011; 11: 1936-1951, doi: 10.1002/pmic.201000500.
    » https://doi.org/10.1002/pmic.201000500
  • 54
    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, doi: 10.1093/infdis/jiq006.
    » https://doi.org/10.1093/infdis/jiq006
  • 55
    Poh CM, Chan YH, Ng LFP. Role of T cells in Chikungunya virus infection and utilizing their potential in anti-viral immunity. Front Immunol 2020; 11: 287, doi: 10.3389/fimmu.2020.00287.
    » https://doi.org/10.3389/fimmu.2020.00287
  • 56
    Hawman DW, Stoermer KA, Montgomery SA, Pal P, Oko L, Diamond MS, et al. Chronic joint disease caused by persistent Chikungunya virus infection is controlled by the adaptive immune response. J Virol 2013; 87: 13878-13888, doi: 10.1128/JVI.02666-13.
    » https://doi.org/10.1128/JVI.02666-13
  • 57
    Carissimo G, Teo TH, Chan YH, Lee CYP, Lee B, Torres-Ruesta A, et al. Viperin controls Chikungunya virus-specific pathogenic T cell IFNγ Th1 stimulation in mice. Life Sci Alliance 2019; 2: e201900298, doi: 10.26508/lsa.201900298.
    » https://doi.org/10.26508/lsa.201900298
  • 58
    Lee WWL, Teo TH, Her Z, Lum FM, Kam YW, Haase D, et al. Expanding regulatory T cells alleviates Chikungunya virus-induced pathology in mice. J Virol 2015; 89: 7893-7904, doi: 10.1128/JVI.00998-15.
    » https://doi.org/10.1128/JVI.00998-15
  • 59
    Chua CL, Sam IC, Chiam CW, Chan YF. The neutralizing role of IgM during early Chikungunya virus infection. PLoS One 2017; 12: e0171989, doi: 10.1371/journal.pone.0171989.
    » https://doi.org/10.1371/journal.pone.0171989
  • 60
    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: e0003684, doi: 10.1371/journal.pntd.0003684.
    » https://doi.org/10.1371/journal.pntd.0003684
  • 61
    Kam YW, Simarmata D, Chow A, Her Z, Teng TS, Ong EKS, et al. Early appearance of neutralizing immunoglobulin G3 antibodies is associated with Chikungunya virus clearance and long-term clinical protection. J Infect Dis 2012; 205: 1147-1154, doi: 10.1093/infdis/jis033.
    » https://doi.org/10.1093/infdis/jis033
  • 62
    Amdekar S, Parashar D, Alagarasu K. Chikungunya virus-induced arthritis: role of host and viral factors in the pathogenesis. Viral Immunol 2017; 30: 691-702, doi: 10.1089/vim.2017.0052.
    » https://doi.org/10.1089/vim.2017.0052
  • 63
    Hennessy EJ, Parker AE, O'neill LAJ. Targeting Toll-like receptors: emerging therapeutics? Nat Rev Drug Discov 2010; 9: 293-307, doi: 10.1038/nrd3203.
    » https://doi.org/10.1038/nrd3203
  • 64
    Diebold SS, Brencicova E. Nucleic acids and endosomal pattern recognition: how to tell friend from foe? Front Cell Infect Microbiol 2013; 3: 37, doi: 10.3389/fcimb.2013.00037.
    » https://doi.org/10.3389/fcimb.2013.00037
  • 65
    Dunlevy F, McElvaney NG, Greene CM. TLR3 sensing of viral infection. Open Infect Dis J 2010; 4: 1-10, doi: 10.2174/1874279301004010001.
    » https://doi.org/10.2174/1874279301004010001
  • 66
    Ranjith-Kumar CT, Miller W, Sun J, Xiong J, Santos J, Yarbrough I, et al. Effects of single nucleotide polymorphisms on Toll-like receptor 3 activity and expression in cultured cells. J Biol Chem 2007; 282: 17696-17705, doi: 10.1074/jbc.M700209200.
    » https://doi.org/10.1074/jbc.M700209200
  • 67
    Blasius AL, Beutler B. Intracellular toll-like receptors. Immunity 2010; 32: 305-315, doi: 10.1016/j.immuni.2010.03.012.
    » https://doi.org/10.1016/j.immuni.2010.03.012
  • 68
    Sengupta S, Mukherjee S, Bhattacharya N, Tripathi A. Differential genotypic signatures of Toll-like receptor polymorphisms among dengue-chikungunya mono-and co-infected Eastern Indian patients. Eur J Clin Microbiol Infect Dis 2021; 1-13, doi: 10.1007/s10096-020-04125-x.
    » https://doi.org/10.1007/s10096-020-04125-x
  • 69
    Khanmohammadi S, Rezaei N. Role of Toll‐like receptors in the pathogenesis of COVID‐19. J Med Virol 2021; 93: 2735-2739, doi: 10.1002/jmv.26826.
    » https://doi.org/10.1002/jmv.26826
  • 70
    Rauseo DM, Fernández-Mestre M. X-linked Toll-like receptor 7 polymorphism associated with susceptibility to Chikungunya Fever. Asian Pac J Trop Med 2019; 12: 137, doi: 10.4103/1995-7645.254940.
    » https://doi.org/10.4103/1995-7645.254940
  • 71
    Xiao W, Liu Z, Lin J, Li J, Wu K, Ma Y, et al. Association of Toll‐like receptor 7 and 8 gene polymorphisms with Graves' disease in Chinese Cantonese population. Tissue Antigens 2015; 85: 29-34, doi: 10.1111/tan.12479.
    » https://doi.org/10.1111/tan.12479
  • 72
    Gantier MP, Irving AT, Kaparakis‐Liaskos M, Xu D, Evans VA, Cameron PU, et al. Genetic modulation of TLR8 response following bacterial phagocytosis. Hum Mutat 2010; 31: 1069-1079, doi: 10.1002/humu.21321.
    » https://doi.org/10.1002/humu.21321
  • 73
    Ojeda N, Salazar C, Cárdenas C, Marshall SH. Expression of DC-SIGN-like C-Type lectin receptors in Salmo salar. Dev Comp Immunol 2020; 113: 103806, doi: 10.1016/j.dci.2020.103806.
    » https://doi.org/10.1016/j.dci.2020.103806
  • 74
    Fang X, Hu Z, Shang W, Zhu J, Xu C, Rao X. Genetic polymorphisms of molecules involved in host immune response to dengue virus infection. FEMS Immunol Med Microbiol 2012; 66: 134-146, doi: 10.1111/j.1574-695X.2012.00995.x.
    » https://doi.org/10.1111/j.1574-695X.2012.00995.x
  • 75
    Alagarasu K, Damle IM, Bachal R V, Mulay AP, Shah PS, Dayaraj C. Association of promoter region polymorphisms of CD209 gene with clinical outcomes of dengue virus infection in Western India. Infect Genet Evol 2013; 17: 239-242, doi: 10.1016/j.meegid.2013.04.024.
    » https://doi.org/10.1016/j.meegid.2013.04.024
  • 76
    Pabalan N, Chaisri S, Tabunhan S, Phumyen A, Jarjanazi H, Steiner TS. Associations of DC-SIGN (CD209) promoter-336G/A polymorphism (rs4804803) with dengue infection: A systematic review and meta-analysis. Acta Trop 2018; 177: 186-193, doi: 10.1016/j.actatropica.2017.10.017.
    » https://doi.org/10.1016/j.actatropica.2017.10.017
  • 77
    Chaaithanya IK, Muruganandam N, Anwesh M, Rajesh R, Ghosal SR, Kartick C, et al. HLA class II allele polymorphism in an outbreak of Chikungunya fever in middle Andaman, India. Immunology 2013; 140: 202-210, doi: 10.1111/imm.12128.
    » https://doi.org/10.1111/imm.12128
  • 78
    Thanapati S, Hande A, Das R, Gurav Y, Tripathy AS. Association of human leukocyte antigen class II allele and haplotypes in Chikungunya viral infection in a western Indian population. Trans R Soc Trop Med Hyg 2014; 108: 277-282, doi: 10.1093/trstmh/tru030.
    » https://doi.org/10.1093/trstmh/tru030
  • 79
    Bouquillard E, Combe B. A report of 21 cases of rheumatoid arthritis following Chikungunya fever. A mean follow-up of two years. Joint Bone Spine 2009; 76: 654-657, doi: 10.1016/j.jbspin.2009.08.005.
    » https://doi.org/10.1016/j.jbspin.2009.08.005
  • 80
    Qidwai T, Khan F. Tumour necrosis factor gene polymorphism and disease prevalence. Scand J Immunol 2011; 74: 522-547, doi: 10.1111/j.1365-3083.2011.02602.x.
    » https://doi.org/10.1111/j.1365-3083.2011.02602.x
  • 81
    Nicklin MJH, Weith A, Duff GW. A physical map of the region encompassing the human interleukin-1α, interleukin-1β, and interleukin-1 receptor antagonist genes. Genomics 1994; 19: 382-384, doi: 10.1006/geno.1994.1076.
    » https://doi.org/10.1006/geno.1994.1076
  • 82
    Tripathy AS, Ganu MA, Sonam L, Alagarasu K, Walimbe AM, Thanapati S, et al. Association of IL1RN VNTR polymorphism with Chikungunya infection: a study from Western India. J Med Virol 2019; 91: 1901-1908, doi: 10.1002/jmv.25546.
    » https://doi.org/10.1002/jmv.25546
  • 83
    Hovanessian AG, Justesen J. The human 2′-5′ oligoadenylate synthetase family: unique interferon-inducible enzymes catalyzing 2′-5′ instead of 3′-5′ phosphodiester bond formation. Biochimie 2007; 89: 779-788, doi: 10.1016/j.biochi.2007.02.003.
    » https://doi.org/10.1016/j.biochi.2007.02.003
  • 84
    Bréhin AC, Casadémont I, Frenkiel MP, Julier C, Sakuntabhai A, Despràs P. The large form of human 2′, 5′-Oligoadenylate Synthetase (OAS3) exerts antiviral effect against Chikungunya virus. Virology 2009; 384: 216-222, doi: 10.1016/j.virol.2008.10.021.
    » https://doi.org/10.1016/j.virol.2008.10.021
  • 85
    Kristiansen H, Gad HH, Eskildsen-Larsen S, Despres P, Hartmann R. The oligoadenylate synthetase family: an ancient protein family with multiple antiviral activities. J Interferon Cytokine Res 2011; 31: 41-47, doi: 10.1089/jir.2010.0107.
    » https://doi.org/10.1089/jir.2010.0107
  • 86
    Sadler AJ, Williams BRG. Interferon-inducible antiviral effectors. Nat Rev Immunol 2008; 8: 559-568, doi: 10.1038/nri2314.
    » https://doi.org/10.1038/nri2314
  • 87
    Sengupta S, Bhattacharya N, Tripathi A. Increased CRP, anti-CCP antibody, IL-2R, COMP levels in prognosis of post-chikungunya chronic arthritis and protective role of their specific genotypes against arthritic manifestation. Virus Res 2022; 23: 198998, doi: 10.1016/j.virusres.2022.198998.
    » https://doi.org/10.1016/j.virusres.2022.198998
  • 88
    Volanakis JE. Human C-reactive protein: expression, structure, and function. Mol Immunol 2001; 38: 189-197, doi: 10.1016/S0161-5890(01)00042-6.
    » https://doi.org/10.1016/S0161-5890(01)00042-6
  • 89
    Verma S, Li SH, Badiwala MV, Weisel RD, Fedak PWM, Li RK, et al. Endothelin antagonism and interleukin-6 inhibition attenuate the proatherogenic effects of C-reactive protein. Circulation 2002; 105: 1890-1896, doi: 10.1161/01.CIR.0000015126.83143.B4.
    » https://doi.org/10.1161/01.CIR.0000015126.83143.B4
  • 90
    Sengupta S, Bhattacharya N, Tripathi A. Association of C-reactive protein polymorphisms with serum-CRP concentration and viral load among dengue-chikungunya mono/co-infected patients. Antiviral Res 2022; 197: 105225, doi: 10.1016/j.antiviral.2021.105225.
    » https://doi.org/10.1016/j.antiviral.2021.105225

Publication Dates

  • Publication in this collection
    08 Sept 2023
  • Date of issue
    2023

History

  • Received
    13 Feb 2023
  • Accepted
    22 July 2023
Associação Brasileira de Divulgação Científica Av. Bandeirantes, 3900, 14049-900 Ribeirão Preto SP Brazil, Tel. / Fax: +55 16 3315-9120 - Ribeirão Preto - SP - Brazil
E-mail: bjournal@terra.com.br