Chikungunya virus infection: an overview

 
CONTINUE READING
Chikungunya virus infection: an overview
NEW MICROBIOLOGICA, 36, 211-227, 2013

             Chikungunya virus infection: an overview
                        Claudia Caglioti, Eleonora Lalle, Concetta Castilletti, Fabrizio Carletti,
                                       Maria Rosaria Capobianchi, Licia Bordi
                 Laboratory of Virology, “L. Spallanzani” National Institute for Infectious Diseases, Rome, Italy

                                                          SUMMARY

    Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus belonging to the Togaviridae family, first isolated
    in Tanzania in 1952. The main vectors are mosquitoes from the Aedes species. Recently, the establishment of an en-
    velope mutation increased infectivity for A. albopictus. CHIKV has recently re-emerged causing millions of infections
    in countries around the Indian Ocean characterized by climate conditions favourable to high vector density. Importation
    of human cases to European regions with high density of suitable arthropod vectors (such as A. albopictus) may trig-
    ger autochthonous outbreaks. The clinical signs of CHIKV infection include non-specific flu-like symptoms, and a char-
    acteristic rash accompanied by joint pain that may last for a long time after the resolution of the infection. The death
    rate is not particularly high, but excess mortality has been observed in concomitance with large CHIKV outbreaks. De-
    regulation of innate defense mechanisms, such as cytokine inflammatory response, may participate in the main clin-
    ical signs of CHIKV infection, and the establishment of persistent (chronic) disease. There is no specific therapy, and
    prevention is the main countermeasure. Prevention is based on insect control and in avoiding mosquito bites in en-
    demic countries. Diagnosis is based on the detection of virus by molecular methods or by virus culture on the first days
    of infection, and by detection of an immune response in later stages. CHIKV infection must be suspected in patients
    with compatible clinical symptoms returning from epidemic/endemic areas. Differential diagnosis should take into ac-
    count the cross-reactivity with other viruses from the same antigenic complex (i.e. O’nyong-nyong virus).

    KEY WORDS: CHIKV, Arbovirus, Virus dissemination, Immunopathogenesis, Geographic distribution, Diagnosis,
    Treatment and prevention.

Received May 26, 2013                                                                                         Accepted May 30, 2013

INTRODUCTION                                                       ceptors (Strauss and Strauss, 1994), followed by
                                                                   clathrin-mediated endocytosis of the attached par-
Chikungunya virus (CHIKV), an arbovirus trans-                     ticle (Lee et al., 2013), low pH-mediated membrane
mitted by mosquito vectors, is an alphavirus be-                   fusion and delivery of the viral nucleocapsid into
longing to the Togaviridae family. Alphaviruses                    the cytoplasm (Sorisseau et al., 2007). To date no
are small spherical enveloped viruses, with a 60-                  CHIKV interacting protein has been characterized,
70 nm diameter. The genome is a single-strand                      but in a very recent study, Wintachai et al. identi-
RNA molecule of positive polarity, encoding four                   fied prohibitin as CHIKV-binding protein ex-
non structural (nsP1-4) and three structural pro-                  pressed by microglial cells (Wintachai et al., 2012).
teins (C, E1, E2). Viral replication is initiated by               The replication cycle is fast, taking around 4 hours.
attachment of the viral envelope to host cell re-                  Alphaviruses are sensitive to dissecation and to
                                                                   temperatures above 58°C (Strauss and Strauss,
                                                                   1994; Khan et al., 2002). About 30 species of arthro-
Corresponding author                                               pod-borne viruses are included in the alphavirus
Maria Rosaria Capobianchi                                          genus, antigenically classified into 7 complexes.
Laboratory of Virology                                             These viruses are widely distributed throughout
Padiglione Baglivi
                                                                   the world, with the exception of Antarctica.
National Institute for Infectious Diseases
INMI “L. Spallanzani”                                              Besides CHIKV, several arthropod-transmitted
Via Portuense, 292 - 00149 Rome, Italy                             alphaviruses cause human disease, characterized
E-mail: maria.capobianchi@inmi.it                                  by similar clinical presentation: Barmah Forest
Chikungunya virus infection: an overview
212                       C. Caglioti, E. Lalle, C. Castilletti, F. Carletti, M.R. Capobianchi, L. Bordi

(BFV) and Ross River viruses (RRV) (Oceania),                      GEOGRAPHIC DISTRIBUTION
O’nyong-nyong (ONNV) and Semliki Forest virus-
es (SFV) (Africa), Mayaro (South America),                         CHIKF has an epidemiological pattern with both
Sindbis (SINV) and Sindbis-like viruses (Africa,                   sporadic and epidemics cases in West Africa,
Asia, Scandinavia and Russia) (Taubiz et al.,                      from Senegal to Cameroun, and in many other
2007). Chikungunya fever (CHIKF) derives its                       African countries (Democratic Republic of
name from Makonde, a language spoken in south                      Congo, Nigeria, Angola, Uganda, Guinea, Malawi,
Tanzania, and means “that which bends up”, re-                     Central African Republic, Burundi, and South
ferring to the posture of patients afflicted with                  Africa). Moreover, many epidemics occurred in
severe joint pain characterizing this infection.                   Asia (Burma, Thailand, Cambodia, Vietnam,
First isolated in Tanzania in 1952 (Robinson,                      India, Sri Lanka, Timor, Indonesia, and the
1955), CHIKV attracted worldwide attention                         Philippines) in the 1960s and in the 1990s
when it caused a massive outbreak in the Indian                    (Pialoux et al., 2007; Jain et al., 2008).
Ocean islands (Enserik, 2006). Since 1952,                         Major epidemics appear and disappear cyclical-
CHIKV has caused a number of epidemics, both                       ly, usually with an inter-epidemic period rang-
in Africa and in Southeast Asia, many of them in-                  ing from 7 to 20 years. The huge outbreak that
volving hundreds of thousands of people. After a                   increased concern about CHIKV started in
few years of relative dormancy in La Réunion                       Kenya in 2004, where the seroprevalence rates
Island, CHIKV transmission has restarted, re-                      reached 75% in Lamu island (Pialoux et al.,
newing concerns about the possibility of renewed                   2007), before reaching the Comores, Seychelles,
autochthonous transmission in Mediterranean                        and Mauritius islands. The virus reached La
countries.                                                         Réunion island in March-April 2005, probably

FIGURE 1 - Geographic distribution of CHIKV shown in the most recent map (May 2012) retrieved from the CDC
website (http://www.cdc.gov/chikungunya/map/index.html, last accessed May 2013).
Chikungunya overview                                        213

as a result of importation of cases among immi-           viding information on the emergence or re-emer-
grants from the Comores and rapidly spread to             gence of an infectious pathogen in a source re-
several countries in the Indian Ocean and India           gion, and can be used to map the location, dy-
(Enserik, 2006; Mavalankar et al., 2007).                 namics and movement of pathogenic strains
Compared to earlier outbreaks, this episode was           (Pistone et al., 2009).
massive, occurred in highly medicalized areas             The geographic range of CHIKV is mainly in
such as La Réunion, and had very significant              Africa, Asia and Australia (Figure 1).
economic and social impact. Since the beginning
of the outbreak in the Indian Ocean region, more
than 1,000 imported CHIKV cases have been de-             PHYLOGENESIS
tected among European and American travellers
returning from the affected areas (Fusco et al.,          Three lineages of CHIKV, with distinct genotyp-
2006; Taubiz et al., 2007), giving rise, in 2007, to      ic and antigenic characteristics, have been iden-
the first autochthonous (human-to-mosquito-to-            tified. Isolates that caused the 2004-06 Indian
human transmission) European outbreak in Italy            Ocean outbreak form a distinct cluster within the
(Rezza et al., 2007; Charrel and de Lambellerie,          large eastern/central Africa (ECSA) phylogenetic
2008). During the period December 2006-July               group, in addition to the Asian and west African
2009, no confirmed cases were detected on La              phylogenetic groups (Powers et al., 2000;
Réunion and Mayotte Islands, but new outbreaks            Schuffenecker et al., 2006).
were reported in Madagascar. After a few years            The divergence of each distinct lineage reflects,
of relative dormancy in La Réunion, CHIKV                 to some extent, the path of global transmission
transmission restarted in 2009 and 2010, lead-            and occasional outbreaks. According to phyloge-
ing to re-importation to Europe (May 2010)                netic analysis performed by Volk and colleagues
(D’Ortenzio et al., 2011).                                (2010), the currently circulating CHIKV strains
During the last three years (2011-2013) concerns          have an ancestor that existed within the last 500
about Chikungunya outbreaks arose again due               years. Interestingly, despite their close geographic
to increasing number of CHIKV infections, start-          distance, the two African lineages did not cluster
ing from 2011, when a massive outbreak with               together, indicating limited genetic exchange be-
more than 11,000 cases occurred in the Republic           tween the two lineages in Africa. The only ex-
of Congo (Brazzaville) (ProMED-mail:                      ception was a 1963 bat isolate from Senegal,
20110613.1806). During 2012, 29 cases of CHIKV            which grouped in the ECSA clade. This finding
infection were reported in India (Rajasthan)              is the first to suggest that the main West African
(ProMED-mail: 20120716.1203694), and two ad-              and ECSA lineages may overlap spatially in the
ditional outbreaks were recorded: one in                  enzootic cycle, at least occasionally (Volk et al.,
Cambodia, with almost 1,500 cases (ProMED-                2010).
mail: 20120920.1303166) and one in the main is-           Moreover, phylogenetic analysis of CHIKV strains
land of Papua New Guinea, with a total of 633             circulating in A. albopicus-human transmission
suspected cases (ProMED-mail: 20121010.                   cycles, obtained during outbreaks, identified the
1335814); Bali has also had sporadic outbreaks            independent acquisition of a common mutation
(ProMED-mail: 20130320.1594512). In Samar                 in E1 glycoprotein (E1gp), namely A226V, in
(Philippines) 600 cases were recorded in 2012,            strains isolated from different geographic regions
but in 2013 the infection rate has been increas-          (Schuffenecker et al., 2006; de Lambellerie et al.,
ing, with 500 cases recorded until March; these           2008a). This mutation, together with M269V and
numbers appear to be increasing day by day                D284E E1gp mutations, have been described as
(ProMED-mail: 20130128.1518853). Considering              molecular signatures of the Indian Ocean out-
the capacity of CHIKV to emerge, re-emerge, and           break (Arankalle et al., 2007; Tsetsarkin et al.,
quickly spread in novel areas, heightened sur-            2007; Vazeille M, 2007). In particular, the A226V
veillance and preparedness seem to be a priori-           mutation, which was absent in the strains isolat-
ty. In particular, travellers act as carriers who in-     ed during the initial phases of the outbreak in La
advertently ferry pathogens between countries.            Réunion, appeared in >90% of the isolates after
They can thus serve as a sentinel population pro-         December 2005. This change could be related to
214                          C. Caglioti, E. Lalle, C. Castilletti, F. Carletti, M.R. Capobianchi, L. Bordi

virus adaptation to the mosquito vector species                       widespread distribution of this vector, particu-
(see below). Together with the lack of herd im-                       larly in Italy (Knudsen, 1995). In a previous pa-
munity, this might explain the abrupt and esca-                       per we characterized 7 viral isolates (5 imported
lating nature of the La Réunion outbreak. The                         and 2 autochthonous cases) with respect to the
A226V mutation was clearly demonstrated to in-                        molecular E1 signatures of the Indian Ocean
crease viral fitness in the A. albopictus vector                      Outbreak, particularly the A226V mutation. These
(Vazeille et al., 2007; Tsetsarkin et al., 2007) that,                isolates had been obtained from 3 travellers re-
in turn, may expand the potential for CHIKV to                        turning from Mauritius in 2006, 2 returning from
diffuse to the Americas and Europe, due to the                        India in 2006 and 2007, and 2 autochthonous cas-

FIGURE 2 - Phylogenetic tree of CHIKV strains performed on partial E1 gene. Sequences of a 1013 bp fragment of
E1 gene (nucleotide positions 10145-11158, with respect to the reference strain S27). The CHIKV strains isolated from
human cases in Italy (3 strains deriving from patients returning to Italy from Mauritius, 2 strains from patients re-
turning from India, 2 strains from patients involved in the 2007 Italian outbreak) are indicated with the strain name
in bold. Their GenBank accession numbers are: EU188924 for ITA1_TAM_06; EU190879 for ITA2_BMI_06; EU190881
for ITA3_CGO_06; EU190884 for ITA4_MRA_06; EU272130 for ITA5_JEM_07; EU272132 for ITA7_BI_07; EU272133
for ITA8_VEN_07 (Bordi et al., 2008). The sequences used for comparison are indicated with their GenBank acces-
sion number. CHIKV strains carrying the A226V mutation are underlined.
Chikungunya overview                                           215

es that occurred during the 2007 Italian outbreak           the Indian Ocean region, India and then
(Bordi et al., 2008) (Figure 2).                            Southeast Asia since 2005. As previously stated,
All the strains isolated in Italy, both imported and        a mutation in the E1gp gene, that results in the
autochthonous, displayed two molecular signa-               A226V amino acid substitution, dramatically in-
tures of the Indian Ocean outbreak (M269V and               creased the infectivity of some epidemic strains
D284E). The A226V mutation was present in all               for an alternative urban vector, A. albopictus
imported and autochthonous cases, with the ex-              (ProMED archive 20100926.3495). Therefore, the
ception of the isolate imported from the Indian             urban transmission cycle relies only on A. aegyp-
subcontinent in 2006. The absence of this muta-             ti and/or A. albopictus, anthropophilic vectors that
tion in the isolate imported in 2006 from India             can initiate human-mosquito-human transmis-
was in agreement with published data (Arankalle             sion, and human amplification hosts. This en-
et al., 2007), and with available GenBank se-               demic/epidemic cycle results in high levels of hu-
quence data indicating that the virus strains cir-          man exposure to mosquito transmission, partic-
culating in India in 2006 lacked this mutation.             ularly because these vectors live in close proxim-
The presence of A226V in the isolate imported               ity to people. The behaviour and ecology of A. ae-
from India in July 2007 and in the isolates from            gypti, in particular, are ideal for epidemic trans-
the 2007 Italian outbreak (originating from a case          mission because adult females prefer to feed on
imported from India) supports the view that the             humans, often take several blood meals during a
virus envelope sequence of strains from India               single gonotrophic cycle, oviposit in artificial con-
changed over time, acquiring the E1 mutation as-            tainers as their preferred larval sites, and rest in-
sociated with enhanced fitness in A. albopictus             side houses with ready access to human hosts
after 2006. So it appears that the acquisition and          (Weaver et al., 2012).
fixation of the A226V mutation may be a com-                A. albopictus is zoophilic and anthropophilic, ag-
mon pathway of Chikungunya explosion in epi-                gressive, silent, active all-day long, and has a lifes-
demic areas, in a parallel interplay with the mos-          pan longer than other mosquitoes (up to 8
quito vector dynamics. It is noteworthy that the            weeks). In recent decades it has expanded to sev-
outbreak in Singapore, where the A226V muta-                eral areas previously known to be Aedes-free
tion was absent, was rapidly controlled.                    (Charrel et al. 2007). It seems that most new in-
                                                            troductions of A. albopictus have been caused by
                                                            vegetative eggs contained in timber and tyres ex-
VECTOR AND RESERVOIR                                        ported from Asia throughout the world. Other
                                                            emerging events also contributed to the intro-
Two distinct transmission cycles have been well             duction of A. albopictus mosquitoes into previ-
documented for CHIKV: an enzootic sylvatic cy-              ously unaffected areas, such as climate change
cle and an endemic/epidemic urban cycle. The                and the increasing use of plastic containers in de-
African sylvatic cycle likely involves several ar-          veloping countries. Indeed, climate changes may
boreal Aedes mosquitoes species as vectors (A.              have several effects on vector biology: increasing
furcifer, A. vittatus, A. fulgens, A. luteocephalus, A.     temperatures may improve survival at higher lat-
dalzieli, A. vigilax, A. camptorhynchites) and non-         itudes and altitudes, increase the growth rates of
human primates as reservoir/amplifying hosts. In            vector populations, and alter their seasonality; in-
Africa, the enzootic transmission cycle can spill           creased rainfall may have an effect on the larval
over to infect people who live nearby, and en-              habitat and population size, and finally an in-
zootic mosquito vectors may be involved in in-              crease in humidity could favourably affect vector
ter-human transmission during small outbreaks.              survival (Gubler et al., 2001). The use of plastic
A. furcifer, probably a principal enzootic vector, is       containers in developing countries, where they
known to enter human villages (Diallo, 1999),               are usually not correctly disposed of and remain
where it presumably transmits the virus from                in the environment for years, has also been linked
monkeys to humans (Peyrefitte et al., 2007;                 with the spread of the mosquitoes: acting as rain-
Peyrefitte et al., 2008). Endemic/epidemic trans-           water receptacles, and being exposed to sunlight,
mission cycles were established when the virus              they can become perfect “incubators” for mos-
was introduced into Asia around 1950, and into              quito eggs, where the ideal conditions of tem-
216                          C. Caglioti, E. Lalle, C. Castilletti, F. Carletti, M.R. Capobianchi, L. Bordi

perature and humidity are achieved easily and                         hibitors of chemokine pathways associated with
naturally.                                                            monocyte/macrophage recruitment may be a
Human beings serve as the main CHIKV reser-                           promising approach in humans, to be further ex-
voir during epidemic periods. In Africa some an-                      plored.
imals (monkeys, rodents, and birds) constitute                        It is widely recognized that passive vaccination is
the virus reservoir during non-epidemic periods,                      an appropriate preventive and therapeutic option
sustaining virus circulation in the environment                       for many viral infections in humans, including
in the absence of human cases. Outbreaks might                        those spread by viral vertical transmission, espe-
occur in monkeys when herd immunity is low;                           cially when no alternative therapy is available
the animals develop viremia but no pronounced                         (Dessain et al., 2008). CHIKV infection seems to
physical manifestations (Wolfe et al., 2001; Inoue                    elicit long-lasting protective immunity, and ex-
et al., 2003). An animal reservoir has not been                       periments performed using animal models have
identified in Asia, where humans appear to be                         shown a partial cross-protection among CHIKV
the only host.                                                        and other alphaviruses (Hearn and Rainey, 1963;
                                                                      Edelman et al., 2000). Since human polyvalent im-
                                                                      mune globulins, purified from plasma samples ob-
TREATMENT AND PREVENTION                                              tained from donors in the convalescent phase of
                                                                      CHIKV infection, exhibited high neutralizing ac-
There are no specific drugs against CHIKV and                         tivity in vitro and a powerful prophylactic and ther-
patients are symptomatically treated with non-                        apeutic efficacy against CHIKV infection in in vi-
steroidal anti-inflammatory drugs, fluids, and                        vo mouse models (Couderc et al., 2009), it could be
medicines to relieve symptoms of fever and                            used in humans for prevention and treatment, es-
aching, such as ibuprofen, naproxen, acetamino-                       pecially in individuals at risk of severe CHIKV dis-
phen, or paracetamol. Steroids have occasional-                       ease, such as neonates born to viremic mothers
ly been used but their efficacy was not significant                   and adults with underlying conditions. Polyclonal
(Taubitz et al., 2007). Some time ago chloroquine,                    immune globulins present the advantage of a
a drug useful for prophylaxis and treatment of                        broad reactivity but the therapeutic intervention is
malaria, showed promising results for treating                        limited, due to the short viremia in the acute phase
chronic Chikungunya arthritis (Brighton, 1984),                       of CHIKV infection: thus the only benefit this
even if a further trial conducted on La Réunion                       treatment has to offer would be to help reducing
Island proved that there was no justification for                     viremia faster (Kam et al., 2009). As an alternative
the use of chloroquine to treat acute                                 approach, more specific human monoclonal an-
Chikungunya disease (de Lamballerie et al.,                           tibodies (MAbs) could be used. In a recent study
2008b); overall, the usefulness of chloroquine                        two unique human MAbs, specific for the CHIKV
treatment remains unclear. Ribavirin (200 mg                          E1gp, strongly and specifically neutralized CHIKV
twice a day for seven days) given to patients who                     infection in vitro (Warter et al., 2011).
continued to have crippling lower limb pains and                      To date a number of CHIKV vaccines have been
arthritis for at least two weeks after a febrile                      developed, but none have been licensed. While a
episode, seems to be effective against CHIKV,                         number of significant questions remain to be ad-
leading to faster resolution of joint and soft tissue                 dressed related to vaccine validation, such as the
manifestations (Ravichandran and Manian,                              most appropriate animal models (species, age,
2008). Briolant and colleagues (2004) screened                        immune status), the dose and route of immu-
various active antiviral compounds against virus-                     nization, the potential interference from multi-
es of the alphavirus genus in vitro and demon-                        ple vaccinations against different viruses, and
strated that 6-azauridinet was more effective than                    lastly, the practical cost of the vaccine, since most
ribavirin against CHIKV. Moreover, the combi-                         of the epidemic geographical regions belong to
nation of interferon (IFN)-α2b and ribavirin had                      the developing countries, there is real hope that
a synergistic antiviral effect on CHIKV (Briolant                     a vaccine to prevent this disease will not be too
et al., 2004). Since inhibitors of monocyte chemo-                    long in arriving.
taxis can greatly alleviate alphavirus-induced                        Although no licensed vaccines are currently avail-
arthritides in mice (Rulli et al., 2009) the use of in-               able for CHIKV, potential vaccine candidates
Chikungunya overview                                          217

have been tested in humans and animals with                a photoactive hydrophobic azide molecule that
varying success. Several vaccine strategies have           they used in a previous study (Sharma et al.,
been undertaken:                                           2007) to completely inactivate VEEV, in addition
1. whole inactivated virus preparation;                    to UV irradiation. The INA-inactivated
2. attenuated live vaccines;                               CHIKV181/25 formulation may address the issue
3. recombinant proteins or virus like particles;           of residual virulence associated with live attenu-
4. DNA vaccination.                                        ated CHIKV181/25, but the INA-inactivation re-
Due to the ease of preparation, the first developed        sults in a relatively weaker binding capacity of
vaccines were formulations of whole-virus grown            CHIKV181/25 to the neutralizing polyclonal an-
in cell cultures and inactivated either by forma-          ti-CHIKV E2 glycoprotein (E2gp) so that further
lin or tween-ether (Harrison et al., 1967; Eckels et       investigations are necessary (Sharma et al., 2012).
al., 1970; Harrison et al., 1971; White et al., 1972).     Alternative genetic strategies such as viral
Further vaccines focused on attenuated strains             chimeras offer the promise of more stable atten-
of CHIKV obtained after serial passages in cell            uation (Kennedy et al., 2011). For instance, a re-
cultures (Levitt et al., 1986; Edelman et al., 2000).      cent study showed that chimeric alphaviruses,
One of these promising candidates is TSI-GSD-              encoding CHIKV-specific structural genes (but
218, a serially passaged and plaque-purified live          no structural or nonstructural proteins capable
CHIKV vaccine, tested for safety and immuno-               of interfering with development of cellular an-
genicity in human Phase II trials by the US Army           tiviral response), induce protective immune re-
Medical Research Institute (Edelman et al., 2000).         sponse against subsequent CHIKV challenge
Some chimeric candidate vaccines were devel-               (Wang et al., 2011).
oped using either Venezuelan Equine                        A novel CHIKV vaccine candidate, CHIKV/IRES
Encephalitis virus (VEEV) attenuated vaccine               (internal ribosome entry site), was generated by
strain TC-83, a naturally attenuated strain of             manipulation of the structural protein expression
Eastern Equine Encephalitis virus, or SINV as a            of a wt-CHIKV strain via the encephalomyocardi-
backbone and the structural protein genes of               tis virus IRES, and exhibited a high degree of
CHIKV. Vaccinated mice were fully protected                murine attenuation that was not dependent on an
against disease and viremia after CHIKV chal-              intact IFN type I response, highly attenuated and
lenge (Wang et al., 2008). Traditional attenuation         efficacious after a single dose (Plante et al., 2011).
approaches, relying on cell culture passages, typ-         Another approach, recently undertaken by Akata
ically result in attenuation that depends only on          and colleagues (2010), was the use of virus-like
small numbers of attenuating point mutations.              particles (VLPs) expressing CHIKV structural pro-
In addition to the risk of reactogenicity, attenua-        teins that resemble replication-competent al-
tion based on small numbers of mutations can               phaviruses (Akahata et al., 2010). Immunization
also result in residual alphavirus infectivity for         of monkeys with these VLPs elicited neutralizing
mosquito vectors. This risk, underscored by the            antibodies against envelope proteins from different
isolation of the TC-83 VEEV vaccine strain from            CHIKV strains that could confer passive protec-
mosquitoes in Louisiana during an equine vacci-            tion against lethal CHIKV challenge into new mice.
nation campaign designed to control the 1971               The last frontier in the approach of CHIKV vac-
epidemic (Pedersen et al., 1972), is especially high       cine design is the DNA vaccine strategy. An adap-
when a vaccine that relies on a small number of            tive constant-current electroporation technique
point mutations is used in a non-endemic loca-             was used to immunize mice (Muthumani et al.,
tion that could support a local transmission cycle.        2008) and rhesus macaques (Mallilankaraman et
In 2012, the United States Army developed and              al., 2011) with an intramuscular injection of plas-
tested a live attenuated strain of CHIKV,                  mid coding for the CHIKV-capsid, E1 and E2.
CHIKV181/25         for     vaccine    application.        Vaccination induced robust antigen-specific cellu-
CHIKV181/25 demonstrated an excellent im-                  lar and humoral immune responses in both cases.
munogenic profile, however, transient arthralgia           Kumar and colleagues (2012) aimed to develop
was observed in about 8% of vaccine recipients.            candidate vaccines following two different strate-
Sharma and colleagues tried to inactivate                  gies: one based on recombinant E2gp; the other
CHIKV181/25 with 1,5 iodonapthyl azide (INA),              based on chemically inactivated whole virus, both
218                        C. Caglioti, E. Lalle, C. Castilletti, F. Carletti, M.R. Capobianchi, L. Bordi

with promising results (Kumar et al., 2012). Since                  the thorax. The clinical presentation may also in-
a vaccine is not currently available, protection                    volve facial oedema and, in children, a bullous
against mosquito bites and vector control are the                   rash with pronounced sloughing, localised pe-
main preventive measures. Individual protection                     techiae and gingivorrhagia (Fourie and Morrison,
relies on the use of mosquito repellents and meas-                  1979; Brighton et al., 1983). Radiological findings
ures to limit skin exposure to mosquitoes.                          are normal, and biological markers of inflamma-
Bednets should be used during the night in hos-                     tion (erythrocyte sedimentation rate and C-reac-
pitals and day-care facilities but Aedes mosqui-                    tive protein) are normal or moderately elevated
toes are active all-day long. Control of both adult                 (Fourie and Morrison, 1979; Kennedy et al., 1980).
and larval mosquito populations uses the same                       Iridocyclitis and retinitis are the most common
model as for dengue and has been relatively ef-                     ocular manifestations associated with CHIKF; less
fective in many countries and settings. Breeding                    frequent ocular lesions include episcleritis. All oc-
sites must be removed, destroyed, frequently                        ular manifestations have a benign course with
emptied, and cleaned or treated with insecticides.                  complete resolution and preservation of vision.
Control of A. aegypti has rarely been achieved and                  Retinitis shows gradual resolution over a period of
never sustained (Reiter et al., 2006). Recent data                  6 to 8 weeks (Mahendradas et al., 2008). Erratic,
show the different degrees of insecticide resist-                   relapsing, and incapacitating arthralgia is the hall-
ance in A. albopictus and A. aegypti (Cui et al.,                   mark of Chikungunya, although it rarely affects
2006). Large-scale prevention campaigns using                       children. These manifestations are normally mi-
dichlorodiphenyltrichloroethane have been ef-                       gratory and involve the small joints of hands,
fective against A. aegypti but not A. albopictus.                   wrists, ankles, and feet with pain on movement.
However, vector control is an endless, costly, and                  The symptoms generally resolve within 7-10 days,
labor-intensive measure and is not always well                      except for joint stiffness and pain: up to 12% of
accepted by local populations, whose coopera-                       patients still have chronic arthralgia three years
tion is crucial. Control of CHIKV infection, oth-                   after onset of the illness. Arthralgia experienced
er than use of drugs for treatment of disease, de-                  by CHIKV patients closely resembles the
velopment of vaccines, individual protection from                   symptoms induced by other viruses like RRV and
mosquitoes and vector control programs, also in-                    BFV (et al., 2002; Jacups et al., 2008).
volves surveillance that is fundamental for early                   Neurological complications such as meningo-en-
identification of cases and quarantine measure-                     cephalitis were reported in a few patients during
ment. A model used in investigation of the trans-                   the first Indian outbreak in 1973, and during the
mission potential of CHIKV in Italy has proven                      2006 Indian outbreak (Chatterjee et al., 1965 a,
useful to provide insight into the possible impact                  b; Ravi, 2006). Moreover, during the 2006 Indian-
of future outbreaks in temperate climate regions                    Ocean outbreak, rare cases of Guillain-Barré
and the effectiveness of the interventions per-                     syndrome associated with CHIKV infection have
formed during the outbreak (Poletti et al., 2011).                  been described (Wielanek et al., 2007; Lebrun et
                                                                    al., 2009). The possible mechanisms underlying
                                                                    these processes remain unknown, even if it was
CLINICAL MANIFESTATIONS                                             found that mouse CNS tissues such as the cho-
                                                                    roid plexi could also be targets of CHIKV, len-
After infection with CHIKV, there is a silent in-                   ding more credence to the fact that CHIKV in-
cubation period lasting about 2-4 days (range 1-12                  fections do affect CNS cells and tissues (Couderc
days) (Lam et al., 2001). Clinical onset is abrupt,                 et al., 2008). Other rare complications described
with high fever, headache, back pain, myalgia,                      after CHIKV infection are mild hemorrhage,
and arthralgia; the latter can be intense, affecting                myocarditis, and hepatitis (Lemant et al., 2008).
mainly the extremities (ankles, wrists, phalanges)                  CHIKV is not generally considered a life-threat-
but also the large joints (Robinson, 1955; Lam et                   ening disease. Usually the clinical course is fair-
al., 2001; Hochedez et al., 2006; Quatresous, 2006;                 ly mild, but fatal cases directly or indirectly linked
Saxena et al., 2006). Skin involvement is present                   to infection with CHIKV were observed during
in about 40-50% of cases, and consists of a pru-                    the Indian-Ocean outbreak (Josseran et al., 2006).
riginous maculopapular rash predominating on                        The main evidence of a mortality linked to
Chikungunya overview                                       219

CHIKF epidemics was obtained in La Réunion,              lymph circulation and then to the blood through
Mauritius, and India, by comparing expected and          the thoracic duct. Once in the blood, the virus
observed mortality data. In all cases, during the        will have access to various parts of the body, in-
months when the epidemics were raging, the ob-           cluding the liver, muscle, joints and brain. In
served mortality significantly exceeded the ex-          these tissues, the infection is associated with a
pected rate. In particular, in La Réunion the            marked infiltration of mononuclear cells, in-
monthly crude death rates in February and                cluding macrophages, that can be considered
March 2006 were respectively 34.4% and 25.2%             Trojan horses for virus spread to sanctuary body
higher than expected. This corresponded to 260           sites. The pathological events associated with tis-
excess deaths (an increase of 18.4%) with a rough        sue infection are mostly subclinical in the liver
estimate of the case-fatality rate for CHIKF of          (hepatocyte apoptosis) and lymphoid organs
≈1/1,000 cases. The case-fatality rate calculated        (adenopathy), whereas mononuclear cell infil-
on increased crude death rates in Mauritius and          tration and viral replication in the muscles and
Ahmedabad, India, is substantially higher than           joints are associated with very strong pain, with
that calculated in La Réunion: approximately             some patients presenting arthritis (Dupuis-
4.5% (15,760 confirmed or suspected cases and            Maguiraga et al., 2012).
743 excess deaths) and 4.9% (60,777 confirmed or         During the first week of CHIKV infection, viremia
suspected cases and 2,944 excess deaths), re-            can reach very high levels (viral loads of 3.3×109
spectively (Beesoon et al., 2008; Mavalankar et al.,     copies/ml) (Parola et al., 2006). Thus, it remains
2008). These differences may be attributed to            unclear if the virus detected in the blood is re-
many factors (greater disease severity, preexisting      leased from virus-infected peripheral blood
patient conditions, different patient management,        mononuclear cells, or is spilled out from other
or coincident excess deaths from other causes)           replication sites.
but may also be due to a different efficacy of the
surveillance systems for CHIKF, that probably
worked poorly in Mauritius and India, leading to         IMMUNOPATHOGENESIS
underestimation of the total number of cases
(Fusco et al., 2010). The possible link between          The innate immune response is the first barrier
CHIKV infection and multiorgan failure is still          against viruses, being able to inhibit viral repli-
under investigation.                                     cation through cytolytic and non-cytolytic mech-
                                                         anisms. The IFN system plays an important role
                                                         in limiting virus spread at an early stage of in-
VIRUS DISSEMINATION                                      fection. In vitro growth of all alphaviruses can be
AND TARGET ORGANS                                        greatly suppressed by the antiviral effects of IFN-
                                                         α/β when it is added to cells prior to infection
Following intradermal inoculation by infected            (Sourisseau et al., 2007; Courderc et al., 2008;
mosquitoes, CHIKV directly enters the subcuta-           Schilte et al., 2010). The finding that aberrant
neous capillaries where its replication starts im-       Type I interferon signalling in mice led to severe
mediately (Figure 3) with some viruses infecting         forms of CHIKF (Couderc et al., 2008) further
susceptible cells in the skin, such as macrophages       highlighted the important roles cytokines play in
or fibroblasts and endothelial cells. Local viral        the pathology of CHIKV infection.
replication seems to be minor and limited in time,       Moreover, in a recent study Wauquier and col-
with the locally produced virus probably being           leagues (2011) demonstrated that CHIKV infec-
transported to secondary lymphoid organs close           tion in humans elicits strong innate immunity in-
to the site of inoculation, where infected migra-        volving the production of numerous proinflam-
tory cells produce new viruses which can, in turn,       matory mediators. Interestingly, high levels of
infect susceptible resident cells. Even if the host      Interferon IFN-α were consistently found.
is mounting a response to control the virus in the       Production of interleukin (IL), IL-4, IL-10, and
skin dermis, the virus disseminates quite rapidly        IFN-γ suggested the engagement of the adaptive
to the blood circulatory system. Viruses produced        immunity. This was confirmed by flow cytometry
in the draining lymph nodes are released into the        of circulating T lymphocytes that showed a CD8+
220                        C. Caglioti, E. Lalle, C. Castilletti, F. Carletti, M.R. Capobianchi, L. Bordi

FIGURE 3 - Schematic representation of CHIKV dissemination to different tissues and organs.

T lymphocyte response in the early stages of the                    for viral dissemination in other sanctuary body
disease and a CD4+ T lymphocyte-mediated re-                        sites, such as the nervous system, and, in turn, may
sponse in the later stages (Wauquier et al., 2011).                 contribute to the development of clinical manifes-
CHIKV interactions with monocytes and with oth-                     tations mediated by excess immune response.
er blood leukocytes induced a robust and rapid in-                  Usually, CHIKF is a self-limiting disease, with a
nate immune response with the production of spe-                    defined duration of clinical course (7-10 days).
cific chemokines and cytokines, including IFN-α.                    Recovery is associated with a vigorous immune
The involvement of monocytes during the early                       response, that may confer protection from re-in-
phase of CHIKV infection in vivo is massive, and                    fection. However, in some cases, chronic disease
infected monocyte/macrophages migrate in the                        (arthralgia) may be established. Chronic symp-
synovial tissues of chronically CHIKV-infected pa-                  toms may persist even after clearance of the virus
tients, where they contribute to the inflammation                   from the blood, but it is possible that an active
process. This may explain the persistence of joint                  viral reservoir persists locally in the joints. Five
symptoms despite the short duration of viremia                      studies have tried to identify the factors associ-
(Her et al., 2010). Infected monocyte/                              ated with chronic Chikungunya disease in groups
macrophages may be the main cells responsible                       of patients in Singapore (Chow et al., 2011), La
Chikungunya overview                                         221

Réunion (Hoarau et al., 2010), Dakshina Kannada           fection of insect cells (Tsetsarkin et al., 2007). A
(India) (Manimunda et al., 2010; Chaaitanya et            recent study by our group investigated the possi-
al., 2011), and Emilia Romagna (Italy) (Kelvin et         ble involvement of A226V mutation in enhanc-
al., 2011).                                               ing human pathogenesis by testing the replica-
Regulatory mechanisms silencing the vigorous              tion competence in primate cell cultures of two
(even localized) inflammatory response seem to            isolates, differing for the presence or absence of
be required to prevent the establishment of               this mutation (Bordi et al., 2011). We observed
chronic disease weeks or even months after viral          that the presence of A226V mutation did not in-
clearance from the blood. The absence of such             fluence the replication kinetics on primate cells.
mechanisms leads to chronic arthralgia. In fact,          Moreover, the two isolates displayed very similar
in patients from the La Réunion study, various            time course of cytopathic effect onset, number
markers of inflammation (IFN-α, IL-6, monocyte            and extent of CHIKV antigen-positive cells, as
chemotactic protein-1/CCL-2, IL-8, and matrix             well as the t-shape of the virus-positive multicel-
metalloproteinase 2) were detected in the syn-            lular foci, thus suggesting a similar mechanism of
ovial fluid of a patient suffering from chronic           spread of the virus in the infected cell cultures.
pain, but not in patients who fully recovered             In addition, we considered the possibility that the
(Hoarau et al., 2010). The persistence of a local         A226V mutation could be associated with partial
reservoir of CHIKV in joints may therefore be             resistance to the antiviral activity of recombinant
characteristic of chronic disease, consistently           IFN-α in classical experiments of virus replica-
with findings in the macaque model, in which              tion inhibition. Surprisingly, the A226V-carrying
CHIKV was detected after up to 90 days espe-              strain was more susceptible than the wt virus to
cially in joint tissues, leading to chronic local in-     the antiviral action of IFN-α.
flammation (Labadie et al., 2010). Moreover,              Overall, our result did not support the concept
Hoarau et al. (2010) reported high plasma con-            that A226V mutation confers a replicative ad-
centrations of IL-12 and IFN-α mRNA in blood              vantage in primate cell cultures, nor did it support
mononuclear cells after the convalescent phase            the possibility that partial resistance to the in-
in patients with chronic disease between 6                hibitory action of IFN-α could account for the ex-
months and 1 year after infection. In patients            plosive spread of the mutated strain in the hu-
from Singapore, the concentrations of these two           man population in the countries where this mu-
cytokines, measured by alternative techniques,            tation had occurred. However, the possibility that
peaked in the acute phase and returned to normal          the interplay between the virus and the innate de-
levels at 2-3 months, even in patients who still          fence system may act at different levels of the
had clinical symptoms. According to these find-           virus/host interaction is to be taken into consid-
ings, Chaaithanya and colleagues (2011) and               eration, by exploring, for instance, other steps of
Kelvin and colleagues (2011) reported high lev-           the IFN response activation.
els of Th1-type cytokines in the blood of patients        At the moment, understanding CHIKV immuno-
with chronic disease (Chaaitanya et al., 2011;            biology is still in its infancy and there is a long
Kelvin et al., 2011). Thus, despite certain dis-          way to go before answers related to the interac-
crepancies, the available studies suggest that            tion between virus and host immunity are ob-
chronic disease is associated with a de-regulation        tained. These will certainly be important in de-
of inflammation during the acute and convales-            signing novel antiviral control strategies against
cence phases. This lack of regulation results in a        the spread of CHIKV infection.
deleterious inflammatory process that persists for
≥ 1 year after the first clinical signs (Dupuis-
Maguiraga et al., 2012).                                  DIAGNOSIS
Concerning the possible implication of viral fac-
tors in the pathogenesis, attention has focused           Chikungunya infection is diagnosed on the basis
on the A226V mutation, that has been associated           of clinical, epidemiological and laboratory criteria.
with enhanced replication and fitness of CHIKV            An acute onset of fever and severe arthralgia or
in A. albopictus vector, and has also been shown          arthritis that is not explained by other medical
to modulate the cholesterol requirement for in-           disorders is considered a possible CHIKV case.
222                         C. Caglioti, E. Lalle, C. Castilletti, F. Carletti, M.R. Capobianchi, L. Bordi

The case becomes probable if the patient has lived                   The detection of CHIKV-specific immune re-
in or visited epidemic areas in a time frame con-                    sponse is based on serological methods such as
sistent with the incubation period (WHO                              enzyme-linked assays (ELISA), indirect im-
Guidelines for prevention and control of                             munofluorescence assays (IFA), hemoagglutina-
Chikungunya fever http://www.searo.who.int/                          tion inhibition (HI) and micro-neutralization
LinkFiles/Publication_SEA-CD-182.pdf (accessed                       (MNt).
Aug 01, 2011).                                                       IFA and ELISA are rapid and sensitive techniques
However, laboratory confirmation is crucial, be-                     for detection of CHIKV-specific antibodies, and
cause the case should be distinguished from var-                     can distinguish between IgG and IgM. IgM are
ious disorders with similar clinical manifesta-                      detectable 2-3 days after the onset of symptoms
tions, such as dengue fever, other alphaviruses                      and persist for several weeks, up to 3 months
and arthritic diseases and also endemic malaria.                     (Sam and AbuBakar, 2006; Litzba et et al., 2008).
The interpretation of laboratory findings is de-                     Rarely, IgM can be detected for longer periods, up
pendent on knowledge of the kinetics of viremia                      to 1 year. CHIKV-specific IgG appear soon after
and antibody response in human beings. The de-                       IgM antibodies (2-3 days) and persist for years.
tection of viral nucleic acid or of infectious virus                 Various in-house ELISA techniques using whole
in serum samples is useful during the initial                        antigen or recombinant capsid or envelope anti-
viremic phase, at the onset of symptoms and nor-                     gens have been described (Cho et al., 2008).
mally for the following 5-10 days, when CHIKV                        Commercial serological assays are available and
RNA reaches very high levels (viral loads of                         results obtained from a comparison of the assays
3.3x109 copies/ml) and can be easily detected.                       suggested that the sensitivity for detection of an
Afterwards, the diagnosis is based mainly on the                     early antibody response before day 5 is depend-
detection of specific immune response by sero-                       ent on the strain of the virus used for the assay or
logical methods.                                                     the source of the antigen; assays based on re-
Molecular assays constitutes a rapid and sensi-                      combinant antigens might be too specific with
tive technique for diagnosis of CHIKV infection                      regard to mutations (Cho et al., 2008; Litzba et
during the early stages of illness before an anti-                   al., 2008; Yap et al., 2010).
body response is evident. Conventional RT-PCR                        Testing of a couple of sera collected in the acute
(Hasebe et al., 2002; Pfeffer et al., 2002) are avail-               and convalescent phases of the disease is manda-
able, together with real time loop-mediated RT-                      tory for the identification of recent infection us-
PCR (Parida et al., 2007) and real time TaqMan                       ing serological methods that cannot distinguish
RT-PCR assay targeting the envelope E1 gene                          IgG Ab from IgM Ab (i.e. HI and MNt). It is also
(Pastorino et al., 2005) or the non-structural the                   very useful to confirm results obtained with oth-
nsP1 gene (Carletti et al., 2007). Moreover a one-                   er methods, especially taking into account the
step SYBR green-based real time assay targeting                      possibility of rare persistence of IgM antibodies.
the non-structural nsp2 gene was described more                      Moreover, rapid bedside tests are commercially
recently (Ho et al., 2010).                                          available, but their sensitivity and specificity are
Viral isolation can be performed from serum of                       poorly established, and the possibility of false-
infected patients on insect or mammalian cell                        positive reactions resulting from cross-reactivity
lines (i.e. C6/36 or Vero E6) or by intracerebral in-                with other arthropod-borne alphaviruses has to
oculation of 1-day-old mice during the early                         be considered (Blackburn et al., 1995). In fact,
phase of the disease when the viral load is very                     CHIKV is a member of the SFV antigenic com-
high and the immune response is still not de-                        plex, and is most closely related to ONNV. In this
tectable. In fact, the presence of early antibody                    respect, diagnosis based exclusively on CHIKV-
seems to prevent isolation of the virus, hence                       specific serological testing is useful only for trav-
virus isolation has been shown to be successful                      ellers returning from a geographic area affected
largely in antibody-negative samples obtained on                     by epidemic CHIKV diffusion (Pile et al., 1999),
or before day 2 of illness (Panning et al., 2006).                   while in other cases differential diagnosis is nec-
Moreover, viral isolation is useful for epidemiol-                   essary, taking into account the most common
ogy or pathogenesis studies or for thorough mo-                      viruses circulating in the region where the infec-
lecular characterization (Fusco et al., 2010).                       tion has presumably been acquired.
Chikungunya overview                                            223

REFERENCES                                                   CHARREL R.N., DE LAMBALLERIE X., RAOULT D. (2007).
                                                                Chikungunya outbreaks - the globalization of vec-
AKAHATA W., YANG Z.Y., ANDERSEN H., SUN S.,                     tor-borne diseases. N. Engl. J. Med. 356, 769-771.
   HOLDAWAY H.A., KONG W.P, LEWIS M.G., HIGGS S.,            CHARREL R., DE LAMBALLERIE X. (2008). Chikungunya in
   ROSSMANN M.G., SRINIVAS R., NABEL G.J. (2010). A             north-eastern Italy: a consequence of seasonal syn-
   VLP vaccine for epidemic Chikungunya virus pro-              chronicity. Euro Surveill. 13, pii: 8003.
   tects nonhuman primates against infection. Nat.           CHATTERJEE S.N., CHAKRAVARTI S.K., MITRA A.C., SARKAR
   Med. 16, 334-338.                                            J.K. (1965a). Virological investigation of cases with
ARANKALLE V.A., SHRIVASTAVA S., CHERIAN S., GUNJIKAR            neurological complications during the outbreak of
   R.S., WALIMBE A.M., JADHAV S.M., SUDEEP A.B.,                haemorrhagic fever in Calcutta. J. Indian Med.
   MISHRA A.C. (2007). Genetic divergence of                    Assoc. 45, 314-316.
   Chikungunya viruses in India (1963-2006) with spe-        CHATTERJEE S.N., SARKAR J.K. (1965b). Electron mi-
   cial reference to the 2005-2006 explosive epidemic.          croscopic studies of suckling mouse brain cells in-
   J. Gen. Virol. 88, 1967-1976.                                fected with Chikungunya virus. Indian J. Exp. Biol.
BEESOON S., FUNKHOUSER E., KOTEA N., SPIELMAN A.,               3, 227-234.
   ROBICH R.M. (2008). Chikungunya fever, Mauritius,         CHO B., KIM J., CHO J.E., JEON B.Y., PARK S. (2008).
   2006. Emerg. Infect. Dis. 14, 337-338.                       Expression of the capsid protein of Chikungunya
BLACKBURN N.K., BESSELAAR T.G., GIBSON G. (1995).               virus in a baculovirus for serodiagnosis of
   Antigenic relationship between chikungunya virus             Chikungunya disease. J. Virol. Methods. 154, 154-
   strains and o’nyong nyong virus using monoclonal             159.
   antibodies. Res. Virol. 146, 69-73.                       CHOW A., HER Z., ONG E.K., CHEN J.M., DIMATATAC F.,
BORDI L., CARLETTI F., CASTILLETTI C., CHIAPPINI R.,            KWEK D.J., BARKHAM T., YANG H., RéNIA L., LEO
   SAMBRI V., CAVRINI F., IPPOLITO G., DI CARO A.,              Y.S., NG L.F. (2011). Persistent arthralgia induced
   CAPOBIANCHI M.R. (2008). Presence of the A226V               by Chikungunya virus infection is associated with
   mutation in autochthonous and imported Italian               interleukin-6 and granulocyte macrophage colony-
   chikungunya virus strains. Clin. Infect. Dis. 47,            stimulating factor. J. Infect. Dis. 203, 149-57.
   428-429.                                                  COUDERC T., CHRéTIEN F., SCHILTE C., DISSON O.,
BORDI L., MESCHI S., SELLERI M., LALLE E., CASTILLETTI          BRIGITTE M., GUIVEL-BENHASSINE F., TOURET Y.,
   C., CARLETTI F., CHIAPPINI R., DI CARO A.,                   BARAU G., CAYET N., SCHUFFENECKER I., DESPRèS P.,
   CAPOBIANCHI M.R. (2011). Chikungunya virus iso-              ARENZANA-SEISDEDOS F., MICHAULT A., ALBERT M.L.,
   lates with/without A226V mutation show differ-               LECUIT M. (2008). A mouse model for Chikungunya:
   ent sensitivity to IFN-α, but similar replication ki-        young age and inefficient Type-I interferon sig-
   netics in non human primate cells. New Microbiol.            nalling are risk factors for severe disease. Plos
   34, 87-91.                                                   Pathogens. 4, e29.
BRIGHTON S.W., PROZESKY O.W., DE LA HARPE A.L.               COUDERC T., KHANDOUDI N., GRANDADAM M., VISSE C.,
   (1983). Chikungunya virus infection. A retrospec-            GANGNEUX N., BAGOT S., PROST J.F., LECUIT M.
   tive study of 107 cases. S. Afr. Med. J. 63, 313-315.        (2009). Prophylaxis and therapy for Chikungunya
BRIGHTON S.W. (1984). Chloroquine phosphate treat-              virus infection. J. Infect. Dis. 200, 516-523.
   ment of chronic Chikungunya arthritis: an open pi-        CUI F., RAYMOND M., QIAO C.L. (2006). Insecticide re-
   lot study. S. Afr. Med. J. 66, 217-218.                      sistance in vector mosquitoes in China. Pest. Manag.
BRIOLANT S., GARIN D., SCARAMOZZINO N., JOUAN A.,               Sci. 62, 1013-1022.
   CRANCE J.M. (2004). In vitro inhibition of                D’ORTENZIO E., GRANDADAM M., BALLEYDIER E., JAFFAR-
   Chikungunya and Semliki Forest viruses replica-              BANDJEE M.C., MICHAULT A., BROTTET E., BAVILLE
   tion by antiviral compounds: synergistic effect of           M., FILLEUL L. (2011). A226V strains of
   interferon-alpha and ribavirin combination.                  Chikungunya virus, Réunion Island, 2010. Emerg.
   Antiviral. Res. 61, 111-117.                                 Infect. Dis. 17, 309-311.
CARLETTI F., BORDI L., CHIAPPINI R., IPPOLITO G.,            DE LAMBELLERIE X., LEROY E., CHARREL R.N.,
   SCIARRONE M.R., CAPOBIANCHI M.R., DI CARO A.,                TTSETSARKIN K., HIGGS S., GOULD E.A. (2008a).
   CASTILLETTI C. (2007). Rapid detection and quan-             Chikungunya virus adapts to tiger mosquito via
   tification of Chikungunya virus by a one-step re-            evolutionary convergence: a sign of things to come?
   verse transcription polymerase chain reaction re-            Virol. J. 5, 33.
   al-time assay. Am. J. Trop. Med. Hyg. 77, 521-524.        DE LAMBALLERIE X., BOISSON V., REYNIER J.C., ENAULT S.,
CHAAITANYA I.K., MURUGANANDAM N., SUNDARAM S.G.,                CHARREL R.N., FLAHAULT A., ROQUES P., LE GRAND R.
   KAWALEKAR O., SUGUNAN A.P., MANIMUNDA S.P.,                  (2008b). On chikungunya acute infection and
   GHOSAL S.R., MUTHUMANI K., VIJAYACHARI P. (2011).            chloroquine treatment. Vector Borne Zoonotic. Dis.
   Role of proinflammatory cytokines and chemokines             8, 837-839.
   in chronic arthropathy in CHIKV infection. Viral.         DESSAIN S.K., ADEKAR S.P., BERRY J.D. (2008). Exploring
   Immunol. 24, 265-271.                                        the native human antibody repertoire to create an-
224                           C. Caglioti, E. Lalle, C. Castilletti, F. Carletti, M.R. Capobianchi, L. Bordi

   tiviral therapeutics. Curr. Top. Microbiol. Immunol.                    DAS T., LI-PAT-YUEN G., DASSA B., DENIZOT M.,
   317, 155-183.                                                           GUICHARD E., RIBERA A., HENNI T., TALLET F.,
DUPUIS-MAGUIRAGA L., NORET M., BRUN S., LE GRAND                           MOITON M.P., GAUZèRE B.A., BRUNIQUET S., JAFFAR
   R., GRAS G., ROQUES P. (2012). Chikungunya dis-                         BANDJEE Z., MORBIDELLI P., MARTIGNY G., JOLIVET
   ease: infection-associated markers from the acute                       M., GAY F., GRANDADAM M., TOLOU H., VIEILLARD V.,
   to the chronic phase of arbovirus-induced arthral-                      DEBRé P., AUTRAN B., GASQUE P. (2010). Persistent
   gia. PLoS Negl. Trop. Dis. 6, e1446.                                    chronic inflammation and infection by
ECKELS K.H., HARRISON V.R., HETRICK F.M. (1970).                           Chikungunya arthritogenic alphavirus in spite of a
   Chikungunya virus vaccine prepared by Tween-                            robust host immune response. J. Immunol. 184,
   ether extraction. Appl. Microbiol. 19, 321-325.                         5914-5927.
EDELMAN R., TACKET C.O., WASSERMAN S.S, BODISON                        HOCHEDEZ P., JAUREGUIBERRY S., DEBRUYNE M., BOSSI
   S.A., PERRY J.G., MANGIAFICO J.A. (2000). Phase II                      P., HAUSFATER P., BRUCKER G., BRICAIRE F., CAUMES
   safety and immunogenicity study of live                                 E. (2006). Chikungunya infection in travelers.
   Chikungunya virus vaccine TSI-GSD-218. Am. J.                           Emerg. Infect. Dis. 12, 1565-1567.
   Trop. Med. Hyg. 62, 681-685.                                        INOUE S., MORITA K., MATIAS R.R., TUPLANO J.V.,
ENSERINK M. (2006). Infectious diseases. Massive out-                      RESUELLO R.R., CANDELARIO J.R., CRUZ D.J., MAPUA
   break draws fresh attention to little-known virus.                      C.A., HASEBE F., IGARASHI A., NATIVIDAD F.F. (2003).
   Science. 311, 1085.                                                     Distribution of three arbovirus antibodies among
FOURIE E.D., MORRISON J.G. (1979). Rheumatoid                              monkeys (Macaca fascicularis) in the Philippines.
   arthritic syndrome after Chikungunya fever. S. Afr.                     J. Med. Primatol. 32, 89-94.
   Med. J. 56, 130-132.                                                JACUPS S.P., WHELAN P.I., CURRIE B.J. (2008). Ross River
FUSCO F.M., PURO V., DI CARO A., NICASTRI E., CARANNANTE                   virus and Barmah Forest virus infections: a review
   N., FAELLA F.S., BARZON L., DI CESARE S., PALù G.,                      of history, ecology, and predictive models, with im-
   CAPOBIANCHI M.R., IPPOLITO G. (2006). Cases of                          plications for tropical northern Australia. Vector
   Chikungunya fever in Italy in travellers returning                      Borne Zoonotic. Dis. 8, 283-297.
   from the Indian Ocean and risk of introduction of                   JAIN M., RAI S., CHAKRAVARTI A. (2008). Chikungunya:
   the disease to Italy. Infez. Med. 14, 238-245.                          a review. Trop. Doc. 38, 70-72.
FUSCO F.M., NICASTRI E., NISII C., DI CARO A, IPPOLITO G.              JOSSERAN L., PAQUET C., ZEHGNOUN A., CAILLERE N., LE
   (2010). Chikungunya fever, a re-emerging disease.                       TERTRE A., SOLET J.L., LEDRANS M. (2006).
   Tropical and Emerging Infectious Diseases, Maltezou                     Chikungunya disease outbreak, Réunion Island.
   H.C. and Gikas A., 93-110, ISBN: 978-81-308-0389-0.                     Emerg. Infect. Dis. 12, 1994-1995.
HARRISON V.R., BINN L.N., RANDALL R. (1967).                           KAM Y.W., ONG E.K., RéNIA L., TONG J.C., NG L.F.
   Comparative immunogenicities of Chikungunya                             (2009). Immuno-biology of Chikungunya and im-
   vaccines prepared in avian and mammalian tissues.                       plications for disease intervention. Microbes Infect.
   Am. J. Trop. Med. Hyg. 16, 786-791.                                     11, 1186-1196.
HARRISON V.R, ECKELS K.H., BARTELLONI P.J. HAMPTON                     KELVIN A.A., BANNER D., SILVI G., MORO M.L., SPATARO
   C. (1971). Production and evaluation of a forma-                        N., GAIBANI P., CAVRINI F., PIERRO A., ROSSINI G.,
   lin-killed Chikungunya vaccine. J. Immunol. 107,                        CAMERON M.J., BERMEJO-MARTIN J.F., PAQUETTE S.G.,
   643-647.                                                                XU L., DANESH A., FAROOQUI A., BORGHETTO I.,
HASEBE F., PARQUET M.C., PANDEY B.D., MATHENGE E.G.,                       KELVIN D.J., SAMBRI V., RUBINO S. (2011).
   MORITA K., BALASUBRAMANIAM V., SAAT Z., YUSOP A.,                       Inflammatory cytokine expression is associated
   SINNIAH M., NATKUNAM S., IGARASHI A. (2002).                            with Chikungunya virus resolution and symptom
   Combined detection and genotyping of Chikungunya                        severity. PLoS Negl. Trop. Dis. 5, e1279.
   virus by a specific reverse transcription-polymerase                KENNEDY A.C., FLEMING J., SOLOMON L. (1980).
   chain reaction. J. Med. Virol. 67, 370-4.                               Chikungunya viral arthropathy: a clinical descrip-
HEARN H.J. JR., RAINEY C.T. (1963). Cross-protection                       tion. J. Rheumatol. 7, 231-236.
   in animals infected with Group A arboviruses. J.                    KENNEY J.L., VOLK S.M., PANDYA J., WANG E., LIANG X.,
   Immunol. 90, 720-724.                                                   WEAVER S.C. (2011). Stability of RNA virus attenu-
HER Z., MALLERET B., CHAN M., ONG E.K., WONG S.C.,                         ation approaches. Vaccine. 29, 2230-2234.
   KWEK D.J., TOLOU H., LIN R.T., TAMBYAH P.A., RéNIA                  KHAN A.H., MORITA K., PARQUET MD MDEL C., HASEBE
   L., NG L.F. (2010). Active infection of human blood                     F., MATHENGE E.G., IGARASHI A. (2002). Complete
   monocytes by Chikungunya virus triggers an innate                       nucleotide sequence of Chikungunya virus and ev-
   immune response. J. Immunol. 184, 5903-5913.                            idence for an internal polyadenylation site. Gen.
HO P.S., NG M.M., CHU J.J. (2010). Establishment of                        Virol. 83, 3075-3084.
   one-step SYBR green-based real time-PCR assay                       KNUDSEN A.B. (1995). Global distribution and contin-
   for rapid detection and quantification of                               uing spread of Aedes albopictus. Parassitologia.37,
   Chikungunya virus infection. Virol. J. 7, 13.                           91-97.
HOARAU J.J., JAFFAR BANDJEE M.C., KREJBICH TROTOT P.,                  KUMAR M., SUDEEP A.B., ARANKALLE V.A. (2012).
You can also read