Você está na página 1de 11

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/286982506

Chikungunya virus: recent advances in


epidemiology, host pathogen interaction &
vaccine strategies

Article in Pathogens and Disease December 2015


DOI: 10.1093/femspd/ftv119

CITATIONS READS

6 209

7 authors, including:

Asimul Islam Syed Naqui Kazim


Jamia Millia Islamia Jamia Millia Islamia
107 PUBLICATIONS 654 CITATIONS 52 PUBLICATIONS 843 CITATIONS

SEE PROFILE SEE PROFILE

Shobha Broor Shama Parveen


SGT Medical College, Hospital & Research Ins Jamia Millia Islamia
346 PUBLICATIONS 3,739 CITATIONS 96 PUBLICATIONS 511 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Comparative Analysis of Selected Routing Protocols for WLAN Based Wireless Sensor Networks
(WSNs) View project

Direct and Indirect Protection by Influenza Vaccine given to Children in India View project

All content following this page was uploaded by Shama Parveen on 04 February 2016.

The user has requested enhancement of the downloaded file. All in-text references underlined in blue are added to the original document
and are linked to publications on ResearchGate, letting you access and read them immediately.
Pathogens and Disease, 74, 2016, ftv119

doi: 10.1093/femspd/ftv119
Advance Access Publication Date: 10 December 2015
Minireview

MINIREVIEW

Chikungunya virus: recent advances in epidemiology,


host pathogen interaction and vaccine strategies
Farah Deeba1 , Asimul Islam1 , Syed Naqui Kazim1 , Irshad Hussain Naqvi2 ,

Downloaded from http://femspd.oxfordjournals.org/ by Shama Parveen on February 4, 2016


Shobha Broor3 , Anwar Ahmed4 and Shama Parveen1,
1
Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India,
2
Dr. M. A. Ansari Health Centre, Jamia Millia Islamia, New Delhi 110025, India, 3 Department of Microbiology,
SGT University, Gurgaon 122001, Haryana, India and 4 Protein Research Chair, Department of Biochemistry,
King Saud University, Riyadh 11451, Saudi Arabia

Corresponding author: Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India. Tel: +91 011 26984167 ext.
4492; E-mail: shamp25@yahoo.com
One sentence summary: This review provides a comprehensive outline of the Chikungunya viral infection with emphasis on the epidemiology, host
pathogen interaction and vaccine stratagies.
Editor: Alfredo Garzino-Demo

ABSTRACT
The Chikungunya virus is a re-emerging alphavirus that belongs to the family Togaviridae. The symptoms include fever,
rashes, nausea and joint pain that may last for months. The laboratory diagnosis of the infection is based on the serologic
assays, virus isolation and molecular methods. The pathogenesis of the Chikungunya viral infection is not completely
understood. Some of the recent investigations have provided information on replication of the virus in various cells and
organs. In addition, some recent reports have indicated that the severity of the disease is correlated with the viral load and
cytokines. The Chikungunya virus infection re-emerged as an explosive epidemic during 200409 affecting millions of
people in the Indian Ocean. Subsequent global attention was given to research on this viral pathogen due to its broad area
of geographical distribution during this epidemic. Chikungunya viral infection has become a challenge for the public health
system because of the absence of a vaccine as well as antiviral drugs. A number of potential vaccine candidates have been
tested on humans and animal models during clinical and preclinical trials. In this review, we mainly discuss the
hostpathogen relationship, epidemiology and recent advances in the development of drugs and vaccines for the
Chikungunya viral infection.

Keywords: Chikungunya virus; disease dissemination; hostpathogen interactions

INTRODUCTION infection is underestimated because of its overlapping symp-


toms with dengue fever.
The name Chikungunya was derived from a Makonde word that
The Chikungunya virus has caused many outbreaks
means one which bends up; it refers to the posture of the pa-
worldwide in the last decade, including the Indian Ocean
tient due to extreme joint pain. The symptoms of Chikungunya
region, African countries and American continent. During
virus (CHIKV) infection are high fever, rashes, nausea, arthral-
the 200409 epidemic the CHIKV was found to infect another
gia, myalgia etc. (Schuffenecker et al. 2006). The Ades aegepti
species of mosquito, i.e. Aedes albopictus due to a single point
mosquito is the main vector involved in the transmission of
mutation in the genome (Schuffenecker et al. 2006). This severe
the disease. However, the actual burden of Chikungunya viral

Received: 11 September 2015; Accepted: 5 December 2015



C FEMS 2015. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com

1
2 Pathogens and Disease, 2016, Vol. 74, No. 3

Downloaded from http://femspd.oxfordjournals.org/ by Shama Parveen on February 4, 2016


Figure 1. Chikungunya virus replication cycle. The CHIKV enters into the host cell through receptor-mediated endocytosis forming an endosome. The endosome enters
the host cell and genome of the virus then replicates in the cytoplasm. The CHIKV genome carries non-translated region (NTR) at both 3 and 5 ends and non-coding
junction region (J region) between the genes coding for structural and non-structural proteins. The replicated negative strand of RNA is transcribed into positive sense
subgenomic RNA, also known as 26S RNA, that serves as mRNA for structural proteins. The genomic and subgenomic RNAs are translated into polyprotein precursors
that finally cleave into non-structural proteins (nsp1, nsp2, nsp3 and nsp4) and structural proteins (capsid, E1 and E2). The positive RNA strand, structural proteins
and capsid proteins finally assemble into the mature virion. ER, endoplasmic reticulum.

outbreak occurred in 2004 in the Indian Ocean region including teins (nsp1, nsp2, nsp3 and nsp4) that are involved in the repli-
India. CHIKV infection is affecting the socioeconomic status of cation of the virus. The nsp1 protein functions as a cytoplasmic
various geographical regions because of the absence of an ef- RNA capping enzyme, whereas the nsp2 protein has two sepa-
fective vaccine and antiviral drugs. Chikungunya viral infection rate domains with helicase and protease activity. The nsp3 pro-
is a major health concern in India also due to the presence of tein is essential for negative strand RNA synthesis. The nsp4
a large immunologically naive population. Therefore, regular protein works as an RNA-dependent RNA polymerase (Solig-
surveillance for the Chikungunya viral infection should be done nat et al. 2009). One-third of the genome from the 3 end has
in the endemic regions such as India. This will assist in the a polyadenylate tail that consists of 26S mRNA whose prod-
prediction and control of outbreaks. In addition, the generation ucts include the structural proteins. The structural proteins of
of awareness among the general public about the disease is CHIKV are the surface glycoproteins (E1 and E2), capsid pro-
essential for its effective control and management. tein and small peptides (E3 and 6K) (Weaver et al. 2005; Pow-
ers and Logue 2007; Abere et al. 2012). The viral surface is cov-
ered with membrane-anchored spikes which are triplets of a
GENOME
heterodimer of envelope E1 and E2 glycoproteins. The E2 is ex-
CHIKV is a member of the genus Alphavirus, in the family To- posed and has a role in viral attachment, whereas E1 is the
gaviridae (Powers et al. 2000). The CHIKV is a small (6070 nm in fusion protein that facilitates the viral entry. The epitopes in
diameter) and enveloped virus. The 11.8 kb genome of CHIKV is a the E2 glycoprotein are recognized by neutralizing antibodies
linear, positive sense RNA molecule. Figure 1 shows a schematic and therefore E2 is the vaccine candidate (Brehin et al. 2008).
representation of the genome of CHIKV and its replication in The lineages of CHIKV are reported with different genotypic
the host cell. Two-thirds of the genome from the 5 end has a characteristics. The genotypes are classified on the basis of
methyl guanylate cap that consists of a non-structural polypro- E1 gene sequence and the full genome of CHIKV. Three geno-
tein precursor. This precursor is cleaved into non-structural pro- types (East, Central and South African (ECSA), West African and
Deeba et al. 3

Asian) have been described for the Chikungunya virus (Santhosh and detection of antibodies (IgM/IgG) to the virus. The CHIKV
et al. 2008). isolation is performed on serum samples collected within the
first 7 days of illness. The virus can be isolated in insect and
mammalian cell lines (C636, Vero). The cultured virus can be
VECTOR AND DISEASE TRANSMISSION confirmed by RT-PCR or immunofluorescence assay. Virus iso-
CHIKV is an arbovirus that spreads mainly through Aedes ae- lation is time consuming and needs expertise therefore it is no
gypti and Aedes albopictus mosquitoes. The CHIKV life cycle in- longer being used for the detection of CHIKV infection. The con-
volves: (i) an enzootic sylvatic cycle, and (ii) an epidemic ur- ventional PCR and real-time PCR assays have been developed
ban cycle. The sylvatic cycle is maintained primarily in African targeting the envelope and the non-structural genes of CHIKV.
forests involving several arboreal Aedes mosquito species as RT-PCR can be used for early identification of the CHIKV in-
vectors and non-human primates like monkeys, rodents, birds, fection before detection of the antibodies, i.e. within the first
etc. as reservoirs (Apandi et al. 2009; Caglioti et al. 2013). It is 710 days of infection. DNA sequencing coupled with the RT-PCR
believed that initially in Africa, the enzootic transmission cy- technique can be used to identify the corresponding genotype.
cle spilled over and infected nearby human populations (Thon- Real-time PCR is used to determine the viral load in the clini-
non et al. 1999; Peyrefitte et al. 2007). The mosquitoes are also cal samples. The sensitivity of real-time RT-PCR was found to be
involved in inter-human transmission during outbreaks. Dur- around 10-fold higher than conventional RT-PCR and as low as
ing epidemics, human beings serve as reservoirs in the urban 20 copies of RNA transcript can be identified by real-time RT-PCR
cycle. (Edwards et al. 2007).

Downloaded from http://femspd.oxfordjournals.org/ by Shama Parveen on February 4, 2016


The diagnosis of the CHIKV infection can also done by de-
tection of the immune response by serological assays. The im-
CLINICAL MANIFESTATIONS munoassays for IgM/IgG (ELISA) and indirect immunofluores-
cent assay (IFA) are used for detection of antibodies to the virus.
The alphaviruses are clinically divided into two subgroups: en-
The indirect immunofluorescent assay is also a method for the
cephalogenic (e.g. Venezuelan equine encephalitis virus, West-
detection of Chikungunya virus in the clinical samples (Yer-
ern equine encephalitis virus), which infect neurons, and
golkar et al. 2006). The IgM antibodies develop within 23 days
arthritogenic (e.g. Chikungunya virus, Sindbis virus) which are
after the onset of symptoms of infection and may persist for sev-
basically associated with polyarthritis and rashes. The infection
eral months (Sam and AbuBakar 2006; Malvy et al. 2009). The IgG
with CHIKV is followed by a silent period of incubation that lasts
antibodies develop in the convalescent phase and may persist
24 days. The incubation period is followed by an abrupt onset
for several months to years (Cavrini et al. 2009). The sensitivity
of symptoms, including fever, headache, nausea, myalgia and
and specificity of serological assays are poorly established be-
rashes (Pialoux et al. 2007). The most prominent and typical clin-
cause of the possibility of false positive results due to cross re-
ical feature is the polyarthralgia that can sometimes persist for
activity with antibodies to dengue or other arboviruses (Pialoux
as long as a few months to years (Schilte et al. 2013). Chikun-
et al. 2007). Therefore, limitations with detection of the Chikun-
gunya viral infection is commonly not found to be associated
gunya viral infection by IgM capture ELISA exist. Recently a study
with mortality. The severity of disease in the case of CHIKV
has tried to improve the ELISA by developing antigen for the
infection is directly attributed to the severe and prolonged
assay using Eilat virus instead of the Chikungunya virus (Eras-
symptoms or effects. In some cases lymphopenia, thrombo-
mus et al. 2015). The antigen that was developed does not re-
cytopenia and minor hemorrhagic signs such as epistaxis or
quire biosafety containment facilities and can be used for ELISA
gingivorrhagia have also been reported (Schuffenecker et al.
without inactivation. However, this assay needs to be tested and
2006). During the 2006 outbreak in Reunion Island neurological
evaluated before its implementation on clinical samples.
complications such as meningoencephilitis were also reported
in a small proportion of patients (Quatresous 2006). During this
epidemic optic neuritis, anterior uveitis and dendritic lesions oc-
curred as primary acute onset inflammatory reactions (Lalitha HOSTPATHOGEN INTERACTIONS
et al. 2007; Mittal et al. 2007). Certain follow-up studies of pa-
The Chikungunya virus first replicates in the skin fibroblasts af-
tients with Chikungunya viral infection have shown evidence
ter inoculation into the skin of humans through the mosquito
of cardiac complications such as myocarditis (Obeyesekere and
bite. Only fibroblasts of the skin have been shown to be sus-
Hermon 1973).
ceptible to CHIKV infection amongst various types of cells
Chikungunya viral infection is generally a self-limiting febrile
(Talarmin et al. 2007; Robin et al. 2009). Once replicated in the
illness. The resolution of CHIKV infection takes place within the
skin, it further disseminates through blood to liver, muscle tis-
first 15 days of illness after which the disease enters a stage
sues, joints, lymph nodes and brain (Fig. 2). On reaching muscles,
where chronic rheumatism remains as the major feature (Man-
the Chikungunya virus replicates in muscle satellite and fibrob-
imunda et al. 2010). The symptoms often mimic those of dengue
last cells and not in muscle fibres (Ozden et al. 2007). The CHIKV
fever and its spread is also common in dengue endemic regions,
replicates in joint fibroblasts and the synovial tissues (Hoarau,
therefore the two diseases are commonly indistinguishable (Ravi
Jaffar Bandjee and Krejbich Trotot 2010). In addition, the Chikun-
2006).
gunya virus RNA and proteins have been identified in the syn-
ovial tissues during severe and persistent arthralgia (Parola et al.
DETECTION OF CHIKUNGUNYA 2006; Manimunda et al. 2007; Ozden et al. 2007; Hoarau, Jaffar
Bandjee and Krejbich Trotot 2010).
VIRUS INFECTION
When the virus disseminates to brain it seems to infect the
Diagnosis of Chikungunya viral infection is based upon the clini- lining of the choroid plexus of stromal cells of the nervous sys-
cal symptoms and laboratory diagnosis. Routinely used diagnos- tem. The CHIKV tissue tropism studied in a mouse model was
tic methods include isolation of virus in cell culture, identifica- found to match with the tissue tropism reported in the in vitro
tion of viral RNA by reverse transcriptase (RT)-PCR/real-time PCR system. The onset of symptoms coincides with a rise in viral
4 Pathogens and Disease, 2016, Vol. 74, No. 3

Downloaded from http://femspd.oxfordjournals.org/ by Shama Parveen on February 4, 2016


Figure 2. A schematic representation of the systemic Chikungunya virus dissemination. The Chikungunya virus enters the host cell by the mosquito bite. The virus
then replicates into fibroblasts of skin and enters nearby lymph nodes through which it disseminates into the blood. The Chikungunya virus further migrates to
different organs and tissues through blood vessels.

CXCL10, CCL2 and IL6 in the acute phase indicates inflammation


initiating the adaptive T cell immune response. The CXCL9 and
CXCL10 are also part of the chemokine program regulating the
migration of monocytes/macrophages, natural killer cells and
memory T cells (Deshmane et al. 2009). The chemokines are cru-
cial mediators of innate as well as adaptive immune responses
against various viral infections (Rot and von Andrian 2004). Both
CXCL9 and CXCL10 are induced by INF and work as a chemo-
attractant for T cells. Both these chemokines signal through a
common receptor, i.e. CXCR3. Therefore, they may have a role
in activation of T cells also, as CXCR3 identifies the T cell sub-
Figure 3. A graphical representation showing levels of various cytokines and sets (Qin et al. 1998). The CCL2 is also referred to as monocyte
chemokines after CHIKV infection. The graph shows an increase and decrease chemotactic protein 1 (MCP-1) that leads monocytes, memory T
in levels of different cytokines with respect to the onset of symptoms and its cells and dendritic cells to the sites of inflammation.
resolution.
IL6 is an immune mediator of fever that activates muscle
metabolism so as to increase body temperature (Cronstein 2007).
titer. Thus activation of the innate immune response occurs, The levels of granulocyte macrophage colony-stimulating fac-
which results in the production of type I interferons as the pri- tor (GM-CSF) receptor and IL6 levels in plasma were significantly
mary response. The type I interferons are mainly involved in higher in patients with persistent arthralgia (Chow et al. 2011).
the innate immune responses against viral infections. These IL6 is a pleotropic cytokine and has a destructive role in rheuma-
interferons are known to induce the expression of interferon- toid arthritis due to which joint inflammation and pain occur
stimulated genes (ISGs) having antiviral, antiproliferative and (Cronstein 2007; Schaible et al. 2010). The increased levels of IL6
immune-modulatory functions (Kumar et al. 2012). The immune may also contribute to the severe joint pain in the CHIKV infec-
response elicited by CHIKV involves the production of many cy- tion. The IL6 is also a major B cell growth factor (Hirano et al.
tokines, chemokines and growth factors. The levels of many cy- 1985) and a known inducer of IgG3 (Kawano, Noma and Yata
tokines (INF , CXCL9, CXCL10, CCL2 (MCP-1), IL6 and INF ) were 1994). The high IgG3 titer is induced by high viremia and is also
found to be higher in the acute phase and their level decreased correlated with the higher IL6 level. An early increase of IgG3
with the convalescent phase (Fig. 3) (Rulli et al. 2011; Wauquier levels was found to be associated with efficient viral clearance in
et al. 2011). Interferon (INF ) is detectable even on the first vivo (Kam et al. 2012). The higher level of C-reactive protein (CRP)
day of infection and its level correlates with the plasma viral is activated by higher IL6 levels. Higher CRP levels, in turn, ac-
load (Dupuis-Maguiraga et al. 2012). The high level of CXCL9, tivate macrophage-mediated phagocytosis, lymphopenia (very
Deeba et al. 5

low levels of lymphocytes), lower monocyte level, neutrophilia were documented in 19992000 in Gabon affecting around
and high viral load (Chow et al. 2011). Eighty percent of the indi- 50 000 people (Pastorino et al. 2004). A subsequent epidemic was
viduals infected with CHIKV were found to have lymphopenia. reported from Indonesia in 200103 (Laras et al. 2005).
These patients experienced a significantly lower level of circu- The most recent re-emerging epidemic was reported from
lating B and T lymphocytes (Borgherini et al. 2007). Such low lev- the Indian Ocean region, including India, from 2004 to 2009 that
els of lymphocytes cannot be directly due to the infection, but affected millions of people in various countries. This epidemic
it is the higher levels of type I INFs that induce cell death in in the Indian Ocean region was believed to have originated from
lymphocytes (Schwartz and Albert 2010). The elevated level of Kenya. Strains of this outbreak formed a separate clade in the
RANTES (Regulated on activation, normal T cell expressed and ECSA genotype that is known as Indian Ocean Lineage (IOL) (Yer-
secreted) is observed during many viral infections. Likewise, the golkar et al. 2006; Volk et al. 2010; Weaver and Forrester 2015).
elevated level of RANTES is also reported during dengue viral in- Thus, this epidemic provided a genotypic shift of the CHIKV
fection. An investigation by Ng and colleagues observed that the from Asian to ECSA genotype in the Indian Ocean region. This
level of RANTES decreased significantly during the Chikungunya ECSA genotype has been reported in the subsequent epidemio-
virus infection. Therefore, a decrease in the level of RANTES can logical studies from this region (Vashishtha et al. 1998; Arankalle
be considered a biomarker to differentiate between the Chikun- et al. 2007). A unique molecular signature of this outbreak was
gunya and dengue infections since both these viral infections the E1 A226V mutation that was absent in the initial phase of
have overlapping symptoms (Kelvin et al. 2011). the epidemic, but later on was found in almost 90% of the iso-
The onset of the adaptive immune response through activa- lates (Schuffenecker et al. 2006). Due to this mutation the virus

Downloaded from http://femspd.oxfordjournals.org/ by Shama Parveen on February 4, 2016


tion and proliferation of CD8+ T cells occurs in the early stage was able to infect A. albopictus, which was not the usual host of
of infection. The percentages of CD3+ and CD8+ lymphocytes CHIKV until this epidemic. The mutation A226V in the E1 pro-
were found to be significantly higher in patients infected with tein was shown to provide cholesterol independence to a Sem-
CHIKV on days 0, 1 and 2 in comparison with healthy controls liki Forest virus population (Tsetsarkin et al. 2007). The ability
(Wauquier et al. 2011). The level of cytokines such as TNF-, IL- of a virus to infect insect cells by providing a better adaptation
1, IL2, IL5 and IL12 were found to increase with patient recov- to the lipid composition of these cells could be improved by the
ery (Fig. 3) (Ng et al. 2009; Chow et al. 2011). TNF and IL-1 play mutation that affects cholesterol dependency. The experimen-
a major role in inflammatory diseases and contribute mainly to tal demonstrations showed that viruses without the E1 A226V
the joint pain in diseases like rheumatoid arthritis. However, in mutation were not as successful at replicating in A. albopictus
Chikungunya viral infection the levels of TNF and IL-1 were (Vazeille et al. 2007). On the other hand, the mutated ones had a
found to increase in the convalescent phase and did not signifi- higher efficiency of replication and dissemination in A. albopic-
cantly differ in patients reporting mild or severe conditions (Ng tus (Parola et al. 2006). Therefore, this mutation was attributed to
et al. 2009). This indicates no or a mild contribution of TNF and the higher epidemic potential of the virus during this epidemic.
IL-1 in persistent arthralgia in CHIKV patients. Since A. albopictus has a long life span and is active all day long,
the spread of CHIKV infection became rapid and reached many
more geographical regions than expected. The overall geograph-
ical distribution of the Aedes mosquitoes has changed since this
EPIDEMIOLOGY
outbreak. The Aedes albopictus is now spreading to regions that
The CHIKV is believed to have originated in Africa and then were earlier inhabited by the Aedes aegypti. Consequently, the
spread to all over the world, causing many epidemics and af- circulation patterns of the Chikungunya virus, as well as the dy-
fecting millions of people (Powers et al. 2000). The geograph- namics of the epidemics caused by this virus, are expected to be
ical distribution of CHIKV commonly includes tropical coun- modified in the future.
tries, but the viral infection also extended into the temper- The first outbreak of Chikungunya virus infection in India
ate regions after the 2004 epidemic (Robinson 1955). It was took place in 196364 in Kolkata (Shah, Gibbs and Banerjee 1964).
first reported from East Africa around Tanzania during an epi- Subsequent epidemics were reported from many parts of India,
demic of fever in 195253. This fever was later described in including Chennai, Pondicherry, Vellore, Visakhapatnam, Raj-
1955 as a dengue like disease (Lumsden 1955; Sergon et al. mundry, Kakinada and Nagpur in 196465 (Rao 1966; Rodrigues
2008). After the first report, the Chikungunya virus infection et al. 1972) and Barsi in 1973 (Padbidri and Gnaneswar 1979)
was documented worldwide involving many countries including affecting millions of people. The virus was postulated to have
Tanzania, Ghana, Mozambique, Burkina Faso, Comoros, Kenya, disappeared from India and South East Asia after 1973 (Burke,
Central African Republic, Benin, Uganda, Nigeria, Zimbabwe, Nisalak and Nimmannitya 1985; Neogi et al. 1995; Bhatia and
Senegal, South Africa, Cameroon, Congo, Guinea, Mayotte, Bu- Narain 2009), but during 2004 it suddenly re-emerged as an ex-
rundi, Sudan, Malawi, Namibia, Gabon, Sri Lanka, Myanmar, plosive epidemic in India affecting around 1.3 million people
Thailand, Vietnam, Indonesia, Malaysia, Taiwan, Laos, Philip- in 13 different states after a gap of almost 32 years (Wauquier
pines, Pakistan, Singapore, Cambodia, India, China, Sri Lanka, et al. 2011). Several states in India experienced massive out-
etc. Indian Ocean regions with reported cases are La Reunion, breaks of Chikungunya viral infection during 200409, including
Madagascar, Mauritius, Seychelles etc. (Hammon, Rudnick and Andhra Pradesh, Karnataka, Maharashtra, Rajasthan, Gujarat,
Sather 1960; Shah, Gibbs and Banerjee 1964; Thaung et al. 1975; Tamilnadu, Orissa and Madhya Pradesh (Cavrini et al. 2009).
Thonnon et al. 1999; Lam et al. 2001; Muyembe-Tamfum et al. Subsequent smaller outbreaks of Chikungunya viral infection
2003; Laras et al. 2005; Schuffenecker et al. 2006; Lanciotti et al. have been reported from many parts of India after the massive
2007; Renault et al. 2007; Cavrini et al. 2009). Chikungunya has 200409 epidemic. The main affected regions were Delhi,
been reported to re-emerge unexpectedly in many countries Haryana, Kolkata, Kerala, Andaman and Nicobar Island (Pialoux
with a gap of 720 years (Powers et al. 2000). The first Asian et al. 2007; Niyas et al. 2010; Singh et al. 2010; Shrinet et al. 2012;
outbreak was documented in 1958 in Bangkok and Thailand Afreen et al. 2014). These studies have reported 845% CHIKV
(Jupp and McIntosh 1988), after which there were outbreaks in positivity using different diagnostic methods, including PCR,
many parts of India in 196365. Later on re-emerging epidemics real-time PCR and serological assays.
6 Pathogens and Disease, 2016, Vol. 74, No. 3

A detailed investigation of the evolution of Chikungunya PREVENTION AND CONTROL


virus strains in India from 1963 to 2006 was carried out dur-
Vector control is the only way to reduce CHIKV infections due
ing the massive outbreak. The phylogenetic characterization
to the absence of an effective vaccine and antiviral drugs. Pre-
showed that the strains identified during 196373 clustered
vention of mosquito breeding can be achieved by elimination
into the Asian genotype. All the isolates identified during
of stagnant water in drains, room air coolers, broken bottles,
the 200409 epidemic clustered into the ECSA genotype mak-
tin cans, etc. Insecticides should be used to control the adult
ing a separate group, IOL (Indian Ocean Lineage) (Arankalle
and larval mosquito population. Protection from mosquito bites
et al. 2007). Another investigation clustered the CHIKV strains
can be achieved by using mosquito-repellent creams, wearing
in the ECSA genotype by phylogenetic analysis of the E2
clothes which cover the body, use of mosquito nets over the
protein gene during another small outbreak in Odisha in
beds and the use of wire mesh on doors and windows. There-
201011 (Sahu et al. 2013). Various mutations were also identi-
fore, vector surveillance should be carried out in the endemic
fied in the E2 protein. The T cell and B cell epitopes were also
areas for effective control of infection. In addition, the general
predicted for the E protein and two of them were found to be
public should be educated on various aspects of the disease that
associated with antigenicity. Structural characterization by ho-
will assist in better management of this viral infection.
mology modeling revealed three domains (A, B and C) in the
E2 protein. Most of the mutations were identified in the do-
main B, which modulates the binding of virion with the host
THERAPEUTICS AND CURE
cell, and domain C is the transmembrane domain. The most

Downloaded from http://femspd.oxfordjournals.org/ by Shama Parveen on February 4, 2016


probable B and T cell epitopes were identified in the conserved No antiviral drugs are available for CHIKV infections, therefore
domain A. the treatment is completely symptomatic. Antipyretics, anal-
An investigation of full genome analysis of five isolates from gesics and anti-inflammatory drugs are usually given for the
Pune during the 200409 epidemic identified numerous muta- treatment of fever, pain, etc. Attempts are being made to develop
tions throughout the genome along with the typical A226V mu- therapeutics and drugs for the treatment of CHIKV infection
tation (Arankalle et al. 2007). Comprehensive full genomic anal- by various scientists all over the world. Chloroquinone has an-
ysis of two outbreak strains from Gwalior revealed the pres- tiviral activity against many viruses, including CHIKV, but a re-
ence of three unique mutations, two in the E1 protein and cent study from India showed that chloroquinone was not effec-
one in the nsp1 protein (Santhosh et al. 2008). These unique tive in the treatment of early musculoskeletal pain and arthri-
molecular features, including the A226V mutation in the E1 tis in acute CHIKV infection compared with the non-steroidal
protein, were found to be associated with high evolutionary anti-inflammatory drug (NSAID) e.g. meloxicam (Chopra, Saluja
and epidemic potential of these CHIKV strains. Full genome and Venugopalan 2014). Ribavarin also has antiviral activity
analysis of the six isolates of these outbreak strains from Ker- against Chikungunya virus. Ribavarin in combination with INF-
ala revealed the presence of 37 novel mutations (Sreekumar 2b showed a synergistical antiviral effect against Chikungunya
et al. 2010). At least 20 mutations mapped to the functionally viral infection in in vitro conditions (Briolant et al. 2004). Another
significant domains of the corresponding viral proteins. They study of the combination of ribavarin with doxycycline showed
also concluded that 18 mutations in the structural proteins af- a decrease in CHIKV replication in cell culture and decreased vi-
fected the T cell epitope immunogenicity. Another investiga- ral load and inflammation in mice models (Rothan et al. 2015).
tion from Pune studied the evolution of the CHIKV during the Another broad spectrum antiviral drug, favipiravir, was shown
massive epidemic using 27 full genomes and partial E1 gene to inhibit replication of different Chikungunya virus strains in
datasets. The mean substitution rate of the CHIKV was calcu- cell cultures in a recent investigation (Delang et al. 2014). The
lated to be 8.8 104 substitutions site1 year1 (Cherian et al. mice experiments with favipiravir protected mice from severe
2009). The evolutionary time scale of the CHIK viruses was es- neurological disease and a decrease in mortality rate was also
timated to be 300 years old. In addition, the progenitor of the observed.
200507 viruses was identified 9 years ago that originated in The use of human immunoglobulins purified from convales-
Central Africa. cent phase CHIKV patients proved to have a good prophylactic
Numerous phylogenetic studies have also provided insights and therapeutic efficacy against CHIKV infection in mouse mod-
into the molecular evolution of Chikungunya virus in India els (Couderc et al. 2008). In one particular study two different
(Santhosh et al. 2008). Comparison of synonymous and non- monoclonal antibodies, 5F10 and 8B10, were prepared against
synonymous substitution rates in different genomic regions CHIKV. These antibodies were formulated by activation, immor-
suggested purifying selection pressures (Arankalle et al. 2007; talization, amplification and cloning of B cells from a donor with
Sahu et al. 2013). This analysis also revealed the presence of a history of CHIKV infection. Both these antibodies specifically
many positively selected sites in the genome. An investigation neutralize CHIKV infection strongly in in vitro conditions (Warter
from Pune identified 10 positively selected sites throughout the et al. 2011). The level of MCP-1 was found to be elevated dur-
genome (Arankalle et al. 2007). Five positively selected sites were ing the CHIKV infection. Therefore, one particular study demon-
identified in the E2 region in most of the isolates in a study from strated that treatment of infected mice with bindarit, which is
Odisha (Sahu et al. 2013). However, the affect of these positively an inhibitor of MCP-1 synthesis, showed symptoms that were
selected sites on the pathogenesis of Chikungunya virus infec- mild and of short duration. The histology of joint and muscle tis-
tions should be investigated further. The phylogenetic investi- sue of infected mice treated with bindarit also showed very low
gations from different parts of India have thus contributed to inflammation compared with the untreated mice (Hoarau, Jaf-
the understanding of the molecular evolution and epidemiology far Bandjee and Krejbich Trotot 2010). Trials of antiviral therapy
of this re-emerging viral pathogen. Therefore, regular surveil- using RNA interference-mediated inhibition of viral replication
lance for the Chikungunya viral infection should be carried out are promising but sequence dependent. A reduction of CHIKV
in endemic regions like India. The data generated from the titer up to 99.6% in specially designed small interfering (si)RNA
surveillance will assist in the formulation of control measures to for conserved regions of nsp3 and E1 gene was reported in an
prevent outbreaks in this region. in vitro study. Complete inhibition of viral replication targeting
Deeba et al. 7

ns1 and E2 protein genes in in vivo conditions was reported in these vaccine candidates was achieved in the cell cultures. In ad-
another investigation (Dash et al. 2008; Parashar et al. 2013). dition, all the vaccine candidates generated a robust immune re-
sponse in mice. Wang and colleagues developed another recom-
binant vaccine candidate comprising the non-replicating aden-
VACCINES ovirus vector expressing the structural proteins of CHIKV (Wang
The recurring epidemics, re-emergence and widespread global et al. 2011). The vaccine induced a good humoral immune re-
distribution of CHIKV have necessitated the development of an sponse in mice. In an effort to develop a vaccine against CHIKV
effective vaccine. Challenges to vaccine development for the infection, Plante and colleagues replaced the subgenomic pro-
CHIKV infection are enormous. The vaccine should induce high moter in a CHIKV clone by internal ribosome entry site (IRES)
levels of antibodies to completely protect against the CHIKV in- from encephalomyocarditis virus and thus altered the levels
fection. In addition, the vaccine should be economical so that and host-specific mechanism of structural protein gene expres-
developing countries can easily afford them since most of the sion (Muthumania et al. 2008). This vaccine was found to be
epidemics occur in these regions. Many clinical and preclinical highly immunogenic and efficacious in mice. In addition, the
trials for development of an effective and economical vaccine vaccine was found to be incapable of replicating in the mosquito
are being carried out across the globe (Singh et al. 2013). cells. Another potential vaccine candidate is MVA-CHIKV that is
based on modified vaccinia virus Ankara (MVA) expressing all
the structural genes of Chikungunya virus (capsid, E3, E2, 6K and
WHOLE VIRUS-INACTIVATED VACCINES E1) (Plante et al. 2011). The trials of the above-mentioned vaccine

Downloaded from http://femspd.oxfordjournals.org/ by Shama Parveen on February 4, 2016


candidate were carried out on mice that showed both potent
The initial vaccine candidate that was developed was a whole
and broad cellular immune responses even on administration
virus grown in a green monkey kidney cell line inactivated by
of high viral doses.
formalin (Harrison et al. 1971). The preliminary clinical trials
showed good immune response with few side effects. Three dif-
ferent CHIKV strains grown in cell culture that were inactivated DNA VACCINES
by Tweenether showed good immune response in preclinical
trials (Eckels, Harrison and Hetrick 1970). Another approach for the design of a vaccine against Chikun-
The formalin-inactivated whole virus vaccine was generated gunya virus includes the development of DNA vaccines. During
from the ECSA genotype of the 2006 Indian outbreak strain. an investigation on DNA-based vaccine, several modifications
This vaccine had a very good immunogenic potential to neutral- were made in the consensus sequences of capsid and envelope
ize the infectivity of the virus by enhancing both the humoral protein genes of CHIKV that included codon optimization, addi-
and cell-mediated immune response in a mouse model (Tiwari tion of a Kozak sequence, and a substituted immunoglobulin E
et al. 2009). Another vaccine candidate was also derived from the leader sequence inserted in a plasmid (Wanga et al. 2008). The
ECSA genotype of CHIKV. The whole virus was inactivated by vaccine was able to induce both humoral and cellular immune
-propiolactone (BPL)formalin, which proved to be a promis- response in mice. Vaccination using envelope-based synthetic
ing vaccine candidate in the mouse model (Kumar, Sudeep and DNA vaccine using a combined E1, E2 and E3 gene construct
Arankalle 2012). The inactivated whole virus vaccine derived was found to generate a humoral as well as a cell-mediated im-
from an Asian isolate completely protected against mice viremia mune response in rhesus macaques, each of which is capable of
and arthritis in another study (Gardner et al. 2010). providing protection against the CHIKV infection (Garcia-Arriaza
et al. 2014).

LIVE ATTENUATED VACCINES


VIRUS-LIKE PARTICLES
The first live attenuated vaccine strain for CHIKV was developed
by Levitt and colleagues in 1986 by passaging a Thailand strain of The selective expression of structural proteins (C-E3-E2-6K-E1)
CHIKV in cell culture (Levitt et al. 1986). The vaccine was able to of the West African strain of CHIKV forms non-infectious virus-
replicate in both species of the vector A. aegepti and A. albopictus. like particles (VLPs) in vitro and showed high titers of neutral-
The live attenuated vaccine was developed by serially passaging izing antibodies both in monkeys and immunodeficient mice
plaque-purified CHIKV in the MRC-5 (Human fetal lung fibrob- (Akahata et al. 2010). The recombinant baculoviruses were used
last) cell line. This vaccine was safe and highly immunogenic to generate CHIKV VLPs in insect cells in another study (Metz
with side effects well within the tolerable limits. In the phase II et al. 2013). The VLPs provided protection against viremia and
trial, investigators observed sero-conversion in 98% of the vac- arthritis in the mouse model. The VLPs generated in another re-
cines by day 28 and the presence of neutralizing antibodies in cent investigation proved to be a most promising vaccine can-
85% of volunteers for 12 months (Edelman et al. 2000). CHIKV didate as the phase I clinical trials showed immunogenic, well-
with point mutations at amino acid numbers 12 and 82 in the tolerated and safe effects (Chang et al. 2014).
E2 glycoprotein proved to be a good candidate for development
of live attenuated vaccine in another investigation (Gorchakov
et al. 2012). CONCLUSION
The Chikungunya virus is a re-emerging infection affecting mil-
lions of people worldwide. The Chikungunya viral infection is
SYNTHETIC VACCINES
associated with a number of challenges such as overlapping
Three different chimeric alphavirus vaccines were developed clinical symptoms with dengue virus, recurring epidemics, per-
by using naturally attenuated strains of Venezuelan equine en- sistent arthralgia and mutations in viral genome leading to
cephalitis virus, Eastern equine encephalitis virus and Sind- enhanced geographical spread and inability to control the
bis virus as a backbone expressing structural protein genes of vector-mediated transmission of the disease. The recurring and
CHIKV (Mallilankaraman et al. 2011). An efficient replication of persistent epidemics in different geographical regions have
8 Pathogens and Disease, 2016, Vol. 74, No. 3

enhanced efforts to develop drugs and vaccines to combat Cherian SS, Walimbe AM, Jadhav SM et al. Evolutionary rates
Chikungunya virus infection. and timescale comparison of Chikungunya viruses inferred
The Chikungunya viral infection is a major health concern in from the whole genome/E1 gene with special reference to the
India. Therefore, continuous viral surveillance should be carried 200507 outbreak in the Indian subcontinent. Infect Genet Evol
out in the regions reporting regular outbreaks in the monsoon 2009;9:1623.
season. This surveillance will describe the circulating genotypes Chopra A, Saluja M, Venugopalan A. Effectiveness of chloro-
of Chikungunya virus in these regions. The information on the quine and inflammatory cytokine response in patients with
circulating strains of Chikungunya virus will assist efforts to de- early persistent musculoskeletal pain and arthritis follow-
velop vaccines and antiviral drugs. These data will also facilitate ing chikungunya virus infection. Arthritis Rheumatol 2014;66:
the prediction and control of the epidemics. In addition, an in- 31926.
crease in the general awareness of the disease by educating the Chow A, Her Z, Ong EKS et al. Persistent arthralgia induced by
general public can be beneficial for a better management of this Chikungunya virus infection is associated with interleukin-
emerging viral infection. 6 and granulocyte macrophage colony- stimulating factor.
J Infec Dis 2011;203:14957.
Couderc T, Chretien F, Schilte C et al. A mouse model for chikun-
FUNDING gunya: young age and inefficient type-I interferon signaling
Work in our laboratory is supported by grants from the Univer- are risk factors for severe disease. PLoS Pathog 2008;4:e29.
sity Grants Commission and the Council of Scientific and Indus- Cronstein BN. Interleukin-6a key mediator of systemic and lo-

Downloaded from http://femspd.oxfordjournals.org/ by Shama Parveen on February 4, 2016


trial Research, Government of India. cal symptoms in rheumatoid arthritis. Bull NYU Hosp Jt Dis
2007;65:S115.
Conflict of interest. None declared. Dash PK, Tiwari M, Santhosh SR et al. RNA interference mediated
inhibition of Chikungunya virus replication in mammalian
cells. Biochem Biophys Res Commun 2008;376:71822.
REFERENCES
Delang L, Segura Guerrero N, Tas A et al. Mutations in the
Abere B, Wikan N, Ubol S et al. Proteomic analysis of chikun- chikungunya virus non-structural proteins cause resistance
gunya virus infected microgial cells. PLoS One 2012;7:e34800. to favipiravir (T-705), a broad-spectrum antiviral. J Antimicrob
Afreen N, Deeba F, Khan WH et al. Molecular characterization Chemother 2014;69:277084.
of dengue and chikungunya virus strains circulating in New Deshmane SL, Kremlev S, Amini S et al. Monocyte chemoattrac-
Delhi, India. Microbiol Immunol 2014;58:68896. tant protein-1 (MCP-1): an overview. J Interferon Cytokine Res
Akahata W, Yang ZY, Andersen H et al. A virus like par- 2009;29:31326.
ticle vaccine for epidemic chikungunya virus protects Dupuis-Maguiraga L, Noret M, Brun S et al. Chikungunya dis-
nonhuman primates against infection. Nat Med 2010;16: ease: infection-associated markers from the acute to the
3348. chronic phase of arbovirus-induced arthralgia. PLoS Negl Trop
Apandi Y, Nazni WA, Noor Azleen ZA et al. The first isolation of Dis 2012;6:e1446.
chikungunya virus from nonhuman primates in Malaysia. J Eckels KH, Harrison VR, Hetrick FM. Chikungunya virus vac-
Gen Mol Virol 2009;1:0359. cine prepared by a Tween-ether extraction. Appl Microbiol
Arankalle VA, Shrivastava S, Cherian S et al. Genetic divergence 1970;19:3215.
of Chikungunya viruses in India (19632006) with special Edelman R, Tacket CO, Wasserman SS et al. Phase II safety and
reference to the 20052006 explosive epidemic. J Gen Virol immunogenicity study of live chikungunya virus vaccine TSI-
2007;88:196776. GSD-218. Am J Trop Med Hyg 2000;62:6815.
Bhatia R, Narain JP. Re-emerging chikungunya fever: some Edwards CJ, Welch SR, Chamberlain J et al. Molecular diagnosis
lessons from Asia. Trop Med Int Health 2009;14:9406. and analysis of Chikungunya virus. J Clin Virol 2007;39:2715.
Borgherini G, Poubeau P, Staikowsky F et al. Outbreak of chikun- Erasmus JH, Needham J, Raychaudhuri S et al. Utilization of an
gunya on Reunion Island: early clinical and laboratory fea- Eilat virus-based chimera for serological detection of chikun-
tures in 157 adult patients. Clin Infect Dis 2007;44:14017. gunya infection. PLoS Negl Trop Dis 2015;9:e0004119.
Brehin AC, Rubrecht L, Sanchez MEN et al. Production and Garcia-Arriaza J, Cepeda V, Hallengard D et al. A novel
characterization of mouse monoclonal antibodies reac- poxvirus-based vaccine (MVA-CHIKV) is highly immuno-
tive to Chikungunya envelope E2 glycoprotein. Virology genic and protects mice against chikungunya infection. J Vi-
2008;371:18595. rol 2014;88:352747.
Briolant S, Garin D, Scaramozzino N et al. In vitro inhibition of Gardner J, Anraku I, Le TT et al. Chikungunya virus arthritis in
Chikungunya and Semliki Forest viruses replication by an- adult wild-type mice. J Virol 2010;84:802132.
tiviral compounds: synergistic effect of interferon- and rib- Gorchakov R, Wang E, Leal G et al. Attenuation of Chikungunya
avirin combination. Antiviral Res 2004;61:1117. virus vaccine strain 181/clone 25 is determined by two amino
Burke DS, Nisalak A, Nimmannitya S. Disappearance of Chikun- acid substitutions in the E2 envelope glycoprotein. J Virol
gunya virus from Bangkok. Trans R Soc Trop Med Hyg 2012;86:608496.
1985;79:41920. Hammon WM, Rudnick A, Sather GE. Viruses associated with
Caglioti C, Lalle E, Castilletti C et al. Chikungunya virus infection: epidemic hemorrhagic fevers of the Philippines and Thai-
an overview. New Microbiol 2013;36:21127. land. Science 1960;131:11023.
Cavrini F, Gaibani P, Pierro AM et al. Chikungunya: an emerg- Harrison VR, Eckels KH, Bartelloni PJ et al. Production and eval-
ing and spreading arthropod-borne viral disease. J Infect Dev uation of a formalin-killed Chikungunya vaccine. J Immunol
Ctries 2009;3:74452. 1971;107:6437.
Chang L-J, Dowd KA, Mendoza FH et al. Safety and tolerability of Hirano T, Taga T, Nakano N et al. Purification to homogeneity
chikungunya virus-like particle vaccine in healthy adults: a and characterization of human B-cell differentiation factor
phase 1 dose-escalation trial. Lancet 2014;384:204652. (BCDF or BSFp-2). Proc Natl Acad Sci USA 1985;82:54904.
Deeba et al. 9

Hoarau JJ, Jaffar Bandjee MC, Krejbich Trotot P. Persistent chronic Muthumania K, Lankaraman KM, Laddy DJ et al. Immunogenic-
inflammation and infection by Chikungunya arthritogenic ity of novel consensus-based DNA vaccines against Chikun-
alphavirus in spite of a robust host immune response. J Im- gunya virus. Vaccine 2008;26:512834.
munol 2010;184:591427. Muyembe-Tamfum JJ, Peyrefitte CN, Yogolelo R et al. Epidemic of
Jupp PG, McIntosh BM. Chikungunya disease. In: Monath TP (ed.). Chikungunya virus in 1999 and 2000 in the Democratic Re-
The Arboviruses: Epidemiology and Ecology. Boca Raton Florida: public of the Congo. Med Trop (Mars) 2003;63:6378.
CRC Press, 1988, 13757. Neogi DK, Bhattacharya N, Mukherjee KK et al. Serosurvey of
Kam YW, Simarmata D, Chow A et al. Early appearance of neu- chikungunya antibody in Calcutta metropolis. J Commun Dis
tralizing immunoglobulin G3 antibodies is associated with 1995;27:1922.
chikungunya virus clearance and long-term clinical protec- Ng LFP, Chow A, Sun YJ et al. IL-1, IL-6, and RANTES
tion. J Infec Dis 2012;205:114754. as biomarkers of Chikungunya severity. PLoS One 2009;4:
Kawano Y, Noma T, Yata J. Regulation of human IgG subclass e4261.
production by cytokines. IFN-gamma and IL-6 act antagonis- Niyas KP, Abraham R, Unnikrishnan RN et al. Molecular charac-
tically in the induction of human IgG1 but additively in the terization of Chikungunya virus isolates from clinical sam-
induction of IgG2. J Immunol 1994;153:494858. ples and adult Aedes albopictus mosquitoes emerged from lar-
Kelvin AA, Banner D, Silvi G et al. Inflammatory cytokine expres- vae from Kerala, South India. Virol J 2010;7:189.
sion is associated with chikungunya virus resolution and Obeyesekere I, Hermon Y. Arbovirus heart disease: Myocardi-
symptom severity. PLoS Negl Trop Dis 2011;5:e1279. tis and cardiomyopathy following dengue and chikun-

Downloaded from http://femspd.oxfordjournals.org/ by Shama Parveen on February 4, 2016


Kumar M, Sudeep AB, Arankalle VA. Evaluation of recombinant gunya feverA follow-up study. Am Heart J 1973;85:
E2 protein-based and whole-virus inactivated candidate vac- 18694.
cines against chikungunya virus. Vaccine 2012a;30:61429. Ozden S, Huerre M, Riviere J-P et al. Human muscle satellite
Kumar S, Jaffer-Bandjee MC, Giry C et al. Mouse macrophage in- cells as targets of Chikungunya virus infection. PLoS One
nate immune response to chikungunya virus infection. Virol 2007;6:e527.
J 2012b;9:313. Padbidri VS, Gnaneswar TT. Epidemiological investigations of
Lalitha P, Rathinam S, Banushree K et al. Ocular involvement as- Chikungunya epidemic at Barsi, Maharashtra State, India. J
sociated with an epidemic outbreak of Chikungunya virus in- Hyg Epidemiol Microbiol Immunol 1979;23:44551.
fection. Am J Ophthalmol 2007;144:5526. Parashar D, Paingankar MS, Kumar S et al. Administration of
Lam SK, Chua KB, Hooi PS et al. Chikungunya infectionan E2 and NS1 siRNAs inhibit chikungunya virus replication in-
emerging disease in Malaysia. Southeast Asian J Trop Med Pub- vitro and protects mice infected with the virus. PLoS Negl Trop
lic Health 2001;32:44751. Dis 2013;7:e2405.
Lanciotti RS, Kosoy OL, Laven JJ et al. Chikungunya virus in Parola P, de Lamballerie X, Jourdan J et al. Novel chikungunya
US travelers returning from India, 2006. Emerg Infect Dis virus variant in travelers returning from Indian Ocean is-
2007;13:7647. lands. Emerg Infect Dis 2006;12:149399.
Laras K, Sukri NC, Larasati RP et al. Tracking the re-emergence Pastorino B, Muyembe-Tamfum JJ, Bessaud M et al. Epidemic
of epidemic chikungunya virus in Indonesia. Trans R Soc Trop resurgence of Chikungunya virus in Democratic Republic of
Med Hyg 2005;99:12841. the Congo: Identification of a new central African strain. J
Levitt NH, Ramsburg HH, Hasty SE et al. Development of an at- Med Virol 2004;74:27782.
tenuated strain of chikungunya virus for use in vaccine pro- Peyrefitte CN, Rousset D, Pastorino BAM et al. Chikungunya virus,
duction. Vaccine 1986;4:15762. Cameroon. Emerg Infect Dis 2007;13:5.
Lumsden WH. An epidemic of virus disease in southern Pialoux G, Gauzere BA, Jaureguiberry S et al. Chikungunya, an
province, Tanganyika Territory, in 195253. II. General de- epidemic arbovirosis. Lancet Infect Dis 2007;7:31927.
scription and epidemiology. Trans R Soc Trop Med Hyg Plante K, Wang E, Partidos CD et al. Novel Chikungunya vaccine
1955;49:3357. candidate with an IRES-based attenuation and host range al-
Mallilankaraman K, Shedlock DJ, Bao H et al. A DNA vaccine teration mechanism. PLoS Pathog 2011;7:e1002142.
against chikungunya virus is protective in mice and induces Powers AM, Brault AC, Tesh RB et al. Re-emergence of chikun-
neutralizing antibodies in mice and nonhuman primates. gunya and onyong-nyong viruses: evidence for distinct ge-
PLoS Negl Trop Dis 2011;5:e928. ographical lineages and distant evolutionary relationships. J
Malvy D, Ezzedine K, Mamani-Matsuda M et al. Destructive Gen Virol 2000;8:4719.
arthritis in a patient with chikungunya virus infection with Powers AM, Logue CH. Changing patterns of chikungunya virus:
persistent specific IgM antibodies. BMC Infect Dis 2009;9:200. reemergence of a zoonotic arbovirus. J Gen Virol 2007;88:
Manimunda SP, Singh SS, Sugunan AP et al. Chikungunya 236377.
fever, Andaman and Nicobar Islands, India. Emerg Infect Dis Qin S, Rottman JB, Myers P et al. The chemokine receptors CXCR3
2007;13:125960. and CCR5 mark subsets of T cells associated with certain in-
Manimunda SP, Vijayachari P, Uppoor R et al. Clinical pro- flammatory reactions. J Clin Invest 1998;101:74654.
gression of chikungunya fever during acute and chronic Quatresous I. E-alert27 January: Chikungunya outbreak in
arthritic stages and the changes in joint morphology as re- Reunion, a French Overseas Department. Eurosurveillance
vealed by imaging. Trans R Soc of Trop Med Hyg 2010;104: Weekly 2006;11:E060202.
3929. Rao TR. Recent epidemics caused by Chikungunya virus in India,
Metz SW, Gardner J, Geertsema C et al. Effective chikungunya 19631965. Sci Cult 1966;32:21520.
virus-like particle vaccine produced in insect cells. PLoS Negl Ravi V. Re-emergence of Chikungunya virus in India. Indian J Med
Trop Dis 2013;7:e2124. Microbiol 2006;24:834.
Mittal A, Mittal S, Bharati MJ et al. Optic neuritis associated with Renault P, Solet JL, Sissoko D et al. A major epidemic of Chikun-
chikungunya virus infection in South India. Arch Ophthalmol gunya virus infection on Reunion Island, France, 20052006.
2007;125:13816. Am J Trop Med Hyg 2007;77:72731.
10 Pathogens and Disease, 2016, Vol. 74, No. 3

Robin S, Ramful D, Zettor J et al. Severe bullous skin lesions as- Solignat M, Gay B, Higgs S et al. Replication cycle of chikungunya:
sociated with chikungunya virus infection in small infants. A re-emerging arbovirus. Virology 2009;393:18397.
Eur J Pediatr 2009;169:6772. Sreekumar E, Issac A, Nair S et al. Genetic characterization
Robinson M. An epidemic of virus disease in southern province, of 20062008 isolates of Chikungunya virus from Kerala,
Tanganyika Territory, in 195253. I. Trans R Soc Trop Med Hyg South India, by whole genome sequence analysis. Virus Genes
1955;49:2832. 2010;40:1427.
Rodrigues FM, Patankar MR, Banerjee K et al. Etiology of the1965 Talarmin F, Staikowsky F, Schoenlaub P et al. Skin and mucosal
epidemic of febrile illness in Nagpur city, Maharashtra State, manifestations of chikungunya virus infection in adults in
India. Bull World Health Organ 1972;46:1739. Reunion Island. Med Trop (Mars) 2007;67:16773.
Rot A, von Andrian UH. Chemokines in innate and adaptive Thaung U, Ming CK, Swe T et al. Epidemiological features of
host defense: basic chemokinese grammar for immune cells. dengue and chikungunya infections in Burma. Southeast
Annu Rev Immunol 2004;22:891928. Asian J Trop Med Public Health 1975;6:27683.
Rothan HA, Bahrani H, Mohamed Z et al. A combination of doxy- Thonnon J, Spiegel A, Diallo M et al. Chikungunya virus outbreak
cycline and ribavirin alleviated chikungunya infection. PLoS in Senegal in 1996 and 1997. Bull Soc Pathol Exot 1999;92:7982.
One 2015;10:e0126360. Tiwari M, Parida M, Santhosh SR et al. Assessment of immuno-
Rulli NE, Rolph MS, Srikiatkhachorn A et al. Protection from genic potential of Vero adapted formalin inactivated vaccine
arthritis and myositis in a mouse model of acute chikun- derived from novel ECSA genotype of Chikungunya virus.
gunya virus disease by bindarit, an inhibitor of monocyte Vaccine 2009;27:251322.

Downloaded from http://femspd.oxfordjournals.org/ by Shama Parveen on February 4, 2016


chemotactic protein-1 synthesis. J Infec Dis 2011;204:102630. Tsetsarkin KA, Vanlandingham DL, McGee CE et al. A single mu-
Sahu A, Das B, Das M et al. Genetic characterization of E2 re- tation in Chikungunya virus affects vector specificity and
gion of Chikungunya virus circulating in Odisha, Eastern In- epidemic potential. PLoS Pathog 2007;3:e201.
dia from 2010 to 2011. Infect Genet Evol 2013;18:11324. Vashishtha M, Phalen T, Marquardt MT et al. A single point mu-
Sam IC, AbuBakar S. Chikungunya virus infection. Med J Malaysia tation controls the cholesterol dependence of Semliki Forest
2006;61:2649. virus entry and exit. J Cell Biol 1998;140:919.
Santhosh SR, Dash PK, Parida MM et al. Comparative full genome Vazeille M, Moutailler S, Coudrier D et al. Two Chikungunya iso-
analysis revealed E1: A226V shift in 2007 Indian Chikungunya lates from the outbreak of La Reunion (Indian Ocean) exhibit
virus isolates. Virus Res 2008;135:3641. different patterns of infection in the mosquito, Aedes albopic-
Schaible HG, von Banchet GS, Boettger MK et al. The role of proin- tus. PLoS One 2007;11:e1168.
flammatory cytokines in the generation and maintenance of Volk SM, Chen R, Tsetsarkin KA et al. Genome-scale phylogenetic
joint pain. Ann N Y Acad Sci 2010;1193:609. analyses of chikungunya virus reveal independent emer-
Schilte C, Staikovsky F, Couderc T et al. Chikungunya virus- gences of recent epidemics and various evolutionary rates.
associated long-term arthralgia: A 36-month prospective J Virol 2010;84:6497.
longitudinal study. PLoS Negl Trop Dis 2013;7:e2137. Wang D, Suhrbier A, Penn-Nicholson A et al. A complex ade-
Schuffenecker I, Iteman I, Michault A et al. Genome microevolu- novirus vaccine against chikungunya virus provides com-
tion of chikungunya viruses causing the Indian Ocean out- plete protection against viraemia and arthritis. Vaccine
break. PLoS Med 2006;3:e263. 2011;29:28039.
Schwartz O, Albert ML. Biology and pathogenesis of Chikun- Wanga E, Volkovac E, Adams AP et al. Chimeric alphavirus vac-
gunya virus. Nat Rev 2010;8:491500. cine candidates for Chikungunya. Vaccine 2008;26:50309.
Sergon K, Njuguna C, Kalani R et al. Seroprevalence of Chikun- Warter L, Lee CY, Thiagarajan R et al. Chikungunya virus
gunya virus (CHIKV) infection on Lamu Island, Kenya, Octo- envelope-specific human monoclonal antibodies with broad
ber 2004. Am J Trop Med Hyg 2008;78:3337. neutralization potency. J Immunol 2011;186:325864.
Shah KV, Gibbs CJ, Banerjee G. Virological investigation of Wauquier N, Becquart P, Nkoghe D et al. The acute phase of
the epidemic of haemorrhagic fever in Calcutta: isolation chikungunya virus infection in humans is associated with
of three strains of chikungunya virus. Indian J Med Res strong innate immunity and T CD8 cell activation. J Infec Dis
1964;52:67683. 2011;204:11523.
Shrinet J, Jain S, Sharma A et al. Genetic characterization of Weaver SC, Forrester NL. Chikungunya: Evolutionary history and
Chikungunya virus from New Delhi reveal emergence of a recent epidemic spread. Antiviral Res 2015;120:329.
new molecular signature in Indian isolates. Virol J 2012;9:100. Weaver SC, Frey TK, Huang HV et al. Togaviridae. In: Fauquet CM,
Singh P, Chhabra M, Mittal V et al. Current research and clinical Mayo MA, Maniloff J, Desselberger U, Ball LA (eds). Virus Tax-
trials for a vaccine against Chikungunya virus. Vaccine (Auckl) onomy: Eighth Report of the International Committee on Taxonomy
2013;3:3546. of Viruses. San Diego, CA, USA, London: Elsevier Academic
Singh P, Mittal V, Rizvi MMA et al. The first dominant co- Press, 2005, 9991008.
circulation of both dengue and chikungunya viruses during Yergolkar PN, Tandale BV, Arankalle VA et al. Chikungunya out-
the post-monsoon period of 2010 in Delhi, India. Epidemiol breaks caused by African genotype, India. Emerg Infect Dis
Infect 2010;140:133742. 2006;12:15803.

View publication stats

Você também pode gostar