You are on page 1of 10

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

net/publication/40756904

Chikungunya Fever: CNS Infection and Pathologies of a Re-Emerging


Arbovirus

Article in Progress in Neurobiology · December 2009


DOI: 10.1016/j.pneurobio.2009.12.006 · Source: PubMed

CITATIONS READS

173 928

13 authors, including:

Trina Das Marie Christine Jaffar-Bandjee

12 PUBLICATIONS 1,088 CITATIONS


University of La Réunion
98 PUBLICATIONS 3,379 CITATIONS
SEE PROFILE
SEE PROFILE

Jean-Jacques Hoarau Pascale Krejbich Trotot


University of La Réunion University of La Réunion
71 PUBLICATIONS 1,548 CITATIONS 59 PUBLICATIONS 2,309 CITATIONS

SEE PROFILE SEE PROFILE

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

Dengue in Reunion island View project

Dengue Virus in La Réunion Island View project

All content following this page was uploaded by Pascale Krejbich Trotot on 17 November 2019.

The user has requested enhancement of the downloaded file.


Progress in Neurobiology 91 (2010) 121–129

Contents lists available at ScienceDirect

Progress in Neurobiology
journal homepage: www.elsevier.com/locate/pneurobio

Chikungunya fever: CNS infection and pathologies of a re-emerging arbovirus


Trina Das, Marie Christine Jaffar-Bandjee, Jean Jacques Hoarau, Pascale Krejbich Trotot, Melanie Denizot,
Ghislaine Lee-Pat-Yuen, Renubala Sahoo, Pascale Guiraud, Duksha Ramful, Stephanie Robin,
Jean Luc Alessandri, Bernard Alex Gauzere, Philippe Gasque *
Immunopathology Infection Research Grouping, University and CHR Gelix Guyon of La Reunion, Saint Denis, 97400, Reunion

A R T I C L E I N F O A B S T R A C T

Article history: Chikungunya virus (CHIKV) is transmitted by Aedes mosquitoes and causes an acute symptomatic illness
Received 18 April 2009 with fever, skin rash, and incapacitating arthralgia, which can evolve into chronic rheumatoid arthritis in
Received in revised form 18 August 2009 elderly patients. This is a tropical disease originally described in central/east Africa in the 1960s, but its
Accepted 14 December 2009
2004 re-emergence in Africa and rapid spread in lands in and around the Indian Ocean (Reunion island,
India, Malaysia) as well as Europe (Italy) led to almost 6 million cases worldwide. The risk of importation
Keywords: and spreading diseases with long-term sequelae is even greater today given the global distribution of the
Alphavirus
vectors (including in the Americas), increased tourism and the apparent capacity of CHIKV to produce
Neuroinfection
Innate immunity
high levels of viremia (109–1012 virus/ml of blood) and new mutants. CHIKV-associated neuropathology
Neurodegeneration was described early in the 1960s, but it is the unprecedented incidence rate in Indian Ocean areas with
efficient clinical facilities that allowed a better description of cases with severe encephalitis,
meningoencephalitis, peripheral neuropathies and deaths among newborns (mother-to-child infection),
infants and elderly patients. Death rates following CHIKV infection were estimated at 1:1000 cases in la
Reunion’s outbreak. These clinical observations have been corroborated by experimental infection in
several mouse models, leading to CNS pathologies. We further describe in this review the capacity of
CHIKV to infect neurons and glial cells, delineate the fundamental innate (intrinsic) immune defence
mechanisms to protect from infection and argue about the possible mechanisms involved in the
encephalopathy.
ß 2010 Elsevier Ltd. All rights reserved.

Contents

1. Chikungunya, epidemiological and clinical data. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122


2. Neurological manifestations of Chikungunya in newborns, infants and adults . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 124
3. CHIKV neuroinfection and possible pathological mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 125
4. CNS innate immune protection against viral neuroinfection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 126
5. Conclusion: exploring novel anti-viral therapies to fight against CHIKV infection. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 127
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128

Abbreviations: APAF-1, apoptotic protease activating factor; CHIKV, Chikungunya virus; CNS, central nervous system; FADD, Fas-associated death domain; IAP, inhibitor of
apoptosis; IFN-a/b, type-I interferons; IFNAR, interferon receptor; ISGs, IFN-stimulated genes, e.g. IFN-stimulated protein of 15 kDa; IPS-1, IFN-b promoter stimulator-1, also
known as MAVS, CARDIF and VISA; IRF, interferon (IFN)-regulatory factor; MDA-5, melanoma differentiation-associated gene 5; Mx, myxovirus resistance () protein; NFkB,
nuclear factor-kB; OAS, oligoadenylate synthetase; PARP, poly(ADP-ribose) polymerase; PKR, IFN-inducible dsRNA-dependent protein kinase; PRR, pattern-recognition
receptors; RIG-I, retinoic-acid-inducible gene I-(RIG-I)-like helicases (RLHs).; RIP1, receptor-interacting protein 1; RNaseL, ribonuclease L; STAT1, signal transducer and
activator of transcription; SVZ, subventricular zone; TLR, Toll-like receptors (TLRs); TRADD, tumour-necrosis factor receptor-associated via death domain.
* Corresponding author at: IRG, University of la Réunion, St Denis, 15 Avenue René Cassin, BP7151, 97715, Ile de la Réunion. Tel.: +262 262 938836; fax: +262 262 938801.
E-mail addresses: gasque@univ-reunion.fr, gasquep@hotmail.com (P. Gasque).

0301-0082/$ – see front matter ß 2010 Elsevier Ltd. All rights reserved.
doi:10.1016/j.pneurobio.2009.12.006
122 T. Das et al. / Progress in Neurobiology 91 (2010) 121–129

Table 1
Main alphaviruses causing human pathologies and their geographical distribution.

1. Chikungunya, epidemiological and clinical data Africa (see Fig. 1, geographical distribution of CHIKV and Aedes
alpbopictus).
CHIKV is an Alphavirus of the Togaviridae family transmitted by CHIKV has also been isolated in western African countries
mosquitoes of the Aedes (Ae) genus (Pialoux et al., 2007; Weaver including Senegal, Benin, the Republic of Guinea, Cote d’Ivoire and
and Barrett, 2004). The alphavirus group comprises 29 viruses, 6 of Nigeria. In South East Asia, frequent outbreaks were reported from
which can cause human joint disorders (arthralgia evolving to the 1960s through to the 2000s in India, Malaysia, Indonesia,
arthritis), namely CHIKV, o’nyong-nyong virus (ONNV), Semliki Cambodia, Vietnam, Myanmar, Pakistan, Singapore and Thailand
Forest virus (SFV) (Africa, Asia, Europe), Ross River (RRV) (see comprehensive review; Powers and Logue, 2007). In 2004,
(Australia and the Pacific), Sindbis virus (SINV) (cosmopolitan), CHIKV re-emerged in Kenya and subsequently spread eastward,
and Mayaro virus (MAYV) (South America, French Guyana) causing millions of cases throughout countries in and around the
(Table 1). Indian Ocean. Estimated cases for India alone were in the order of
Three CHIKV lineages with distinct genotypic and antigenic 1.6–6 million infected people (Mavalankar et al., 2007). Outbreaks
characteristics have been identified: isolates from the Indian occurred almost continuously during 2004–2007 with cases
Ocean outbreak represent a distinct clade within a large east reported in Europe (Italy, UK, Belgium, Germany, Czech Republic,
central and southern African phylogroup distinct from the Asian Norway, Spain and France), Hong Kong, Canada, Taiwan, and Sri
phylogroup and from the West African phylogroup. To date, several Lanka and these were directly associated with the return of tourists
complete CHIKV nucleotide sequences have been determined and from India and affected islands of the Indian Ocean. The risk of
including the human isolates (Ross and S27) recovered during the emerging or re-emerging CHIKV epidemics is ever present because
1952–1953 outbreaks in Tanzania. Between the 1960s and 1990s, of the high attack rates, the high levels of viremia in acutely
the virus was frequently isolated from Central and Southern Africa infected humans (109–1012 virus per ml of blood) (our unpublished
including Sudan, Uganda, Democratic Republic of Congo the data and (Panning et al., 2008)), and the worldwide distribution of
Central African Republic, Malawi, Zimbabwe, Kenya and South the well-adapted vectors responsible for transmitting CHIKV
T. Das et al. / Progress in Neurobiology 91 (2010) 121–129 123

Fig. 1. Worldwide distribution of CHIKV (endemic in yellow shade and imported cases in blue) and Aedes albopictus (before 1980 in green and after 1980 in pink) (adapted
from Powers and Logue, 2007; Charrel et al., 2007).

(Fig. 1). For instance, CHIKV has hardly been reported in Australia, ranges from 3 to 7 days and as few as 5% of asymptomatic cases
the USA and South America but this could be a matter of future were generally reported. The laboratory diagnosis of CHIKV
concerns given that Aedes albopictus is now found in these infection is based on either specific serology or detection of viral
countries (Fig. 1) (Charrel et al., 2007; Lanciotti et al., 2007). RNA by reverse transcription-polymerase chain reaction (RT-PCR).
The CHIKV genome is approximately 11 kb and consists of a RT-PCR is positive during the viremic phase lasting for 1 week after
single-stand of positive-sense RNA with two open reading frames onset of the symptoms while the levels of IgG (and surprisingly
(ORF). The 50 ORF encodes the virus non-structural proteins (nsp) IgM) can last for months. Interestingly, and in contrast to Dengue,
1–4. The nsps are multifunctional and together form the virus the apparition of IgG anti-CHIKV was detected in the first week
replicase. The 30 ORF encodes the virus structural proteins, the following infection and illustrating the rapid and robust adaptive
capsid and the envelope glycoproteins E1 and E2. E3 is a secreted immune response (Panning et al., 2008). The acute signs and
polypeptide of unknown functions (Chevillon et al., 2008). symptoms usually resolve in less than 2 weeks, but arthralgia may
Interestingly, a mutation at residue 226 of the membrane fusion linger for weeks or months. Particularly in the elderly, CHIKV has
glycoprotein E1 (E1-A226 V), which was absent from the strains had rather profound acute arthritogenic activities and can
isolated during the first months of the ongoing outbreak in la contribute to chronic incapacitating arthritis as described for
Réunion, was found in more than 90% of isolates after September several alphaviruses (Harley et al., 2001; Levine et al., 1994;
2005 (Schuffenecker et al., 2006). This type of change originally Suhrbier and La Linn, 2004; Tesh, 1982; Toivanen, 2008).
described in another close alphavirus, SFV, could be related to virus Rheumatic manifestations in 10–20% of the patients typically
adaptation to the mosquito vector species as recently described consisted of a febrile arthritis mainly affecting the extremities
(Tsetsarkin et al., 2007; Vashishtha et al., 1998). Other CHIKV (ankles, wrists, phalanges) (Borgherini et al., 2008; Brighton and
isolates are currently being sequenced to provide spatiotemporal Simson, 1984; Fourie and Morrison, 1979; Simon et al., 2007;
data on the viruses isolated from different locations and to find out Sissoko et al., 2009). Beneficial treatments of these chronic
whether any molecular signatures could be associated with conditions involved the use of paracetamol with a nonsteroid
unusual clinical forms (Powers and Logue, 2007). The vast scale anti-inflammatory drug, glucocorticoids and methrotrexate in the
and sudden emergence of the la Réunion outbreak underline how most severe cases.
little we knew about the biology and pathology of CHIKV. Overall, Skin lesions were also reported and included vesiculobullous
270,000 cases were reported and this outbreak was the first time lesions with desquamation, aphthous-like ulcers and vasculitis
that a number of severe clinical manifestations, particularly lesions (Pialoux et al., 2007; Robin et al., 2009) as well as unusual
following infections of newborns and elderly (see below), and complications such as fulminant hepatitis, cardiologic manifesta-
rare mortality (estimated at 1:1000 of cases) were reported to be tions and neurologic diseases. Knowledge is currently limited
associated with CHIKV (Economopoulou et al., 2009; Gerardin about neurologic forms of CHIKV and its long-term sequelae
et al., 2008; Lemant et al., 2008; Mavalankar et al., 2007; Pialoux among children. Other serious non-neurological complications
et al., 2007; Ramful et al., 2007; Robin et al., 2008, 2009; have been reported in la Réunion and elsewhere, including acute
Schuffenecker et al., 2006). Classically, infected people adopt a hepatitis, bullous dermatosis, heart failure, myocarditis, pneumo-
characteristic stooped walking position which is the hallmark of nia, mild hemorrhage and pre-renal failure. Of critical note, these
the disease and from which it derives its name Chikungunya, severe cases were reported from hospitalized cohorts of patients
meaning ‘to walk bent over’, in the Kimakonde language of and may be related to several underlying medical conditions, most
Mozambique. The classic clinical symptoms after infection by commonly with hypertension, respiratory conditions and diabetes
CHIKV are abrupt febrile illness (temperature usually >38.9 8C) mellitus. In a recent study from India, age >85 years was the only
and maculopapular rash, with articular pains responsible for a risk factor for mortality although the possibility of co-infection by
stooped walk. Other symptoms include myalgia, headache, edema dengue virus (and other pathogens such as the respiratory
of the extremities, and gastrointestinal complaints (Borgherini syncytial or influenza viruses) may be a contributing factor to
et al., 2007; Lakshmi et al., 2008). The incubation period for CHIKV be considered (Sankari et al., 2008; Tandale et al., 2009).
124 T. Das et al. / Progress in Neurobiology 91 (2010) 121–129

2. Neurological manifestations of Chikungunya in newborns, the rate of transmission for viremic women was close to 50%
infants and adults highlighting the intrapartum period as the critical time for
transmission to the neonate (Gerardin et al., 2008; Ramful et al.,
CHIKV infection was first reported to affect the nervous system 2007). Cesarean sections had no protective effect on transmission.
in the 1960–1970s (Carey et al., 1969; Chastel, 1963; Chatterjee Dramatically, at least 50% of the infected neonates had pathologic
et al., 1965; Hammon et al., 1960; Jadhav et al., 1965; MRI findings (combination of white matter lesions and/or brain
Nimmannitya et al., 1969; Thiruvengadam et al., 1965). It was swelling and/or cerebral hemorrhages) evolving sometimes
found to be associated with seizures, meningoencephalopathy, towards persistent disabilities and deaths (Fig. 2) (Gerardin
myelitis, and choroiditis (Arpino et al., 2009). The same clinical et al., 2008; Ramful et al., 2007; Robin et al., 2008). In acute
hallmarks were reported in recent outbreaks but also included conditions, the supratentorial white matter lesions appeared
periventricular encephalitis, meningoencephalitis and deaths in essentially in the corpus callosum and the frontal, parietal and
neonates and adults hospitalized with severe CHIKV (Economo- temporal lobes. These lesions evolved into vasogenic edema and
poulou et al., 2009; Gerardin et al., 2008; Lemant et al., 2008; finally, in some cases, towards cavitations and subcortical
Lewthwaite et al., 2009; Ramful et al., 2007; Robin et al., 2008; athrophy. Persistent disabilities (10–20% of the infected neonates)
Tandale et al., 2009). after 1–2 years included seizures, cerebral palsy with ataxia and
Encephalopathy (associated with the detection of the virus in blindness, and ocular and behavioral or postural deficiencies
the CSF) appears to represent the most common neurologic (Gerardin et al., 2008).
manifestation among CHIKV-infected neonates after mother-to- The neurological signs were less severe in children hospitalized
child transmission. In la Reunion, the prevalence rate of maternal– following CHIKV infection but included cases of encephalitis,
fetal infections was 0.25%. In contrast, during the delivery period, febrile seizures, meningeal syndrome and acute encephalopathy.

Fig. 2. MRI findings in a neonate (12 days of life) with encephalopathy secondary to mother-to-child transmission of CHIKV infection on the 7th day after onset of disease. (a)
Diffusion weighted imaging: hyperintensity signals in the corpus callosum, subcortical and periventricular white matter, respecting the U-fibres. Reduction of the apparent
diffusion coefficient is compatible with cytotoxic edema. (b) T2-weighted imaging: petechial hemorrhages and hematomas (nodular lesions) with hyperintensities in the
subcortical and periventricular frontal and occipitoparietal white matter. (c) Gradient echo MRI: subtentorial hematoma of the left cerebellar hemisphere. (d) T1-weighed
imaging: hyperintensities in the subcortical and periventricular with matter corresponding to areas of hemorrhage. MRI abnormalities appear as white area.
T. Das et al. / Progress in Neurobiology 91 (2010) 121–129 125

The MRI scans were abnormal only in a limited number of cases IFN-a signaling pathway (KO for the IFNA receptor), CHIKV
(less than 20%) but deaths were reported together with long-term neuroinfection was particularly severe, targeting the leptome-
sequelae in 10–20% of the survivors (Robin et al., 2008). Deaths ninges, the choroid plexus and ependymal cells lining the
were observed in previously healthy children and probably due to subventricular zone (SVZ) also recognized as the neural stem cell
massive hemorrhages associated or not with brain swellings niche (Hauwel et al., 2005a,b). This area of the CNS seems to be
(Robin et al., 2008). Behavioral and communication disorders particularly targeted by several RNA viruses and with the key
(including autistic features) were reported in the follow-up of the observation that ependymal cells lining the SVZ express a plethora
infected children (Robin et al., 2008). of receptors (coxsackie adenovirus receptor (CAR), junction-
Neurological manifestations described in adults requiring associated molecules (JAM), complement regulators CD55 and
hospitalization (incidence of 15–25%) involved cases of encepha- CD46) for viruses such as coxsackie and adenovirus, reovirus,
lopathy frequently associated with the presence of IgM anti-CHIKV echovirus, and measles (Antar et al., 2009; Hotta et al., 2003;
in the CSF, encephalitis, Guillain Barre, encephalomyeloradiculitis Kemper and Atkinson, 2009; Lindahl et al., 2000). Is the SVZ really
and rare deaths (Chandak et al., 2009; Economopoulou et al., 2009; considered more immune-privileged then the rest of the brain? We
Ganesan et al., 2008; Lebrun et al., 2009; Lemant et al., 2008; hypothesize that CHIKV targets progenitor and stem cells in the
Tournebize et al., 2009; Wielanek et al., 2007). In the Ganesan’s SVZ as well as ependymal cells, possibly affecting neurogenesis
case studies it was found that CHIKV infection in adults was and neuronal migration which in turn could lead to demyelinating
associated to bilateral frontoparietal white matter lesions with encephalomyelitis, as recently reported in children and neonates
restricted diffusion, which is described as an early sign of viral infected by CHIKV (Gerardin et al., 2008; Ramful et al., 2007; Robin
encephalitis (Nouranifar et al., 2003). At autopsy, the brain was et al., 2008). CNS infections involving subventricular white matter
swollen and with subarachnoid cerebellar hemorrhages. Histolog- lesions and intraparenchymal hemorrhages have been described
ically, focal perivascular lymphocytic infiltrates were also present experimentally and in clinical settings for other alphaviruses such
in the area of active demyelination and some degree of microglial as SFV, RRV, EEV and SINV (Deresiewicz et al., 1997; Fazakerley
activation but no astrogliosis was also noted and which may et al., 2006; Jackson et al., 1987; Mims et al., 1973). Interestingly, it
contribute to bystander neuronal loss as described below. has recently been shown that flavivirus infection of the SVZ can
Unfortunately, no staining for CHIKV was performed in this study affect neurogenesis and have long-term effects with the depletion
(Ganesan et al., 2008). of the neural stem cell pool, but whether this is also the case for the
Although all the CHIKV-related neurological data are still alphavirus CHIKV remains to be ascertained (Das and Basu, 2008).
scarce, the number of cases with CNS involvement appears to The capacity of alphaviruses to interact with platelets/endothelial
support the neurotropic/neuroinfectious activity of CHIKV. cells and hence contribute to hemorrhagic complications is still a
matter of debate (Couderc et al., 2008; Dropulic and Masters, 1990;
3. CHIKV neuroinfection and possible pathological Larke and Wheelock, 1970; Scott et al., 1978).
mechanisms The cell receptor for CHIKV is unknown but CHIKV was shown
to have profound cytopathic effects in vivo and in vitro (Couderc
Historically (since 1965), CHIKV has been shown to infect et al., 2008; Ozden et al., 2007; Sourisseau et al., 2007); there is a
mouse brain and to replicate in primary culture of glial cells rapid shut-off of host cell transcription and translation followed by
(Chatterjee and Sarkar, 1965; Precious et al., 1974). Further but apoptotic death as assessed by the presence of numerous active
indirect evidence is supported by the observation that intracebral caspase-3 and CHIKV double-positive cells (Sourisseau et al.,
injection in newborn mice can be used to amplify the virus before 2007). These data were obtained using the HeLa epithelial cell
preparing the hyperimmune ascitic fluid used to date in most of the model and it remains to be ascertained whether this is also the case
CHIKV ELISA kit (Schuffenecker et al., 2006). More convincing in neuronal and glial cells. To begin to address some of these issues,
evidence comes from experimental infections where mice were we have infected mouse brain mixed cultures with low multiplicity
inoculated subcutaneously leading to rapid (24 h) and robust of infection (MOI) of CHIKV. Original data presented in Fig. 3 clearly
replication (106–107 PFU/ml) of CHIKV in the brain particularly of indicate that neurons (Tuj1 bIII tubulin+ cells) and astrocytes (glial
newborn outbred mice (CD1, ICR) (Ziegler et al., 2008). Interest- fibrillary acidic protein, GFAP+ cells) were extremely efficient at
ingly, infected mice showed signs of illness such as loss of balance, replicating CHIKV. The events leading to apoptosis remain to be
difficulty of walking, dragging of the hind limbs, and skin lesions fully determined although our data clearly indicate that both,
but rare mortality. Surprisingly, no definite histological evidence of intrinsic (mitochondrial) and extrinsic (Fas/TRAIL-like) apoptosis,
tropism to neurons was reported in these two mouse models and pathways are involved consequently to CHIKV infection (pathways
the CNS infection seemed to be tightly controlled by ill- illustrated in Fig. 4). The intrinsic apoptosis pathway is engaged by
characterized anti-viral mechanisms given that the viral titer cytochrome c released by the mitochondria to activate caspase 9
was reduced to basal levels at day 10 post-infection. In a more and sub sequentially caspase 3 (for review Clarke and Tyler, 2009;
recent review, Powers and Logue (2007) reported that CHIKV Griffin, 2005) (see illustration Fig. 4). In the IFNAR/ mouse
injected intranasally in BALB/c mice led to neuronal infection and model, it was shown that CHIKV induced severe vacuolization of
tissue necrosis in the anterior olfactory lobe. A very promising choroid plexus epithelial cells and ependymal cells, but failed to
neuroinfectious model reported recently by Weaver and collea- affect endothelial cells of the blood–brain barrier (Couderc et al.,
gues used intranasal injection of CHIKV Ross strain selected 2008). In the Ziegler study (Ziegler et al., 2008), glial cells and
because of its excessive mouse passage history, which may have neurons were rarely immunopositive for CHIKV antigens, but were
increased its virulence (Wang et al., 2008). Interestingly, 5-week- nevertheless found to be affected plausibly as a consequence of the
old C57BL/6 mice developed encephalitis 7 days post-infection robust anti-viral innate immune and inflammatory responses as
with severe multifocal infection and liquefactive necrosis in the described for many alphaviruses (Amor et al., 1996; Rowell and
cerebral cortex. Immunohistochemistry techniques revealed that Griffin, 2002; Schoneboom et al., 2000; Sharma et al., 2008; Vogel
neurons were infected and induced to programmed cell death et al., 2005). Gene profiling studies have clearly indicated that
while prominent microgliosis and perivascular cuffs were distrib- alphavirus CNS infection is accompanied by an upregulation of
uted throughout the parenchyma. Moreover, the authors reported multiple pro-inflammatory molecules including inducible nitric
neuronal degeneration in the hippocampus and multifocal oxide synthase (iNOS), tumor necrosis factor (TNF-a), and several
lymphocytic leptomeningitis. In inbred mice deficient in the cytokines and chemokines which ultimately would have direct or
126 T. Das et al. / Progress in Neurobiology 91 (2010) 121–129

Fig. 3. CHIKV infection and replication in neurons and astrocytes drive programmed cell death and innate immunity. Cells were either mock-infected or infected for 24–48 h
with CHIKV (MOI of 1). Cells were then double stained using antibodies against CHIKV (in green), markers of astrocytes (GFAP, red), of neurons (Tuj1, not shown), markers of
innate immunity (ISG15, in red), or markers of late apoptosis (cleaved PARP, in red). Of note, none of these antibodies stained mock-infected cells (not shown). (a) Cluster of
mouse embryonic neurons (E16–19) were strongly stained for CHIKV. (b) Primary astrocytes from 1- to 2-day-old mice were stained for CHIKV. CHIKV infection was also
evidenced in the CLTT mouse astrocyte cell line model stained for CHIKV (c), ISG15 (d), and cleaved PARP (e–f). Magnification 400.

indirect neurotoxic activities (Labrada et al., 2002; Sharma et al., clearance of the virus to preserve neuronal connections even if
2008). The innate immune and inflammatory cascade is an attempt this could promote latent viral infection (Binder and Griffin,
by resident cells to eliminate the challenge imposed on the brain, 2001). In this case, a robust and rapid innate immune response
clear the dead neurons, and rescue the normal functioning of the needs to be mobilized to limit viral replication in neuronal cells.
tissue. However, during the process of microglial/astroglial For instance, CNS viral infection rapidly results in the induction
activation, the proinflammatory molecules may exert certain of expression of the type-I interferons (IFNs) IFNa and IFNb, and
detrimental effects and neurons/neural stem cells are likely to be of pro-inflammatory cytokines by resident cells (for comprehen-
affected (Poluektova et al., 2005). sive review see Paul et al., 2007). Type-I IFNs are essential for the
anti-viral CHIKV response as demonstrated in vitro and in vivo
4. CNS innate immune protection against viral neuroinfection (Couderc et al., 2008; Gardner et al., 2008; Sourisseau et al.,
2007). Accumulating evidence indicates that several key cyto-
Viruses are the most abundant pathogens that the host innate plasmic cellular detection systems (RIG-I, MDA-5) are involved in
immune system is challenged by particularly when they find a innate immune sensing of RNA viruses (Bowie and Unterholzner,
privileged niche inside the CNS tissue (Griffin, 2003). Alpha- 2008; Takeuchi and Akira, 2007) to initiate the protective IFN
viruses like many other RNA viruses are known to persist in response (Fig. 4). RNA viruses such as CHIKV can also activate
tissue sanctuaries but our understanding of the mechanisms TLR-7 and TLR-8 localized into endosomes together with TLR3
involved is still in its infancy (Levine and Griffin, 1992; Levine and TLR9 (Fig. 4). This activation step is dependent on the
et al., 1994; Precious et al., 1974). In most tissues, the loss of acidification of the endosomes which is blocked by chloroquine
infected cells preferably by apoptosis is accompanied by (Lund et al., 2003, 2004), a drug that was tested for the treatment
enhanced cell division to replenish the cell population but this of CHIKV but, hence found unlikely to protect from infection
may not be achievable in tissues with little capacity for renewal, (Coombs et al., 1981; de Lamballerie et al., 2009; Maheshwari
such as the CNS. The balance would be to favor non-cytolytic et al., 1991; Seth et al., 1999).
T. Das et al. / Progress in Neurobiology 91 (2010) 121–129 127

Fig. 4. Innate immune sensing, apoptosis signaling pathways and anti-viral responses in the CNS. Several innate immune receptors also called ‘pattern-recognition receptors
(PRRs)’, such as RIG-I (retinoic-acid-inducible gene I), MDA5 (melanoma differentiation-associated gene 5) and TLR (Toll-like receptors) are expressed by neurons and glial
cells (microglia and astrocyte) to initiate signaling pathways that converge at the activation of the transcription factors interferon (IFN)-regulatory factor 3 (IRF3), IRF7 and/or
nuclear factor-kB (NFkB). These key events lead to the expression of type-I interferons (IFN-a/b). IFNs binding to the receptor (IFNAR) then initiate an anti-viral effector
program in the infected cell and neighboring cells, which involves the expression of numerous IFN-stimulated genes (ISGs) and pro-inflammatory cytokines and chemokines.
ISGs such as oligoadenylate synthetase (OAS) and ribonuclease L (RNaseL), IFN-inducible dsRNA-dependent protein kinase (PKR), myxovirus resistance (Mx) protein, are
involved in anti-viral mechanisms that interfere with the life cycle of individual viruses. OAS and PKR are further activated by double-stranded RNA (dsRNA). IFNAR1,
interferon-a receptor; IPS1, IFNB promoter stimulator 1; ISG15, IFN-stimulated gene of 15 kDa; PPP, 5 triphosphate; ssRNA, single-stranded RNA; STAT, signal transducer and
activator of transcription. The innate immune signaling pathways are intimately linked to apoptosis through the mobilization of TRADD, RIP1 and FADD. The intrinsic
apoptosis pathway is initiated by the release of cytochrome C from the mitochondria which promotes the formation of the apoptosome, including APAF-1 (apoptotic protease
activating factor) and caspase 9 which in turn activates the effector caspase 3. The extrinsic pathway mobilized notably by TNF-a, TRAIL or FASL involves death-receptor
signaling pathways linked to FADD and caspase 8. Some viruses such as CHIKV (our unpublished data) can escape apoptosis by inducing the expression of anti-apoptotic
proteins such as IAP and Bcl2.

These innate immune receptors are the first line of defense need to be further established (Brehin et al., 2009). Of preliminary
against pathogens infiltrating the CNS and they are called pattern- note, we found a robust expression of ISG15 by CLTT astrocyte cell
recognition receptors (PRRs) for their capacity to recognize a line in response to CHIKV infection (see Fig. 3). This astrocyte
plethora of pathogens and to drive signaling pathways to promote model will be invaluable to decipher the molecular signaling
anti-viral responses. These PRRs use diverse adaptor proteins and pathways engaged by one or many PRR to drive anti-CHIKV
signaling pathways resulting in activation of a distinct set of responses in the CNS.
transcription factors and include NFkB (p50/p65 subunits) and
IFN-regulatory factors (IRFs). NFkB and IRFs translocation into the 5. Conclusion: exploring novel anti-viral therapies to fight
nucleus will lead to the transcription of pro-inflammatory against CHIKV infection
mediators and survival factors, including IL-6, TNF-a, IL-12, IL-8
and IFN-g. RIG-I and MDA-5 activate IPS-1 (IFN-b promoter A new and comprehensive description of CHIKV-induced
stimulator-1, also known as MAVS, CARDIF and VISA), a protein neuroinfection and pathology has been gained from recent
located on the surface of mitochondria. Downstream this would outbreaks in countries with efficient clinical facilities, including
result in translocation of the transcription factors IRF-3 and NFkB MRI scanning. Severe encephalitis and deaths are clear testimo-
but could also lead to apoptosis through the recruitment of the nies of the severity of the disease particularly following mother-
(TRADD, tumour-necrosis factor receptor-associated via death to-child transmission and in elderly patients with cardiovascular
domain; RIP1, receptor-interacting protein 1 and FADD, Fas- diseases or diabetes. The plausible role of mutated forms of
associated death domain) complex linked to caspase 8 (Fig. 4) CHIKV with high neuroinfection fitness needs to be more firmly
(Balachandran et al., 2000; Michallet et al., 2008; Takahashi et al., addressed, making use of the several mouse models reproducing
2006; Wilson et al., 2009). The precise role of these different the disease and cDNA CHIKV infectious clones (Powers and
adaptor proteins in the signalling cascades to promote cytokine Logue, 2007). From a clinical standpoint it is critical that
production and/or apoptosis following CHIKV infection remains to physicians are educated in recognizing acute cases and that
be ascertained. Recent data indicate that nsP2 of an alphavirus is an diagnostic tools are available to perform discriminative screen-
antagonist of the type-I interferon response (Breakwell et al., 2007) ings from other tropical diseases. Interestingly, several cases of
and CHIKV may control innate immunity in a similar manner. CNS infection have been associated with co-infection (mainly
Coordinated PRR amplification involves IFN-a/b dependent path- Dengue), which may explain some of the severe cases in recent
ways with the translocation of phosphorylated STAT1 and STAT2 outbreaks. Further research is warranted to have a better
molecules into the nucleus to drive the expression of several understanding of CHIKV-induced pathology and the long-term
interferon-stimulated genes (ISGs) and other anti-viral proteins. sequelae particularly in children. Arboviruses have long been
Factors directly affecting viral replication are PKR, OAS, RNaseL, known to persist in infected tissues, driving chronic pathologies
and ISG15, ISG54 and ISG56, which all modulate translation; Mx (Kuno, 2001).
which inhibits viral transcription; the RNA deaminases ADAR-1 The restriction of CHIKV diseases is undoubtedly through
and APOBEC3G, leading to mutations in viral genomes. IFN-a/b prevention measures such as vaccination. A live, attenuated
responsiveness in CNS cells is indicated by induction of various vaccine was originally advanced through phase II clinical trials,
ISGs, including RNaseL, ISG54 and ISG56, Mx, IRF-7 and IRF-9 but its development was halted because of its poor tolerance
following distinct infections (for review Paul et al., 2007). (Edelman et al., 2000). Ongoing studies will help to delineate the
Interestingly, OAS has recently been shown to be active against immunogenicity of the different CHIKV proteins and to design a
CHIKV replication in HeLa cells and its role in neuronal cells will safer and more potent vaccination regime to protect from future
128 T. Das et al. / Progress in Neurobiology 91 (2010) 121–129

outbreaks and life-threatening infection (Muthumani et al., 2008; de Lamballerie, X., Ninove, L., Charrel, R.N., 2009. Antiviral treatment of Chikungu-
nya virus infection. Infect. Disord. Drug Targets 9, 101–104.
Wang et al., 2008). Deresiewicz, R.L., Thaler, S.J., Hsu, L., Zamani, A.A., 1997. Clinical and neuroradio-
graphic manifestations of eastern equine encephalitis. N. Engl. J. Med. 336,
1867–1874.
Acknowledgements Dropulic, B., Masters, C.L., 1990. Entry of neurotropic arboviruses into the central
nervous system: an in vitro study using mouse brain endothelium. J. Infect. Dis.
161, 685–691.
The authors’ work is funded by the University of La Reunion,
Economopoulou, A., Dominguez, M., Helynck, B., Sissoko, D., Wichmann, O., Quenel,
French Ministry of Health (PRHC 2006–2009), French overseas P., Germonneau, P., Quatresous, I., 2009. Atypical Chikungunya virus infections:
Ministry (MOM), the CRVOI (centre de recherche et de veille de clinical manifestations, mortality and risk factors for severe disease during the
l’Océan Indien), the Regional council of La Réunion and Europe 2005–2006 outbreak on Reunion. Epidemiol. Infect. 137, 534–541.
Edelman, R., Tacket, C.O., Wasserman, S.S., Bodison, S.A., Perry, J.G., Mangiafico, J.A.,
(CPER/FEDER/GRII). PG is a fellow of INSERM ‘contrat d’interface’ 2000. Phase II safety and immunogenicity study of live Chikungunya virus
with Pr. P. Debre and Pr. B. Autran (INSERM U945, Salpétrière, vaccine TSI-GSD-218. Am. J. Trop. Med. Hyg. 62, 681–685.
Paris). The authors thank Mrs Claudine Vitry for help in editorial’s Fazakerley, J.K., Cotterill, C.L., Lee, G., Graham, A., 2006. Virus tropism, distribution,
persistence and pathology in the corpus callosum of the Semliki Forest virus-
management of the paper and figure. infected mouse brain: a novel system to study virus-oligodendrocyte interac-
tions. Neuropathol. Appl. Neurobiol. 32, 397–409.
Fourie, E.D., Morrison, J.G., 1979. Rheumatoid arthritic syndrome after Chikungunya
References fever. S. Afr. Med. J. 56, 130–132.
Ganesan, K., Diwan, A., Shankar, S.K., Desai, S.B., Sainani, G.S., Katrak, S.M., 2008.
Amor, S., Scallan, M.F., Morris, M.M., Dyson, H., Fazakerley, J.K., 1996. Role of Chikungunya encephalomyeloradiculitis: report of 2 cases with neuroimaging
immune responses in protection and pathogenesis during Semliki Forest virus and 1 case with autopsy findings. Am. J. Neuroradiol. 29, 1636–1637.
encephalitis. J. Gen. Virol. 77 (Pt 2), 281–291. Gardner, C.L., Burke, C.W., Tesfay, M.Z., Glass, P.J., Klimstra, W.B., Ryman, K.D., 2008.
Antar, A.A., Konopka, J.L., Campbell, J.A., Henry, R.A., Perdigoto, A.L., Carter, B.D., Eastern and Venezuelan equine encephalitis viruses differ in their ability to
Pozzi, A., Abel, T.W., Dermody, T.S., 2009. Junctional adhesion molecule-A is infect dendritic cells and macrophages: impact of altered cell tropism on
required for hematogenous dissemination of reovirus. Cell Host Microbe 5, 59– pathogenesis. J. Virol. 82, 10634–10646.
71. Gerardin, P., Barau, G., Michault, A., Bintner, M., Randrianaivo, H., Choker, G.,
Arpino, C., Curatolo, P., Rezza, G., 2009. Chikungunya and the nervous system: what Lenglet, Y., Touret, Y., Bouveret, A., Grivard, P., Le Roux, K., Blanc, S., Schuffe-
we do and do not know. Rev. Med. Virol. 19, 121–129. necker, I., Couderc, T., Arenzana-Seisdedos, F., Lecuit, M., Robillard, P.Y., 2008.
Balachandran, S., Roberts, P.C., Kipperman, T., Bhalla, K.N., Compans, R.W., Archer, Multidisciplinary prospective study of mother-to-child Chikungunya virus
D.R., Barber, G.N., 2000. Alpha/beta interferons potentiate virus-induced apo- infections on the island of La Reunion. PLoS Med. 5, e60.
ptosis through activation of the FADD/Caspase-8 death signaling pathway. J. Griffin, D.E., 2003. Immune responses to RNA-virus infections of the CNS. Nat. Rev.
Virol. 74, 1513–1523. Immunol. 3, 493–502.
Binder, G.K., Griffin, D.E., 2001. Interferon-gamma-mediated site-specific clearance Griffin, D.E., 2005. Neuronal cell death in alphavirus encephalomyelitis. Curr. Top.
of alphavirus from CNS neurons. Science 293, 303–306. Microbiol. Immunol. 289, 57–77.
Borgherini, G., Poubeau, P., Jossaume, A., Gouix, A., Cotte, L., Michault, A., Arvin- Hammon, W.M., Rudnick, A., Sather, G.E., 1960. Viruses associated with epidemic
Berod, C., Paganin, F., 2008. Persistent arthralgia associated with Chikungunya hemorrhagic fevers of the Philippines and Thailand. Science 131, 1102–1103.
virus: a study of 88 adult patients on reunion island. Clin. Infect. Dis. 47, 469– Harley, D., Sleigh, A., Ritchie, S., 2001. Ross River virus transmission, infection, and
475. disease: a cross-disciplinary review. Clin. Microbiol. Rev. 14, 909–932 table of
Borgherini, G., Poubeau, P., Staikowsky, F., Lory, M., Le Moullec, N., Becquart, J.P., contents.
Wengling, C., Michault, A., Paganin, F., 2007. Outbreak of Chikungunya on Hauwel, M., Furon, E., Canova, C., Griffiths, M., Neal, J., Gasque, P., 2005a. Innate
Reunion Island: early clinical and laboratory features in 157 adult patients. (inherent) control of brain infection, brain inflammation and brain repair: the
Clin. Infect. Dis. 44, 1401–1407. role of microglia, astrocytes, ‘‘protective’’ glial stem cells and stromal ependy-
Bowie, A.G., Unterholzner, L., 2008. Viral evasion and subversion of pattern-recog- mal cells. Brain Res. Brain Res. Rev. 48, 220–233.
nition receptor signalling. Nat. Rev. Immunol. 8, 911–922. Hauwel, M., Furon, E., Gasque, P., 2005b. Molecular and cellular insights into the
Breakwell, L., Dosenovic, P., Karlsson Hedestam, G.B., D’Amato, M., Liljestrom, P., coxsackie-adenovirus receptor: role in cellular interactions in the stem cell
Fazakerley, J., McInerney, G.M., 2007. Semliki Forest virus nonstructural protein niche. Brain Res. Brain Res. Rev. 48, 265–272.
2 is involved in suppression of the type I interferon response. J. Virol. 81, 8677– Hotta, Y., Honda, T., Naito, M., Kuwano, R., 2003. Developmental distribution of
8684. coxsackie virus and adenovirus receptor localized in the nervous system. Brain
Brehin, A.C., Casademont, I., Frenkiel, M.P., Julier, C., Sakuntabhai, A., Despres, P., Res. Dev. Brain Res. 143, 1–13.
2009. The large form of human 20 ,50 -oligoadenylate synthetase (OAS3) exerts Jackson, A.C., Moench, T.R., Griffin, D.E., Johnson, R.T., 1987. The pathogenesis of
antiviral effect against Chikungunya virus. Virology 384, 216–222. spinal cord involvement in the encephalomyelitis of mice caused by neuroa-
Brighton, S.W., Simson, I.W., 1984. A destructive arthropathy following Chikungu- dapted Sindbis virus infection. Lab Invest. 56, 418–423.
nya virus arthritis—a possible association. Clin. Rheumatol. 3, 253–258. Jadhav, M., Namboodripad, M., Carman, R.H., Carey, D.E., Myers, R.M., 1965. Chi-
Carey, D.E., Myers, R.M., DeRanitz, C.M., Jadhav, M., Reuben, R., 1969. The 1964 kungunya disease in infants and children in Vellore: a report of clinical and
Chikungunya epidemic at Vellore, South India, including observations on haematological features of virologically proved cases. Indian J. Med. Res. 53,
concurrent dengue. Trans. R. Soc. Trop. Med. Hyg. 63, 434–445. 764–776.
Chandak, N.H., Kashyap, R.S., Kabra, D., Karandikar, P., Saha, S.S., Morey, S.H., Kemper, C., Atkinson, J.P., 2009. Measles virus and CD46. Curr. Top. Microbiol.
Purohit, H.J., Taori, G.M., Daginawala, H.F., 2009. Neurological complications Immunol. 329, 31–57.
of Chikungunya virus infection. Neurol. India 57, 177–180. Kuno, G., 2001. Persistence of arboviruses and antiviral antibodies in vertebrate
Charrel, R.N., de Lamballerie, X., Raoult, D., 2007. Chikungunya outbreaks—the hosts: its occurrence and impacts. Rev. Med. Virol. 11, 165–190.
globalization of vectorborne diseases. N. Engl. J. Med. 356, 769–771. Labrada, L., Liang, X.H., Zheng, W., Johnston, C., Levine, B., 2002. Age-dependent
Chastel, C., 1963. Human infections in Cambodia by the Chikungunya virus or an resistance to lethal alphavirus encephalitis in mice: analysis of gene expression
apparently closely related agent. II. Experimental pathological anatomy. Bull. in the central nervous system and identification of a novel interferon-inducible
Soc. Pathol. Exot. Filiales 56, 915–924. protective gene, mouse ISG12. J. Virol. 76, 11688–11703.
Chatterjee, S.N., Chakravarti, S.K., Mitra, A.C., Sarkar, J.K., 1965. Virological investi- Lakshmi, V., Neeraja, M., Subbalaxmi, M.V., Parida, M.M., Dash, P.K., Santhosh, S.R.,
gation of cases with neurological complications during the outbreak of hae- Rao, P.V., 2008. Clinical features and molecular diagnosis of Chikungunya fever
morrhagic fever in Calcutta. J. Indian Med. Assoc. 45, 314–316. from South India. Clin. Infect. Dis. 46, 1436–1442.
Chatterjee, S.N., Sarkar, J.K., 1965. Electron microscopic studies of suckling mouse Lanciotti, R.S., Kosoy, O.L., Laven, J.J., Panella, A.J., Velez, J.O., Lambert, A.J., Campbell,
brain cells infected with Chikungunya virus. Indian J. Exp. Biol. 3, 227–234. G.L., 2007. Chikungunya virus in US travelers returning from India, 2006. Emerg.
Chevillon, C., Briant, L., Renaud, F., Devaux, C., 2008. The Chikungunya threat: an Infect. Dis. 13, 764–767.
ecological and evolutionary perspective. Trends Microbiol. 16, 80–88. Larke, R.P., Wheelock, E.F., 1970. Stabilization of Chikungunya virus infectivity by
Clarke, P., Tyler, K.L., 2009. Apoptosis in animal models of virus-induced disease. human blood platelets. J. Infect. Dis. 122, 523–531.
Nat. Rev. Microbiol. 7, 144–155. Lebrun, G., Chadda, K., Reboux, A.H., Martinet, O., Gauzere, B.A., 2009. Guillain-Barre
Coombs, K., Mann, E., Edwards, J., Brown, D.T., 1981. Effects of chloroquine and syndrome after Chikungunya infection. Emerg. Infect. Dis. 15, 495–496.
cytochalasin B on the infection of cells by Sindbis virus and vesicular stomatitis Lemant, J., Boisson, V., Winer, A., Thibault, L., Andre, H., Tixier, F., Lemercier, M.,
virus. J. Virol. 37, 1060–1065. Antok, E., Cresta, M.P., Grivard, P., Besnard, M., Rollot, O., Favier, F., Huerre, M.,
Couderc, T., Chretien, F., Schilte, C., Disson, O., Brigitte, M., Guivel-Benhassine, F., Campinos, J.L., Michault, A., 2008. Serious acute Chikungunya virus infection
Touret, Y., Barau, G., Cayet, N., Schuffenecker, I., Despres, P., Arenzana-Seisde- requiring intensive care during the Reunion Island outbreak in 2005–2006. Crit.
dos, F., Michault, A., Albert, M.L., Lecuit, M., 2008. A mouse model for Chikun- Care Med. 36, 2536–2541.
gunya: young age and inefficient type-I interferon signaling are risk factors for Levine, B., Griffin, D.E., 1992. Persistence of viral RNA in mouse brains after recovery
severe disease. PLoS Pathog. 4, e29. from acute alphavirus encephalitis. J. Virol. 66, 6429–6435.
Das, S., Basu, A., 2008. Japanese encephalitis virus infects neural progenitor cells and Levine, B., Hardwick, J.M., Griffin, D.E., 1994. Persistence of alphaviruses in verte-
decreases their proliferation. J. Neurochem. 106, 1624–1636. brate hosts. Trends Microbiol. 2, 25–28.
T. Das et al. / Progress in Neurobiology 91 (2010) 121–129 129

Lewthwaite, P., Vasanthapuram, R., Osborne, J.C., Begum, A., Plank, J.L., Shankar, Duquerroy, S., Guigon, G., Frenkiel, M.P., Brehin, A.C., Cubito, N., Despres, P.,
M.V., Hewson, R., Desai, A., Beeching, N.J., Ravikumar, R., Solomon, T., 2009. Kunst, F., Rey, F.A., Zeller, H., Brisse, S., 2006. Genome microevolution of
Chikungunya virus and central nervous system infections in children, India. Chikungunya viruses causing the Indian Ocean outbreak. PLoS Med. 3, e263.
Emerg. Infect. Dis. 15, 329–331. Scott, S., Reimers, H.J., Chernesky, M.A., Greenberg, J.P., Kinolugh-Rathbone, R.L.,
Lindahl, G., Sjobring, U., Johnsson, E., 2000. Human complement regulators: a major Packham, M.A., Mustard, J.F., 1978. Effect of viruses on platelet aggregation and
target for pathogenic microorganisms. Curr. Opin. Immunol. 12, 44–51. platelet survival in rabbits. Blood 52, 47–55.
Lund, J., Sato, A., Akira, S., Medzhitov, R., Iwasaki, A., 2003. Toll-like receptor 9- Seth, P., Mani, H., Singh, A.K., Banaudha, K.K., Madhavan, S., Sidhu, G.S., Gaddipati,
mediated recognition of Herpes simplex virus-2 by plasmacytoid dendritic J.P., Vogel, S.N., Maheshwari, R.K., 1999. Acceleration of viral replication and up-
cells. J. Exp. Med. 198, 513–520. regulation of cytokine levels by antimalarials: implications in malaria-endemic
Lund, J.M., Alexopoulou, L., Sato, A., Karow, M., Adams, N.C., Gale, N.W., Iwasaki, A., areas. Am. J. Trop. Med. Hyg. 61, 180–186.
Flavell, R.A., 2004. Recognition of single-stranded RNA viruses by Toll-like Sharma, A., Bhattacharya, B., Puri, R.K., Maheshwari, R.K., 2008. Venezuelan equine
receptor 7. Proc. Natl. Acad. Sci. U.S.A. 101, 5598–5603. encephalitis virus infection causes modulation of inflammatory and immune
Maheshwari, R.K., Srikantan, V., Bhartiya, D., 1991. Chloroquine enhances replica- response genes in mouse brain. BMC Genom. 9, 289.
tion of Semliki Forest virus and encephalomyocarditis virus in mice. J. Virol. 65, Simon, F., Parola, P., Grandadam, M., Fourcade, S., Oliver, M., Brouqui, P., Hance, P.,
992–995. Kraemer, P., Ali Mohamed, A., de Lamballerie, X., Charrel, R., Tolou, H., 2007.
Mavalankar, D., Shastri, P., Raman, P., 2007. Chikungunya epidemic in India: a major Chikungunya infection: an emerging rheumatism among travelers returned
public-health disaster. Lancet Infect. Dis. 7, 306–307. from Indian Ocean islands. Report of 47 cases. Medicine (Baltimore) 86, 123–
Michallet, M.C., Meylan, E., Ermolaeva, M.A., Vazquez, J., Rebsamen, M., Curran, J., 137.
Poeck, H., Bscheider, M., Hartmann, G., Konig, M., Kalinke, U., Pasparakis, M., Sissoko, D., Malvy, D., Ezzedine, K., Renault, P., Moscetti, F., Ledrans, M., Pierre, V.,
Tschopp, J., 2008. TRADD protein is an essential component of the RIG-like 2009. Post-epidemic Chikungunya disease on Reunion Island: course of rheu-
helicase antiviral pathway. Immunity 28, 651–661. matic manifestations and associated factors over a 15-month period. PLoS Negl.
Mims, C.A., Murphy, F.A., Taylor, W.P., Marshall, I.D., 1973. Pathogenesis of Ross Trop. Dis. 3, e389.
River virus infection in mice. I. Ependymal infection, cortical thinning, and Sourisseau, M., Schilte, C., Casartelli, N., Trouillet, C., Guivel-Benhassine, F., Rud-
hydrocephalus. J. Infect. Dis. 127, 121–128. nicka, D., Sol-Foulon, N., Le Roux, K., Prevost, M.C., Fsihi, H., Frenkiel, M.P.,
Muthumani, K., Lankaraman, K.M., Laddy, D.J., Sundaram, S.G., Chung, C.W., Sako, E., Blanchet, F., Afonso, P.V., Ceccaldi, P.E., Ozden, S., Gessain, A., Schuffenecker, I.,
Wu, L., Khan, A., Sardesai, N., Kim, J.J., Vijayachari, P., Weiner, D.B., 2008. Verhasselt, B., Zamborlini, A., Saib, A., Rey, F.A., Arenzana-Seisdedos, F., Despres,
Immunogenicity of novel consensus-based DNA vaccines against Chikungunya P., Michault, A., Albert, M.L., Schwartz, O., 2007. Characterization of reemerging
virus. Vaccine 26, 5128–5134. Chikungunya virus. PLoS Pathog. 3, e89.
Nimmannitya, S., Halstead, S.B., Cohen, S.N., Margiotta, M.R., 1969. Dengue and Suhrbier, A., La Linn, M., 2004. Clinical and pathologic aspects of arthritis due to
Chikungunya virus infection in man in Thailand, 1962–1964. I. Observations on Ross River virus and other alphaviruses. Curr. Opin. Rheumatol. 16, 374–
hospitalized patients with hemorrhagic fever. Am. J. Trop. Med. Hyg. 18, 954– 379.
971. Takahashi, K., Kawai, T., Kumar, H., Sato, S., Yonehara, S., Akira, S., 2006. Roles of
Nouranifar, R.K., Ali, M., Nath, J., 2003. The earliest manifestation of focal encepha- caspase-8 and caspase-10 in innate immune responses to double-stranded
litis on diffusion-weighted MRI. Clin. Imaging 27, 316–320. RNA. J. Immunol. 176, 4520–4524.
Ozden, S., Huerre, M., Riviere, J.P., Coffey, L.L., Afonso, P.V., Mouly, V., de Monredon, Takeuchi, O., Akira, S., 2007. Recognition of viruses by innate immunity. Immunol.
J., Roger, J.C., El Amrani, M., Yvin, J.L., Jaffar, M.C., Frenkiel, M.P., Sourisseau, M., Rev. 220, 214–224.
Schwartz, O., Butler-Browne, G., Despres, P., Gessain, A., Ceccaldi, P.E., 2007. Tandale, B.V., Sathe, P.S., Arankalle, V.A., Wadia, R.S., Kulkarni, R., Shah, S.V., Shah,
Human muscle satellite cells as targets of Chikungunya virus infection. PLoS S.K., Sheth, J.K., Sudeep, A.B., Tripathy, A.S., Mishra, A.C., 2009. Systemic invol-
ONE 2, e527. vements and fatalities during Chikungunya epidemic in India, 2006. J. Clin.
Panning, M., Grywna, K., van Esbroeck, M., Emmerich, P., Drosten, C., 2008. Chi- Virol. 46, 145–149.
kungunya fever in travelers returning to Europe from the Indian Ocean region, Tesh, R.B., 1982. Arthritides caused by mosquito-borne viruses. Annu. Rev. Med. 33,
2006. Emerg. Infect. Dis. 14, 416–422. 31–40.
Paul, S., Ricour, C., Sommereyns, C., Sorgeloos, F., Michiels, T., 2007. Type I interferon Thiruvengadam, K.V., Kalyanasundaram, V., Rajgopal, J., 1965. Clinical and patho-
response in the central nervous system. Biochimie 89, 770–778. logical studies on Chikungunya fever in Madras city. Indian J. Med. Res. 53, 729–
Pialoux, G., Gauzere, B.A., Jaureguiberry, S., Strobel, M., 2007. Chikungunya, an 744.
epidemic arbovirosis. Lancet Infect. Dis. 7, 319–327. Toivanen, A., 2008. Alphaviruses: an emerging cause of arthritis? Curr. Opin.
Poluektova, L., Meyer, V., Walters, L., Paez, X., Gendelman, H.E., 2005. Macrophage- Rheumatol. 20, 486–490.
induced inflammation affects hippocampal plasticity and neuronal develop- Tournebize, P., Charlin, C., Lagrange, M., 2009. Neurological manifestations in
ment in a murine model of HIV-1 encephalitis. Glia 52, 344–353. Chikungunya: about 23 cases collected in Reunion Island. Rev. Neurol. (Paris)
Powers, A.M., Logue, C.H., 2007. Changing patterns of Chikungunya virus: re- 165, 48–51.
emergence of a zoonotic arbovirus. J. Gen. Virol. 88, 2363–2377. Tsetsarkin, K.A., Vanlandingham, D.L., McGee, C.E., Higgs, S., 2007. A single mutation
Precious, S.W., Webb, H.E., Bowen, E.T., 1974. Isolation and persistence of Chikun- in Chikungunya virus affects vector specificity and epidemic potential. PLoS
gunya virus in cultures of mouse brain cells. J. Gen. Virol. 23, 271–279. Pathog. 3, e201.
Ramful, D., Carbonnier, M., Pasquet, M., Bouhmani, B., Ghazouani, J., Noormahomed, Vashishtha, M., Phalen, T., Marquardt, M.T., Ryu, J.S., Ng, A.C., Kielian, M., 1998. A
T., Beullier, G., Attali, T., Samperiz, S., Fourmaintraux, A., Alessandri, J.L., 2007. single point mutation controls the cholesterol dependence of Semliki Forest
Mother-to-child transmission of Chikungunya virus infection. Pediatr. Infect. virus entry and exit. J. Cell Biol. 140, 91–99.
Dis. J. 26, 811–815. Vogel, P., Kell, W.M., Fritz, D.L., Parker, M.D., Schoepp, R.J., 2005. Early events in the
Robin, S., Ramful, D., Le Seach, F., Jaffar-Bandjee, M.C., Rigou, G., Alessandri, J.L., pathogenesis of eastern equine encephalitis virus in mice. Am. J. Pathol. 166,
2008. Neurologic manifestations of pediatric Chikungunya infection. J. Child. 159–171.
Neurol. 23, 1028–1035. Wang, E., Volkova, E., Adams, A.P., Forrester, N., Xiao, S.Y., Frolov, I., Weaver, S.C.,
Robin, S., Ramful, D., Zettor, J., Benhamou, L., Jaffar-Bandjee, M.C., Riviere, J.P., 2008. Chimeric alphavirus vaccine candidates for Chikungunya. Vaccine 26,
Marichy, J., Ezzedine, K., Alessandri, J.L., 2009. Severe bullous skin lesions 5030–5039.
associated with Chikungunya virus infection in small infants. Eur. J. Pediatr. Weaver, S.C., Barrett, A.D., 2004. Transmission cycles, host range, evolution and
169, 67–72. emergence of arboviral disease. Nat. Rev. Microbiol. 2, 789–801.
Rowell, J.F., Griffin, D.E., 2002. Contribution of T cells to mortality in neurovirulent Wielanek, A.C., de Monredon, J., El Amrani, M., Roger, J.C., Serveaux, J.P., 2007.
Sindbis virus encephalomyelitis. J. Neuroimmunol. 127, 106–114. Guillain-Barre syndrome complicating a Chikungunya virus infection. Neurol-
Sankari, T., Hoti, S.L., Govindaraj, V., Das, P.K., 2008. Chikungunya and respiratory ogy 69, 2105–2107.
viral infections. Lancet Infect. Dis. 8, 3–4. Wilson, N.S., Dixit, V., Ashkenazi, A., 2009. Death receptor signal transducers:
Schoneboom, B.A., Catlin, K.M., Marty, A.M., Grieder, F.B., 2000. Inflammation is a nodes of coordination in immune signaling networks. Nat. Immunol. 10,
component of neurodegeneration in response to Venezuelan equine encepha- 348–355.
litis virus infection in mice. J. Neuroimmunol. 109, 132–146. Ziegler, S.A., Lu, L., da Rosa, A.P., Xiao, S.Y., Tesh, R.B., 2008. An animal model for
Schuffenecker, I., Iteman, I., Michault, A., Murri, S., Frangeul, L., Vaney, M.C., Lavenir, studying the pathogenesis of Chikungunya virus infection. Am. J. Trop. Med.
R., Pardigon, N., Reynes, J.M., Pettinelli, F., Biscornet, L., Diancourt, L., Michel, S., Hyg. 79, 133–139.

View publication stats

You might also like