ArticlePDF AvailableLiterature Review

The global emergence of Chikungunya infection: An integrated view

Authors:
  • GPGC Mandian Abbottabad

Abstract

Chikungunya virus (CHIKV) is one of the emerging viruses around the globe. It belongs to the family Togaviridae and genus Alphavirus and is an arthropod borne virus that transmits by the bite of an infected mosquito, mainly through Aedes aegypti and Aedes albopcitus. It is a spherical, enveloped virus with positive single stranded RNA genome. It was first discovered during 1952-53 in Tanganyika, after which outbreaks were documented in many regions of the world. CHIKV has two transmission cycles; an enzootic sylvatic cycle and an urban cycle. CHIKV genome contains 11,900 nucleotides and two open reading frames and shows great sequence variability. Molecular mechanisms of virus host-cell interactions and the pathogenesis of disease are not fully understood. The disease involves three phases; acute, post-acute and chronic with symptoms including high-grade fever, arthralgia, macupapular rashes and headache. There is no licensed vaccine or specific treatment for CHIKV infection. This lack of specific interventions combined with difficulties in making a precise diagnosis together make the disease difficult to manage. In this review we aim to present the current knowledge of global epidemiology, transmission, structure, various aspects of diagnosis as well as highlight potential antiviral drugs and vaccines against CHIKV.
Received: 19 May 2021
-
Revised: 8 August 2021
-
Accepted: 9 August 2021
DOI: 10.1002/rmv.2287
REVIEW
The global emergence of Chikungunya infection: An
integrated view
Khanzadi Nazneen Manzoor
1
|Farakh Javed
2
|Muhammad Ejaz
3
|
Mubashar Ali
3
|Neelam Mujaddadi
3
|Abid Ali Khan
4
|Aamer Ali Khattak
5
|
Assad Zaib
5
|Ibrar Ahmad
6
|Waqar Khalid Saeed
2
|Sobia Manzoor
7
1
Department of Biosciences, COMSATS
institute of Information Technology,
Islamabad, Pakistan
2
Department of Biomedical Sciences, Pak
Autria Fachhochschule: Institute of Applied
Sciences & Technology, Haripur, Pakistan
3
Department of Microbiology, The University
of Haripur, Haripur, Pakistan
4
Institute of Precision Medicine, Hochschule
Furtwangen University, Furtwangen im
Schwarzwald, Germany
5
Department of Medical Lab Technology, The
University of Haripur, Haripur, Pakistan
6
Center for Human Genetics, Hazara
University, Mansehra, Pakistan
7
AttaurRehman school of applied
biosciences, National University of science and
Technology, Islamabad, Pakistan
Correspondence
Farakh Javed, Department of Biomedical
Sciences, PakAutria Fachhochschule:
Institute of Applied Sciences & Technology,
Mang, Haripur, Pakistan.
Email: Farakh.javed@fbse.paf-iast.edu.pk and
farukhbbt@gmail.com
Summary
Chikungunya virus (CHIKV) is one of the emerging viruses around the globe. It
belongs to the family Togaviridae and genus Alphavirus and is an arthropod borne
virus that transmits by the bite of an infected mosquito, mainly through Aedes
aegypti and Aedes albopcitus. It is a spherical, enveloped virus with positive single
stranded RNA genome. It was first discovered during 195253 in Tanganyika, after
which outbreaks were documented in many regions of the world. CHIKV has two
transmission cycles; an enzootic sylvatic cycle and an urban cycle. CHIKV genome
contains 11,900 nucleotides and two open reading frames and shows great
sequence variability. Molecular mechanisms of virus hostcell interactions and the
pathogenesis of disease are not fully understood. The disease involves three phases;
acute, postacute and chronic with symptoms including highgrade fever, arthralgia,
macupapular rashes and headache. There is no licensed vaccine or specific treat-
ment for CHIKV infection. This lack of specific interventions combined with diffi-
culties in making a precise diagnosis together make the disease difficult to manage.
In this review we aim to present the current knowledge of global epidemiology,
transmission, structure, various aspects of diagnosis as well as highlight potential
antiviral drugs and vaccines against CHIKV.
KEYWORDS
Aedes aegypti,Aedes albopictus, alpha virus, Chikungunya virus
1
|
INTRODUCTION
Chikungunya virus (CHIKV) is an arthropod borne virus, transmitted
via the bite of infected mosquito. It was first discovered in epidemic
of dengue like fever in 1952–1953 in Tanganyika (now called
Tanzania) and was isolated from the serum of the patient in acute
febrile phase of illness.
1–3
The word Chikungunya derived from local
Kimakonde or Kiswahili language spoken in some regions of Tanzania
and Mozambique that means ‘that which bends up’, refers to the post
symptoms of Chikungunya viral infection.
4
CHIKV outbreaks
occurred in many regions but its large epidemic reemerged in La
Reunion island of Indian Ocean in early 2000s, infecting thousands
of people.
5
CHIKV belongs to genus Alphavirus of Togaviridae family
that has enveloped positive single stranded RNA genome and belongs
Abbreviations: CHIKV, Chikungunya virus; NTR, nontranslated region; NSAID, nonsteroidal antiinflammatory drugs; ORF, open reading frames; UTR, untranslated region.
Khanzadi Nazneen Manzoor & Farakh Javed equally participated to this study
Rev Med Virol. 2021;e2287. wileyonlinelibrary.com/journal/rmv © 2021 John Wiley & Sons Ltd.
-
1 of 13
https://doi.org/10.1002/rmv.2287
to Semiliki Forest Virus antigenic serocomplex.
6,7
Genome size is
approximately 11.8 kb with two open reading frames (ORFs) that are
separated by an untranslated junction region. ORF1 encodes for
polyproteins that are precursor for nonstructural protein while
ORF2 encodes for structural protein.
8,9
After an incubation period of 1–12 days, averaging of 2–4 days
disease is characterized by highgrade fever, arthralgia, macupapular
rashes and headache.
10
Neonates and people aged more than
60 years, having higher viral load were at higher risk of acute CHIKV
infection. The case fatality is 1:1000; mostly cases involved were
elderly in the Reunion outbreak.
11
Currently, there is no antiviral
drug therapy for treatment or vaccine to prevent CHIKV infection,
treatment is based on clinical symptoms such as use of nonsteroidal
antiinflammatory drugs to reduce the symptoms.
12–14
2
|
HISTORY AND EPIDEMIOLOGY
CHIKV first recognized in 1952–1952 in Tanzania; since then, CHIKV
cases were documented in over 60 countries of Asia, Africa, Europe
and America Figure 1. In 1958 Bangkok (Thailand) outbreak of
CHIKV fever was the first significant urban outbreak of CHIKV.
15
Few epidemics of CHIKV occurred in India between 1963 and 1973,
including Kolkata outbreak in 1963, followed by Maharashtra, Tamil
Nadu and Andhra Pradesh (1964–1965) and Barsi (1973)
outbreaks.
16–19
Between 1998–1999 and 2001–2003 outbreaks
were reported in Malaysia and Indonesia respectively.
20,21
In May
2004, Lamu district, Kenya outbreak caused 13,500 infections. After
that a modest outbreak in nearby Mombasa district was thought to
be associated with pervious outbreak in Lamu district, Kenya.
16,22
In January 2005, another CHIKV outbreak in Grande Comore
Island had 215,000 reported cases.
23
In 2005–2006, severe CHIKV
outbreak in La Reunion Island of Indian Ocean infected almost
266,000 people (total population of 770,000 people) with 213
deaths.
11,24,25
Later, Aedes albopictus infection was also reported in
the outbreak of La Reunion island.
5
Prenatal transmission of CHIKV
infection at the time of delivery was observed in the epidemic of La
Reunion island of Indian Ocean.
26
After about 32 years, CHIKV
outbreak occurred in the several states of India; Andhra Pradesh
being the first state to report in December 2005. Later, the infection
spread to other 12 Indian states including Karnataka, Kerala,
Maharashtra, Tamil Naidu, Gujarat and Madhya Pradesh in which
about 1.3 million peoples were infected with CHIKV.
27–29
In
December 2006, infection spread to neighbouring countries caused
an epidemic in Maldives (3500 confirmed cases) and in Sri Lanka with
60,000 people infected and 80 deaths.
30,31
In July 2007, outbreak of CHIKV occurred in two villages of
Northern Italy, Castiglione di Cervia and Castiglione di Ravenna of
Romagna region in which 205 patients were diagnosed with CHIKV.
This outbreak was autochthonous, due to a single viremic patient
FIGURE 1 World map showing the most common outbreaks of Chikungunya virus (CHIKV) around the globe. Outbreaks of Chikungunya
virus had observed in all subcontinents
2 of 13
-
MANZOOR
ET AL.
from Kerala state of India.
32–34
In 2010, about 253 suspected and
129 laboratory confirmed CHIKV cases were reported in Guangdong
province of China, indicating the maiden outbreak of CHIKV.
35
Two
autochthonous cases were documented in Southeastern France in
2010
36,37
while in October 2014, 12 autochthonous (11 confirmed)
cases of Cameroon origin were reported in Montpelier, France.
38
Republic of Congo documented the CHIKV epidemic in 2011 in which
317 cases (37 RTPCR confirmed) were reported.
39–41
In 2011,
CHIKV antibodies were detected in 460 blood samples in Pool and
7014 samples in Brazzaville, while representative of Integrated
Regional Information Networks reported approximately 8000 sus-
pected cases, with no mortality.
39,41,42
CHIKV detected in Feb 2011 in New Caledonia, where 33
autochthonous cases indicated the entry of CHIKV in Pacific Ocean
43
while in June 2012, the epidemic appeared in Papua New Guinea
where more than 1500 cases were reported until 2013.
43
Until 2015,
CHIKV had been detected in 8 out of 22 Pacific island Territories or
countries.
44
In 2014, about 318 laboratory confirmed cases reported
in French Polynesia.
45
In Brazil, the first CHIKV case was diagnosed
in a patient returning from Indonesia (endemic country) in Rio de
Janiero on 18 August 2010
46
while until 2015 about 20,661 sus-
pected cases (7823 confirmed cases) reported by Brazilian ministry
of health
47
and in 2016, 3394 cases were reported.
42
In November 2013, the first reported CHIKV case in Saint Martin
caused an epidemic in which 26 suspected cases (20 confirmed) were
identified.
48
Later, infection spread to other territories of Caribbean
regions including Martinique, French, Guiana, Guadeloupe and Haiti
and Dominican Republic.
49
In 2013–2014, about 17,000 cases were
documented.
50
CHIKV spread to about 45 territories or countries of
South, central and North America causing more than 2.9 million sus-
pected cases with 296 deaths until July 2016.
51
US National Institute of
Allergy and Infectious Diseases in 2008 categorized CHIKV as Cate-
gory C priority agent due to high risk of spread in the different areas.
51
In Pakistan, the first large epidemic of CHIKV was reported in
Karachi city in December 2016 in which 1018 cases were reported in
Lyari, Malir, Keamari and Ibrahim Hyderi regions.
52
On 26th
December, Ministry of Health Services, Regulation and Coordination
officially reported the first CHIKV outbreak to WHO (World Health
Organization). Approximately 30,000 suspected cases and 4000
confirmed cases were reported by Armed Forces Institute of Pathology
and National Institute of Health (NIH) in Karachi.
52–54
In July 2017,
second CHIKV outbreak was reported in District of Haripur and 18
cases were confirmed as CHIKV positive by NIH Islamabad.
55,56
Next
outbreak was observed in District Manshera but no officially published
data regarding number of suspected or confirmed cases is available.
57
3
|
VECTOR AND TRANSMISSION
CHIKV, a mosquitoborne disease, is mainly transmitted via mainly
bite of infected Aedes aegypti and Aedes albopcitus mosquitos.
16,58
Two CHIKV transmission cycles namely enzootic sylvatic cycle and
urban cycle. An enzootic sylvatic cycle (animalmosquitohuman),
maintained in Africa; between forest dwelling Aedes species (Aedes
taylori,Aedes furcifer,Aedes luteocephalus,Aedes africanus and Aedes
neoafricanus) and other in nonhuman primates (rodents, monkeys,
birds and some unidentified vertebrates).
59–63
The virus occasionally
spreads to human population that live close to sylvatic environment
while sylvatic cycle have also been also reported in Asia.
64,65
Humans
are the main reservoir during epidemic while rodents, monkeys and
birds are reservoirs outside epidemic.
59
Urban cycle (humanmosquitohuman) is maintained by CHIKV
transmission between human and urban mosquitoes.
59,66
Aedes
aegypti is the major vector of CHIKV in urban outbreaks. Besides
Aedes aegypti, the other vector Aedes albopictus was observed as
vector of transmission in virus and this strain is also find associated
in Indian Ocean Island outbreak with harbouring the substitution in
alanine to valine of E1 glycoprotein (E1A226V), results in
enhanced transmitting ability.
59,67–70
In addition to Aedes aegypti
and Aedes albopictus, some other vectors found in endemic areas are
Aedes stephensi and Aedes vittatus.
71
Transmission of virus from
infected male mosquito (Aedes aegypti) to female via venereal route
have been reported.
72
Vertical transmission from mother to child
has also been analysed, causing foetal death and congenital
illness.
73
The breeding habitat of Aedes aegypti and Aedes albopictus
is mainly fresh water. The mosquito's eggs remain resistant until
rainy season giving rise to larvae.
4
Main transmission vector of
CHIKV infection, Aedes aegypti is mostly adapted to urban envi-
ronment while Aedes albopictus is well amended to both rural and
urban environment.
4,59
4
|
VIRUS VARIABILITY AND SEQUENCE
ANALYSIS
CHIKV genome contains 11,900 nucleotides and two ORFs code for
nonstructural and structural proteins.
74
CHIKV genome shows great
sequence variability but cross protection against different genotype
has been observed in patients who has been infected by one geno-
type.
75
There are three geographically related genotype of CHIKV.
One from West Africa, another from Asia and one contains all iso-
lates from Eastern, Central and South Africa (ECSA).
76
However,
genome length among these lineages varies, as ECSA lineage is
shorter than West African and Asian isolates Figure 2. Almost all
genes show nucleotide differences including 26s junction regions and
50and 30UTRs. Along with distinct genotypes, novel variants such as
quasispecies have been identified in patients infected with CHIKV as
shown in Figure 2.
77
Some sites such as E1, E2 and nsp3 show
intergenomic genetic differences that are associated with host
adaptability in alpha virus.
78
Currently intensive sequencing of
CHIKV genome is conducted and nearly 394 CHIKV sequences
including 51 full length genomes reported so far in generic data
banks (Genbank, European Molecular Biology Laboratory and DNA
Data Bank of Japan).
MANZOOR ET AL.
-
3 of 13
FIGURE 2 Evolutionary relationships of taxa. The evolutionary history was inferred using the NeighbourJoining method and Tamura Nei
method. The optimal tree with the sum of branch length =0.27860208 is shown. The percentage of replicate trees in which the associated
taxa clustered together in the bootstrap test (1000 replicates) is shown next to the branches. The tree is drawn to scale, with branch lengths in
the same units as those of the evolutionary distances used to infer the phylogenetic tree. The evolutionary distances were computed using the
Maximum Composite Likelihood method and are in the units of the number of base substitutions per site. The analysis involved 35 nucleotide
sequences. Codon positions included were 1st +2nd +3rd +Noncoding. All positions containing gaps and missing data were eliminated. There
were a total of 1096 positions in the final data set. Evolutionary analyses were conducted in MEGA5
4 of 13
-
MANZOOR
ET AL.
5
|
GENOME ORGANIZATION
CHIKV belongs to the family Togaviridae and genus Alphavirus. CHIKV
is small, enveloped virus, spherical in shape with diameter up to
70 nm.
79
Genome of CHIKV is a single and positive stranded RNA
molecule which contains about 11,900 nucleotides whose organiza-
tion is 50NTRcapnsP1nsP2nsP3nsP4(junctional region)CE3E2
6KE1poly (A)NTR3’ Figure 3. Three segments are present in non
translated (NTR) region that are 50NTR whose length is almost 76
nucleotides, 30NTR which is of 526 nucleotides in length while the
junctional region of 68 nucleotides long (Table 1).
5.1
|
Nonstructural protein
CHIKV have two ORFs like other alpha viruses. ORF contains 7424
nucleotides at 50terminal responsible for encoding nonstructural
proteins nsP14 and these proteins are involved in virus replica-
tion
80
Table 1.
5.1.1
|
nsP1
The nsP1 is 1605 bp in length; its Nterminal region is a methyl-
transferase and guanylyltransferase engaged with topping and
methylation of recently incorporated viral genomic and subgenomic
RNAs. Additionally, nsP1 is a noteworthy part of the infection
replicase complex and capacities to stay replication buildings to have
cell layers amid RNA replication.
81,82
5.1.2
|
nsP2
The nsP2 protein has large net positive charge on it (+21) and con-
sists of 2394 nucleotides. The nsP2 protein consists of three do-
mains, the main containing helicase domain, RNA triphosphatase
domain and nucleoside triphosphatase domain.
83
Though, the second
and third domains are a papainlike protease and are nonfunctional
methyltransferase. In addition, nsP2 contains a nuclear localization
sequence that made an interpretation of nsP2 to be translocated into
the nucleus.
84,85
It displays RNA triphosphatase/nucleoside triphos-
phatase and helicase exercises inside its Nterminal area, the Cend
encodes the viral cysteine protease important for preparing of
nonstructural polyprotein.
86,87
5.1.3
|
nsP3
The nsp3 consist of about 1590 bp in length and no exact capacity or
movement has been credited to this protein. It is made of two do-
mains; the first being a novel macro domain situated in the moder-
ated N terminal region. The Cterminal domain is less moderated and
is phosphorylated in up to 16 positions on serines and
threonines.
88,89
The part of phosphorylation is not all around recor-
ded be that as it may, cancellation of the phosphorylated residues
and diminishes the RNA synthesis level. The C end of nsP3 is in this
manner thought to have a superfluous regulatory part.
90
5.1.4
|
nsP4
The nsP4 is of 1833 bp long and contains the RNAdependent RNA
polymerase, engaged with genome replication and interpretation.
91
It
works as RNAdependent RNApolymerase and could likewise as-
sume a platform part for connection with different nsPs or host
proteins through its N terminal end. It is the most conserved
protein.
92
All of these ns proteins are involved in formation of RNA repli-
case along with some proteins from host cell.
93
5.2
|
Structural proteins
ORF contain 3732 nucleotides fragment at its 30terminal and these
nucleotides are responsible for encoding five structural polyproteins
(sPs), that is, capsid, E3, E2, 6K/TF and E1 protein.
5.2.1
|
Capsid
Capsid consists of 783 nucleotides and contains two domains. First,
the amino terminal domain, is not involved in formation of structural
architecture but takes part in nucleocapsid formation as well as
interaction with RNA to enclose the genome. While the second is
carboxyl terminal domain that serves as serine protease.
94–96
5.2.2
|
E3
This protein is 192 bp long and it is basically α/βprotein which
regulates the spike assembly and interacts with E2 glycoprotein.
97
5.2.3
|
E2
E2 protein is about 1269 bp long. It is outer most region of spike
protein and have three immunoglobulin domains that is, A, B and C
domains as well as two glycosylation sites positions at 263 and 345.
98
The C domain is near to the viral membrane and function as a linker to
the transmembrane region. The B domain is present at the membrane
distal end which allows the contact with E3 protein. While the A
domain is present in the centre of protein.
99
E2 protein involved in
facilitating the virus to enter cells via A and B domains by two
mechanisms which are glycosaminoglycan GAGindependent and
GAGdependent.
100
While E2 is also the main target of neutralizing
antibodies.
98
MANZOOR ET AL.
-
5 of 13
TABLE 1Structural and nonstructural proteins of CHIKV
Protein name Symbol Size (aa) Location in cell Function
Structural
Capsid C 261 Cytoplasm Nucleocapsid formation as well as interacts with RNA to enclose the genome
Envelope 3 E3 64 Cytoplasm Regulates the spike assembly and interacts with E2 glycoprotein
Envelope 2 E2 423 Cytoplasm Involved in facilitating the virus to enter into cells
6K transframe 6K/TF 61 Cytoplasm Enhancing the virus assembly and release
Envelope 1 E1 436 Cytoplasm Involved in pre and post fusion events
Nonstructural
nsP1 protein nsp1 535 Cytoplasm Methylation of recently incorporated viral genomic and sub genomic RNAs
nsP2 protein nsP4 799 Cytoplasm Helicase exercises inside its Nterminal area, viral cysteine protease
nsP3 protein nsP3 530 Cytoplasm Cancellation of the phosphorylated residues and diminishes the RNA synthesis level
nsP4 protein nsP4 61 Cytoplasm Engaged with genome replication and interpretation
FIGURE 3 Genome of Chikugunya virus (CHIKV). CHIKV possess +ssRNA with an open reading frame about 12 kb, encoding five
structural protein that is, capsid, E3, E2, 6K/TF and E1 protein and four nonstructural protein np1 to np5 are coloured in green. The amino
acid lengths of all encoding proteins are indicated in black. Red, black, dark green and purple arrow indicate the cleavage site of viral nsp2
protease, signal peptidase capsid and furin, separately
6 of 13
-
MANZOOR
ET AL.
5.2.4
|
6K/TF
6K/TF is 183 bp in length. During the translation of 6K gene the
protein is produced by a frame shift although it is transframe (TF)
protein that is involved in enhancing the virus assembly and release.
5.2.5
|
E1
E1 protein is 436 bp in length and consists of three βsheet rich
domains (domains I, II and III), involved in pre and post fusion events
of replicating cycle.
101
6
|
PATHOGENESIS AND CLINICAL
MANIFESTATIONS
Molecular mechanisms of virus hostcell interactions and pathogen-
esis of disease are not fully understood. Infected female mosquitoes
suck the blood from person infected with CHIKV and then allow viral
replication in the midgut for some days and then transmit that virus
to another host by biting. Epithelial and endothelial cells, primary
fibroblasts and macrophages are vulnerable to the virus and allow for
its replication.
16
There are three phases of disease and infected
person suffers with at least one of these three phases which are
acute, postacute and chronic stage.
102
6.1
|
First acute phase
Acute phase is comprising of first three weeks of infection. Signs and
symptoms arise after incubation period which is on average about 4–
7 days (in range of 1–12 days). Symptoms do not appear in all
infected persons. Asymptomatic infections are reported in about 3%–
25% patients with antibodies to CHIKV.
16,103
Signs and symptoms
then last for 5–10 days. In 99.5% cases, arthritis with severe pain,
pyrexia and inflammatory arthralgia are typical clinical presentations
of infection. Fever is very high, which is even not controlled by an-
tipyretics, along with painful and swollen peripheral joints. Other
signs and symptoms include headache, macular to maculopapular
rash, myalgia oedema of face and other extremities. In children, there
may be epistaxis and gingival bleeding, but it is not common in adults.
During acute phase, some unusual symptoms also appear which are
neurological and gastrointestinal, damaging of mucus membrane,
malaise, renal and respiratory failure, and pancreatitis.
104–106
Despite
all these, most patients have clinical improvement within 1–
2 weeks.
106
6.2
|
Second postacute phase
This phase commences from 4th week till the end of 3rd month. Sign
and symptoms of this phase are periarticular and synovial
inflammation along with severe arthritis, neuropathy, neuropsychi-
atric disorders and peripheral vascular disorders.
107
6.3
|
Third chronic phase
This phase appears after 3rd month when clinical symptoms still
exist.
107
In Reunion Island from 2005–2006 chronic phase lasted from
few months to several years in some infected patients and in Angola it
lasted up to 15 years.
102,108
Chronic disease was observed in 80%–
93% of patients in Reunion Island, from these patients 57% had it for
15 months and 47% for about 2 years.
104,109
It was observed in a study
that 76 patients suffered with chronic disease for about 3 years and
only 31% of these fully recovered while remaining patients suffered
from intermittent arthralgia due to which these patients were unable
to lift heavy objects and even not walk.
110
Mental health problems
have also been reported in many patients. It was estimated that eco-
nomic burden is about €250.00 per patient every year.
110,111
CHIKV
infections are mostly confused with dengue viral infection because in
both infections there is high fever along with myalgia and same mos-
quito specie transmit both viruses. But despite all this arthralgias in
multiple joints is more common in case of CHIKV only.
112–116
7
|
DIAGNOSIS
7.1
|
Genetic method
Samples taken from patient, RNA of virus or antibodies to CHIKV can
be detected for the confirmation of infection. In past, diagnosis was
based on serological techniques but with the advance molecular
techniques now RNA of virus can be detected in serum by the
reverse transcriptasepolymerase chain reaction (RTPCR) specif-
ically when infection is in acute phase. There is high level of viremia
by CHIKV infection which usually lasts for 4–6 days and persist up to
12 days from disease onset.
117,118
Due to this reason viral culture or
RTPCR during an acute phase can be used for confirmation of
infection. Virus can be easily isolated from serum that is collected
during first 7 days of disease while the nucleic acid of virus can be
detected after few days with the help of real time RTPCR.
117,119
For
detection of CHIKV RT loopmediated isothermal amplification can
also be used.
120
There is an advantage of this technique that no
specialized equipment like thermocyclers is required in it. Isolation of
virus requires 48 h and molecular assays are completed within 1 day.
7.2
|
Virological method
CHIKV infection can be confirmed virologically by isolation of
virus from plasma, sera and tissue or whole blood. That diagnosis
is very specific but not sensitive. Viral antigens can be detected
with help of enzymelinked immunosorbent assay (ELISA) or
immunochromatographic assay. But only some antigen detection
MANZOOR ET AL.
-
7 of 13
kits are available with different sensitivity level towards different
genotypes of virus.
121
7.3
|
Serological method
AntiCHIKV immunoglobulin IgM and IgG antibodies can be detected
by ELISA.
117,119
ELISA is very specific for detection of infection
because it has very little cross reactivity with other alpha viruses. For
diagnosis of CHIKV infection, rapid dipstick variations of CHIKV
ELISA were developed recently but availability of these limited
worldwide.
120
Despite these techniques, plaque reduction neutrali-
zation test (PRNT) and immunofiuorescence assays are also used for
detecting antibodies to CHIKV in the serum.
16
PRNTs are very useful
for diagnosis because of their specificity to alpha viruses and are
thought to be gold standard to confirm the results of serological
tests. The use of live virus is required for PRNTs which is its major
drawback. Immunofiuorescence assays are very specific and sensi-
tive, but these are unable to quantify antibodies as well as requires
special equipment and training. But still these tests are available
commercially and are optionally used in those laboratories where
other infectious agents are to be detected.
16
8
|
PROSPECTIVE TREATMENT AND VACCINES
There are no antiviral drugs or vaccines for treatment and prevention
of CHIKV infection. Antipyretics, analgesics and antiinflammatory
agents may be used for treatment of different symptoms. IgGs anti-
bodies obtained from patients infected with CHIKV show protection
against CHIKV infection in mice.
122
This method becomes very
effective in neonates for treatment and prevention from CHIKV
infection whose mothers are viremic. However, this method is very
costly, and it is very difficult to develop CHIKV antiviral drugs and
vaccines. But due to recent outbreaks of CHIKV in Africa, La Réunion
island and Asia different research avenues have been started for
development of CHIKV antiviral drugs and vaccines (Table 2).
127
It
was reported that chloroquine phosphate is effective for treating
chronic CHIKV arthritis.
128
For treatment of CHIKV induced
arthritis, ribavirin which is an antiviral agent was used and it was
reported to be effective in reducing the swelling of joint and soft
tissue.
129
Bindarit, which is an antiinflammatory drug, has been re-
ported to be effective for treatment of arthritis caused by CHIKV
infection.
130
Two compounds harrington and cephalotoxine alkaloid
inhibited CHIKV replication in early stages.
131
Controlling mosquitoes can be helpful in prevention of disease
but it is very difficult strategy. Most effective way of disease control
would be vaccination. For development of vaccine against CHIKV,
infection different approaches have been used. The first vaccine
against CHIKV was formalin inactivated vaccine.
132
ECSA genotype of
2006 Indian outbreak strain was used for development of formalin
inactivated vaccine which was whole virus vaccine.
133
In 1986, Levitt
and colleagues developed the first live attenuated vaccine. It was
developed by plaquepurified CHIKV in MRC5 (human foetal lung
fibroblast) cell line.
134
At US Army Medical Research Institute of In-
fectious Diseases, liveattenuated CHIKV vaccine was also developed.
That vaccine was safe to be used as well as very well tolerated.
123
Other than these, DNA based vaccine using envelope (structural
protein) sequences was also developed.
135
By using a combined E1, E2
and E3 genes construct was found to be helpful in generating cell
mediated as well as humoral immune response.
124
VLP vaccines were
also developed that have ability of producing high tittered neutral-
izing antibodies.
136
But there was a drawback of all these vaccines.
Multiple immunizations were required for these vaccines and
TABLE 2Vaccines for CHIKV under process and clinical trials
Vaccine Description Trial Developer References
TSIGSD218 Live attenuated virus vaccine developed by United States
Army Medical Research Institute of infectious Diseases.
Produced by serial passage in MRC5 cells. Further
development stops due to funding issues
Phase II US Army Medical Research
Institute of Infectious
Diseases, The Salk Institute
for Biological Studies
[123]
VRCCHKV Chikungunya VLPs that are composed of the E1, E2, and
capsid proteins from the chikungunya virus strain 37997.
Manufactured at VRC, NIAID, Vaccine Pilot Plant
operated by Leidos Biomedical Research. It was safe with
good immunogenicity profile
Phase II National Institute of Allergy and
infectious Diseases (Vaccine
Pilot Plant operated by
Leidos Biomedical Research)
[124]
MVCHIK Live recombinant measlesvirus based chikungunya vaccine.
Live attenuated recombinant viral vectored vaccine based
on the Schwarz strain of measles vaccine. First measles
virus based candidate vaccine for humans
Phase I Themis Bioscience GmbH/
Institut Pasteur
[125]
CHIKV/IRES Liveattenuated vaccine based on the insertion of a
picornavirus IRES sequence into the genome of CHIKV.
Induces strong neutralizing antibody response and
protects mice after a single dose
Phase I Takeda Pharmaceuticals U.S.A.,
Inc./University of Texas
Medical Branch
[126]
Abbreviations: CHIKV, Chikungunya virus; IRES, internal ribosome entry site.
8 of 13
-
MANZOOR
ET AL.
manufacturing was also expensive. This problem was overcome by the
development of live attenuated vaccine. CHIKIRES vaccine that
proved to be safe as well as efficient.
126
Chimeric viruses that contain
CHIKV structural proteins, Eastern equine encephalitis virus and
Venezuelan equine encephalitis virus induced neutralizing antibodies
and thus help in protection of mice from CHIKV infection.
9
9
|
CHIKV AND HEALTH COMMUNICATION IN
THE 21ST CENTURY
Different social networks play crucial role in spreading awareness
about CHIKV and its consequences such as Facebook, Instagram and
Twitter. Moreover, Facebook is main information source that contain
different posts which could be reliable and relevant to disease.
137
CHIKV related contents at twitter is under different themes and
hashtags such as (i) Aedes Aegypti, (ii) Chikungunya, (iii) Preventing
Mosquito Borne Disease and (iv) Health Care Services. Apart from
that, different news channels are the second most important source
for CHIKVrelated information such as Science Daily, Dawn News
and The Star Online. Main concern of Science Daily was to investi-
gate the cause of persistent joint pain after months of infection while,
Dawn News showed the epidemiology of disease in different areas of
Pakistan and its severity compared to last year.
138
The Star News
indicated the new cases in traders and merchants in Simbang Kuala
that was investigated by Kedah Health Department Malaysia.
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS
CoV2), also named as the 2019 novel Coronavirus (2019nCoV), is a
newly emerging zoonotic agent that come into sight in December
2019 and is the causative agent of the Coronavirus Disease 2019
(COVID19) pandemic.
139
Wide spread of virus is due to its ability of
persontoperson transmission. People get infected via direct contact
or by droplets that are produced through sneezing or coughing of
infected person.
140
CHIKV is an arthropod borne virus and spread via
infected mosquito.
16
These attributes of CHIKV reduce likelihood of
its epidemics to develop into a pandemic.
To control pandemic in future it is required to take various pre-
cautionary measures. Quick and accurate method are required for
rapid diagnose and reliable results. Widely used standard method for
viral diagnosis is quantitative reverse polymerase chain reaction.
MALDITOF MS and IR spectroscopy are also introduced for routine
identification of bacteria, viruses and fungi.
141
It is required to conduct
epidemiological surveillance and puts pressure on health care units.
142
10
|
CHIKV, URBANIZATION AND GEOGRAPHIC
EXPANSION
The habitat which is created by us allows Aedes mosquitoes to live with
us. In urban areas, high population densities are of major concern
regarding disease transmission that are of mosquito borne. In urban
zones from 1960 to 2014 population increased from 34% to 54%.
143
As
hypothesized by LaDeau regarding mosquito population there are two
effects of urbanization: buffer temperature fluctuations and mean
temperatures are raised by the urban heat islands. This in turn facili-
tates the growth and viral replication rates. Immature mosquito life
stages are further supported by creation of additional container habi-
tats.
144
In urban environments, the population is increasing which is
creating good habitats for vectors. The risk of new emergence of CHIKV
epidemics increases due to movements of international population and
dissemination of mosquito worldwide progressively. This threat can be
limited in susceptible regions by international collaboration. In
epidemic countries, it can be controlled by controlling the vector as well
as isolating infectious people.
135
11
|
CONCLUSION
CHIKV is a reemerging virus that is known to infect millions of
people worldwide. CHIKV is a major health concern. The main
chronic symptom of CHIKV infection is persisting arthralgia.
Continuous viral surveillance is needed and will help in gathering
data related to viral genotype which could be helpful in development
of viral drugs and vaccines.
ACKNOWLEDGEMENTS
None.
CONFLICT OF INTEREST
There is no conflict of interest exists among the authors.
AUTHOR CONTRIBUTIONS
Farakh Javed conceived the idea and design the flow of manuscript.
Farakh Javed, Nazneen Manzoor, Muhammad Ejaz, Mubashir Ali and
Neelam Mujaddadi wrote the first draft of the manuscript. Farakh
Javed, Nazneen Manzoor, Sobia Manzoor, Waqar Saeed, Aamer Ali
Khattak. Ibrar Ahmad, Abid Ali Khan and Asad Zaib revised the
manuscript. Farakh Javed, Nazneen Manzoor and Mubashir Ali pro-
duced the figures. All authors reviewed and approved the final version
of the manuscript.
DATA AVAILABILTY STATEMENT
Data sharing not applicable to this article as no datasets were
generated or analysed during the current study.
ORCID
Farakh Javed
https://orcid.org/0000-0003-1305-7834
Muhammad Ejaz https://orcid.org/0000-0002-1339-5537
REFERENCES
1. Mason PJ, Haddow AJ. An epidemic of virus disease in Southern
Province, Tanganyika Territory, in 195253; an additional note on
Chikungunya virus isolations and serum antibodies. Trans R Soc
Trop Med Hyg. 1957;51(3):238240.
2. Robinson MC. An epidemic of virus disease in Southern Province,
Tanganyika Territory, in 195253. I. Clinical features. Trans R Soc
Trop Med Hyg. 1955;49(1):2832.
MANZOOR ET AL.
-
9 of 13
3. Ross RW. The Newala epidemic. III. The virus: isolation, pathogenic
properties and relationship to the epidemic. J Hyg (Lond).
1956;54(2):177191.
4. Pialoux G, Gauzere BA, Jaureguiberry S, Strobel M. Chikungunya,
an epidemic arbovirosis. Lancet Infect Dis. 2007;7(5):319327.
doi:10.1016/S1473-3099(07)70107-X
5. Vazeille M, Moutailler S, Coudrier D, et al. Two Chikungunya
isolates from the outbreak of La Reunion (Indian Ocean)
exhibit different patterns of infection in the mosquito, Aedes
albopictus. PLoS One. 2007;142(11):e1168. doi:10.1371/journal.
pone.0001168
6. Powers AM, Brault AC, Tesh RB, Weaver SC. Reemergence of
Chikungunya and O'nyongnyong viruses: evidence for distinct
geographical lineages and distant evolutionary relationships. J
Gen Virol. 2000;81(Pt 2):471479. doi:10.1099/0022-1317-81-
2-471
7. Powers AM, Brault AC, Shirako Y, et al. Evolutionary relationships
and systematics of the alphaviruses. J Virol. 2001;75(21):10118-
10131. doi:10.1128/JVI.75.21.10118-10131.2001
8. Li XF, Jiang T, Deng YQ, et al. Complete genome sequence of a
chikungunya virus isolated in Guangdong, China. J Virol.
2012;86(16):89048905. doi:10.1128/JVI.01289-1286/16/8904
9. Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT.
Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines.
2012;11(9):10871101. doi:10.1586/erv.12.84
10. Horwood PF, Buchy P. Chikungunya. Rev Sci Tech.
2015;34(2):479489.
11. Josseran L, Paquet C, Zehgnoun A, et al. Chikungunya disease
outbreak, Reunion Island. Emerg Infect Dis. 2006;12(12):19941995.
doi:10.3201/eid1212.060710
12. Bettadapura J, Herrero LJ, Taylor A, Mahalingam S. Approaches to
the treatment of disease induced by chikungunya virus. Ind J Med
Res. 2013;138(5):762.
13. da Cunha RV, Trinta KS. Chikungunya virus: clinical aspects and
treatmentA Review. Memórias do Inst Oswaldo Cruz.
2017;112(8):523531.
14. de Lamballerie X, Ninove L, Charrel RN. Antiviral treatment of
chikungunya virus infection. Infect Disord Drug Targets.
2009;9(2):101104.
15. Rianthavorn P, Prianantathavorn K, Wuttirattanakowit N, The-
amboonlers A, Poovorawan Y. An outbreak of chikungunya in
southern Thailand from 2008 to 2009 caused by African strains
with A226V mutation. Int J Infect Dis. 2010;14(Suppl 3):e161e165.
doi:10.1016/j.ijid.2010.01.001S12019712(10)023209
16. Staples JE, Breiman RF, Powers AM. Chikungunya fever: an
epidemiological review of a reemerging infectious disease. Clin
Infect Dis. 2009;49(6):942948. doi:10.1086/605496
17. Naresh Kumar CV, Sai Gopal DV. Reemergence of chikungunya
virus in Indian Subcontinent. Indian J Virol. 2010;21(1):817.
doi:10.1007/s13337-010-0012-112
18. Lahariya C, Pradhan SK. Emergence of chikungunya virus in Indian
subcontinent after 32 years: a review. J Vector Borne Dis.
2006;43(4):151160.
19. Patil SS, Patil SR, Durgawale PM, Patil AG. A study of the outbreak
of Chikungunya fever. J Clin Diagn Res. 2013;7(6):10591062.
doi:10.7860/JCDR/2013/5330.3061
20. Lam SK, Chua KB, Hooi PS, et al. Chikungunya infection–an
emerging disease in Malaysia. Southeast Asian J Trop Med Publ
Health. 2001;32(3):447451.
21. Laras K, Sukri NC, Larasati RP, et al. Tracking the reemergence of
epidemic chikungunya virus in Indonesia. Trans R Soc Trop Med Hyg.
2005;99(2):128141.
22. Sergon K, Njuguna C, Kalani R, et al. Seroprevalence of chikungu-
nya virus (CHIKV) infection on Lamu Island, Kenya, October 2004.
Am J Trop Med Hyg. 2008;78(2):333337.
23. Sergon K, Yahaya AA, Brown J, et al. Seroprevalence of chi-
kungunya virus infection on Grande Comore Island, union of the
Comoros, 2005. Am J Trop Med Hyg. 2007;76(6):11891193.
24. Gerardin P, Guernier V, Perrau J, et al. Estimating Chikungunya
prevalence in La Reunion Island outbreak by serosurveys: two
methods for two critical times of the epidemic. BMC Infect Dis.
2008;288:99. doi:10.1186/1471-2334-8-991471-2334-8-99
25. Borgherini G, Poubeau P, Staikowsky F, et al. Outbreak of chi-
kungunya on Reunion Island: early clinical and laboratory features
in 157 adult patients. Clin Infect Dis. 2007;144(11):14011407.
doi:10.1086/517537
26. Gerardin P, Barau G, Michault A, et al. Multidisciplinary prospec-
tive study of mothertochild chikungunya virus infections on the
island of La Reunion. PLoS Med. 2008;185(3):e60. doi:10.1371/
journal.pmed.00500607PLMERA1274
27. Ravi V. Reemergence of chikungunya virus in India. Indian J Med
Microbiol. 2006;24(2):8384.
28. Manimunda SP, Sugunan AP, Rai SK, et al. Outbreak of chikungunya
fever, Dakshina Kannada District, South India, 2008. Am J Trop Med
Hyg. 2010;83(4):751754. doi:10.4269/ajtmh.2010.09-043383/4/751
29. Cecilia D. Current status of dengue and chikungunya in India. WHO
South East Asia J Public Health. 2014;3(1):2226. doi:10.4103/2224-
3151.206879
30. Kaur P, Ponniah M, Murhekar MV, et al. Chikungunya outbreak,
South India, 2006. Emerg Infect Dis. 2008;14(10):16231625.
doi:10.3201/eid1410.070569
31. Talawar AS, Pujar HS. An outbreak of Chikungunya epidemic in
South IndiaKarnataka. Int J Res Rev Appl Sci. 2010;5:229234.
32. Sambri V, Cavrini F, Rossini G, Pierro A, Landini MP. The 2007
epidemic outbreak of Chikungunya virus infection in the Romagna
region of Italy: a new perspective for the possible diffusion of tropical
diseases in temperate areas? New Microbiol. 2008;31(3):303304.
33. Rezza G, Nicoletti L, Angelini R, et al. Infection with chikungunya
virus in Italy: an outbreak in a temperate region. Lancet.
2007;370(9602):18401846. doi:10.1016/S0140-6736(07)61779-6
34. Angelini R, Finarelli AC, Angelini P, et al. An outbreak of chi-
kungunya fever in the province of Ravenna, Italy. Euro Surveill.
2007;12(9):E070906 1.
35. Qiaoli Z, Jianfeng H, De W, et al. Maiden outbreak of chikungunya
in Dongguan city, Guangdong province, China: epidemiological
characteristics. PLoS One. 2012;7(8):e42830. doi:10.1371/journal.
pone.0042830
36. Grandadam M, Caro V, Plumet S, et al. Chikungunya virus, south-
eastern France. Emerg Infect Dis. 2011;17(5):910913. doi:10.3201/
eid1705.101873
37. Gould EA, Gallian P, De Lamballerie X, Charrel RN. First cases of
autochthonous dengue fever and chikungunya fever in France: from
bad dream to reality!. Clin Microbiol Infect. 2010;16(12):17021704.
doi:10.1111/j.1469-0691.2010.03386.xS1198-743X(14)60569-3
38. Delisle E, Rousseau C, Broche B, et al. Chikungunya outbreak in
Montpellier, France, September to October 2014. Euro Surveill.
2015;20(17):21108.
39. Kelvin AA. Outbreak of chikungunya in the Republic of Congo and
the global picture. J Infect Dev Ctries. 2011;5(6):441444.
40. Moyen N, Thiberville SD, Pastorino B, et al. First reported chi-
kungunya fever outbreak in the republic of Congo, 2011. PLoS One.
2014;9(12):e115938. doi:10.1371/journal.pone.0115938
41. Diop D, Meseznikov G, Sanicas M. Chikungunya outbreaks from
2000 to 2015: a review. MOJ Public Health. 2015;2(6):00043.
42. Wahid B, Ali A, Rafique S, Idrees M. Global expansion of chi-
kungunya virus: mapping the 64year history. Int J Infect Dis.
2017;58:6976. doi:10.1016/j.ijid.2017.03.006
43. Nhan TX, Musso D. The burden of chikungunya in the Pacific. Clin
Microbiol Infect. 2015;21(6):e47e48. doi:10.1016/j.cmi.2015.02.
018S1198-743X(15)00310-9
10 of 13
-
MANZOOR
ET AL.
44. CaoLormeau VM, Musso D. Emerging arboviruses in the Pacific.
Lancet. 2014;384(9954):15711572. doi:10.1016/S0140-6736(14)
61977-2
45. Aubry M, Teissier A, Roche C, et al. Chikungunya outbreak, French
Polynesia, 2014. Emerg Infect Dis. 2015;21(4):724726.
doi:10.3201/eid2104.141741
46. Albuquerque IG, Marandino R, Mendonca AP, et al. Chikungunya
virus infection: report of the first case diagnosed in Rio de Janeiro,
Brazil. Rev Soc Bras Med Trop. 2012;45(1):128129. doi:10.1590/
s0037-86822012000100026
47. Rodrigues Faria N, Lourenco J, Marques de Cerqueira E, Maia de
Lima M, Pybus O, Alcantara LCJ. Epidemiology of chikungunya
virus in Bahia, Brazil, 20142015. PLoS Curr. 2016:8. doi:10.1371/
currents.outbreaks.c97507e3e48efb946401755d468c28b2
48. Cassadou S, Boucau S, PetitSinturel M, Huc P, LeparcGoffart I,
Ledrans M. Emergence of chikungunya fever on the French side
of Saint Martin island, October to December 2013. Euro Surveill.
2014;19(13):20752. doi:10.2807/1560-7917.es2014.19.13.
20752
49. Mowatt L, Jackson ST. Chikungunya in the Caribbean: an epidemic
in the making. Infect Dis Ther. 2014;3(2):6368. doi:10.1007/
s40121-014-0043-9
50. Van Bortel W, Dorleans F, Rosine J, et al. Chikungunya outbreak in
the Caribbean region, December 2013 to March 2014, and the
significance for Europe. Euro Surveill. 2014;19(13):20759.
doi:10.2807/1560-7917.es2014.19.13.20759
51. Yactayo S, Staples JE, Millot V, Cibrelus L, RamonPardo P.
Epidemiology of chikungunya in the Americas. J Infect Dis.
2016;214(suppl 5):S441S445. doi:10.1093/infdis/jiw390
52. Ali I, Dasti JI. Chikungunya virus; an emerging arbovirus in
Pakistan. J Pakistan Med Assoc. 2018;68(2):252257.
53. Rauf M, Fatima Tuz Z, Manzoor S, Mehmood A, Bhatti S. Outbreak
of chikungunya in Pakistan. Lancet Infect Dis. 2017;17(3):258.
doi:10.1016/S1473-3099(17)30074-9
54. Aamir UB, Badar N, Salman M, Ahmed M, Alam MM. Outbreaks of
chikungunya in Pakistan. Lancet Infect Dis. 2017;17(5):483.
doi:10.1016/S1473-3099(17)30191-3
55. Chikungunya Found. Haripur villages undergo emergency fumigation.
Updated 15 July 2017. Accessed. 17 March, 2018. https://
tribune.com.pk/story/1458321/chikungunya-found-haripur-villages-
undergo-emergency-fumigation./
56. D'Alessio M, De Nicola M, Coppola S, et al. Oxidative Bax dimeriza-
tion promotes its translocation to mitochondria independently of
apoptosis. FASEB J. 2005;19(11):15041506. doi:10.1096/fj.04-
3329fje
57. Qi F, Li A, Zhao L, et al. Cinobufacini, an aqueous extract from
Bufo bufo gargarizans Cantor, induces apoptosis through a
mitochondriamediated pathway in human hepatocellular carci-
noma cells. J Ethnopharmacol. 2010;128(3):654661. doi:10.1016/
j.jep.2010.02.022S03788741(10)001376
58. Bala Murugan S, Sathishkumar R. Chikungunya infection: a po-
tential reemerging global threat. Asian Pac J Trop Med.
2016;9(10):933937. doi:10.1016/j.apjtm.2016.07.020
59. Singh SK, Unni SK. Chikungunya virus: host pathogen interaction.
Rev Med Virol. 2011;21(2):7888. doi:10.1002/rmv.681
60. Pardigon N. The biology of chikungunya: a brief review of what we
still do not know. Pathol Biol (Paris). 2009;57(2):127132.
doi:10.1016/j.patbio.2008.02.016S0369-8114(08)00040-0
61. Vourc'h G, Halos L, Desvars A, et al. Chikungunya antibodies
detected in nonhuman primates and rats in three Indian Ocean
islands after the 2006 ChikV outbreak. Vet Res. 2014;45:52.
doi:10.1186/1297-9716-45-521297-9716-45-52
62. Weaver SC, Lecuit M. Chikungunya virus and the global spread of a
mosquitoborne disease. N Engl J Med. 2015;372(13):12311239.
doi:10.1056/NEJMra1406035
63. Ap Y, Nazni W, Azleen ZN, et al. The first isolation of chikungunya
virus from nonhuman primates in Malaysia. J General Mol Virol.
2009;1(3):035039.
64. Diallo D, Sall AA, Buenemann M, et al. Landscape ecology of syl-
vatic chikungunya virus and mosquito vectors in southeastern
Senegal. PLoS Negl Trop Dis. 2012;6(6):e1649. doi:10.1371/journal.
pntd.0001649PNTDD1100975
65. Sam IC, Chua CL, RovieRyan JJ, et al. Chikungunya virus in Ma-
caques, Malaysia. Emerg Infect Dis. 2015;21(9):16831685.
doi:10.3201/eid2109.150439
66. Coffey LL, Failloux AB, Weaver SC. Chikungunya virusvector in-
teractions. Viruses. 2014;6(11):46284663. doi:10.3390/
v6114628v6114628
67. Pages F, Peyrefitte CN, Mve MT, et al. Aedes albopictus mosquito:
the main vector of the 2007 Chikungunya outbreak in Gabon. PLoS
One. 2009;4(3):e4691. doi:10.1371/journal.pone.0004691
68. de Lamballerie X, Leroy E, Charrel RN, Ttsetsarkin K, Higgs S,
Gould EA. Chikungunya virus adapts to tiger mosquito via evolu-
tionary convergence: a sign of things to come? Virol J. 2008;5:33.
doi:10.1186/1743-422X-5-33
69. Thavara U, Tawatsin A, Pengsakul T, et al. Outbreak of chikungu-
nya fever in Thailand and virus detection in field population of
vector mosquitoes, Aedes aegypti (L.) and Aedes albopictus Skuse
(Diptera: Culicidae). Southeast Asian J Trop Med Publ Health.
2009;40(5):951962.
70. DupontRouzeyrol M, Caro V, Guillaumot L, et al. Chikungunya virus
and the mosquito vector Aedes aegypti in New Caledonia (South
Pacific Region). Vector Borne Zoonotic Dis. 2012;12(12):10361041.
doi:10.1089/vbz.2011.0937
71. Powers AM, Logue CH. Changing patterns of chikungunya virus:
reemergence of a zoonotic arbovirus. J Gen Virol. 2007;88(Pt
9):23632377. doi:10.1099/vir.0.82858-0
72. Mavale M, Parashar D, Sudeep A, et al. Venereal transmission of
chikungunya virus by Aedes aegypti mosquitoes (Diptera: Culici-
dae). Am J Trop Med Hyg. 2010;83(6):12421244. doi:10.4269/
ajtmh.2010.09-057783/6/1242
73. Ramful D, Carbonnier M, Pasquet M, et al. Mothertochild
transmission of Chikungunya virus infection. Pediatr Infect Dis J.
2007;26(9):811815. doi:10.1097/INF.0b013e3180616d4f
74. Ooi MK, Gan HM, Rohani A, Hassan SS. First complete genome
sequence of a Chikungunya virus strain isolated from a patient
diagnosed with dengue virus infection in Malaysia. Genome
Announc. 2016;4(4):e0087616. doi:10.1128/genomeA.00876-16
75. Chua CL, Sam IC, Merits A, Chan YF. Antigenic variation of East/
Central/South African and Asian Chikungunya virus genotypes in
neutralization by immune sera. PLoS Negl Trop Dis. 2016;10(8):
e0004960. doi:10.1371/journal.pntd.0004960PNTD-D-15-01358
76. Gokhale MD, Paingankar MS, Sudeep AB, Parashar D. Chikungunya
virus susceptibility & variation in populations of Aedes aegypti
(Diptera: Culicidae) mosquito from India. Indian J Med Res.
2015;142(Suppl):S33S43. doi:10.4103/0971-5916.176614
77. Stapleford KA, Moratorio G, Henningsson R, et al. Wholegenome
sequencing analysis from the Chikungunya virus Caribbean outbreak
reveals novel evolutionary genomic elements. PLoS Negl Trop Dis.
2016;10(1):e0004402. doi:10.1371/journal.pntd.0004402PNTDD
1501720
78. Sam IC, Loong SK, Michael JC, et al. Genotypic and phenotypic
characterization of Chikungunya virus of different genotypes from
Malaysia. PLoS One. 2012;7(11):e50476. doi:10.1371/journal.
pone.0050476PONED1119201
79. Solignat M, Gay B, Higgs S, Briant L, Devaux C. Replication cycle of
chikungunya: a reemerging arbovirus. Virology. 2009;393(2):183197.
doi:10.1016/j.virol.2009.07.024S00426822(09)004565
80. WegerLucarelli J, Aliota MT, Wlodarchak N, Kamlangdee A,
Swanson R, Osorio JE. Dissecting the role of E2 protein domains in
MANZOOR ET AL.
-
11 of 13
alphavirus pathogenicity. J Virol. 2015;90(5):24182433.
doi:10.1128/JVI.02792-15JVI.02792-15
81. Ahola T, Kääriäinen L. Reaction in alphavirus mRNA capping: for-
mation of a covalent complex of nonstructural protein nsP1 with 7
methylGMP. Proc Natl Acad Sci. 1995;92(2):507511.
82. Peränen J, Laakkonen P, Hyvönen M, Kääriäinen L. The alphavirus
replicase protein nsP1 is membraneassociated and has affinity to
endocytic organelles. Virology. 1995;208(2):610620.
83. Rikkonen M, Peränen J, Kääriäinen L. ATPase and GTPase activ-
ities associated with Semliki Forest virus nonstructural protein
nsP2. J Virol. 1994;68(9):58045810.
84. Strauss EG, De Groot RJ, Levinson R, Strauss JH. Identification of
the active site residues in the nsP2 proteinase of Sindbis virus.
Virology. 1992;191(2):932940.
85. Peränen J, Rikkonen M, Liljeström P, Kääriäinen L. Nuclear locali-
zation of Semliki Forest virusspecific nonstructural protein nsP2. J
Virol. 1990;64(5):18881896.
86. Gomez de Cedrón M, Ehsani N, Mikkola ML, Garcıa JA, Kääriäinen
L. RNA helicase activity of Semliki Forest virus replicase protein
NSP2. FEBS Lett. 1999;448(1):1922.
87. Hardy WR, Strauss JH. Processing the nonstructural polyproteins
of sindbis virus: nonstructural proteinase is in the Cterminal half
of nsP2 and functions both in cis and in trans. J Virol.
1989;63(11):46534664.
88. Vihinen H, Saarinen J. Phosphorylation site analysis of Semliki
forest virus nonstructural protein 3. J Biol Chem.
2000;275(36):2777527783.
89. Li G, La Starza MW, Hardy WR, Strauss JH, Rice CM. Phosphory-
lation of Sindbis virus nsP3 in vivo and in vitro. Virology.
1990;179(1):416427.
90. Vihinen H, Ahola T, Tuittila M, Merits A, Kääriäinen L. Elimination
of phosphorylation sites of Semliki Forest virus replicase protein
nsP3. J Biol Chem. 2001;276(8):57455752.
91. Rubach JK, Wasik BR, Rupp JC, Kuhn RJ, Hardy RW, Smith JL.
Characterization of purified Sindbis virus nsP4 RNAdependent
RNA polymerase activity in vitro. Virology. 2009;384(1):201208.
92. Shirako Y, Strauss EG, Strauss JH. Suppressor mutations that allow
Sindbis virus RNA polymerase to function with nonaromatic amino
acids at the Nterminus: evidence for interaction between nsP1
and nsP4 in minusstrand RNA synthesis. Virology.
2000;276(1):148160.
93. Khan AH, Morita K, Parquet MdC, Hasebe F, Mathenge EGM,
Igarashi A. Parquet Md Mdel C, Hasebe F, Mathenge EG, Igarashi
A. Complete nucleotide sequence of chikungunya virus and evi-
dence for an internal polyadenylation site. J Gen Virol. 2002;83(Pt
12):30753084. doi:10.1099/0022-1317-83-12-3075
94. Weiss B, Nitschko H, Ghattas I, Wright R, Schlesinger S. Evidence
for specificity in the encapsidation of Sindbis virus RNAs. J Virol.
1989;63(12):53105318.
95. Forsell K, Suomalainen M, Garoff H. Structurefunction relation of
the NH2terminal domain of the Semliki Forest virus capsid pro-
tein. J Virol. 1995;69(3):15561563.
96. Aggarwal M, Sharma R, Kumar P, Parida M, Tomar S. Kinetic
characterization of transproteolytic activity of Chikungunya virus
capsid protease and development of a FRETbased HTS assay. Sci
Rep. 2015;5:14753. doi:10.1038/srep14753srep14753
97. Snyder AJ, Mukhopadhyay S. The alphavirus E3 glycoprotein
functions in a cladespecific manner. J Virol. 2012;86(24):13609-
13620. doi:10.1128/JVI.01805-12JVI.01805-12
98. Smith TJ, Cheng RH, Olson NH, et al. Putative receptor binding
sites on alphaviruses as visualized by cryoelectron microscopy.
Proc Natl Acad Sci U. S. A. 1995;92(23):1064810652.
99. Voss JE, Vaney MC, Duquerroy S, et al. Glycoprotein organization
of Chikungunya virus particles revealed by Xray crystallography.
Nature. 2010;468(7324):709712. doi:10.1038/nature09555
100. Weber C, Berberich E, von Rhein C, Henss L, Hildt E, Schnierle BS.
Identification of functional determinants in the Chikungunya virus
E2 protein. PLoS Negl Trop Dis. 2017;11(1):e0005318. doi:10.1371/
journal.pntd.0005318PNTDD1600326
101. Snyder JE, Kulcsar KA, Schultz KL, et al. Functional characteriza-
tion of the alphavirus TF protein. J Virol. 2013;87(15):85118523.
doi:10.1128/JVI.00449-13JVI.00449-13
102. Simon F, Javelle E, Cabie A, et al. French guidelines for the man-
agement of chikungunya (acute and persistent presentations).
November 2014. Med Mal Infect. 2015;45(7):243263. doi:10.1016/
j.medmal.2015.05.007S0399077X(15)001444
103. Queyriaux B, Simon F, Grandadam M, Michel R, Tolou H, Boutin JP.
Clinical burden of chikungunya virus infection. Lancet Infect Dis.
2008;8(1):23. doi:10.1016/S1473-3099(07)70294-3
104. Borgherini G, Poubeau P, Jossaume A, et al. Persistent arthralgia
associated with chikungunya virus: a study of 88 adult patients on
reunion island. Clin Infect Dis. 2008;47(4):469475. doi:10.1086/
590003
105. Renault P, Josseran L, Pierre V. Chikungunyarelated fatality rates,
Mauritius, India, and Reunion Island. Emerg Infect Dis.
2008;14(8):13271327. doi:10.3201/eid1408.080201
106. Simon F, Javelle E, Oliver M, LeparcGoffart I, Marimoutou C.
Chikungunya virus infection. Curr Infect Dis Rep.
2011;13(3):218228. doi:10.1007/s11908-011-0180-1
107. MartiCarvajal A, RamonPardo P, Javelle E, et al. Interventions for
treating patients with chikungunya virus infectionrelated rheu-
matic and musculoskeletal disorders: a systematic review. PLoS
One. 2017;12(6):e0179028. doi:10.1371/journal.pone.0179028
108. Brighton SW, Simson IW. A destructive arthropathy following
Chikungunya virus arthritis–a possible association. Clin Rheumatol.
1984;3(2):253258.
109. Brito CA, Sohsten AK, Leitao CC, et al. Pharmacologic management
of pain in patients with Chikungunya: a guideline. Rev Soc Bras Med
Trop. 2016;49(6):668679. doi:10.1590/0037-8682-0279-2016
110. Schilte C, Staikowsky F, Couderc T, et al. Chikungunya virus
associated longterm arthralgia: a 36month prospective longitu-
dinal study. PLoS Negl Trop Dis. 2013;7(3):e2137. doi:10.1371/
journal.pntd.0002137
111. Ramachandran V, Kaur P, Kanagasabai K, Vadivoo S, Murhekar
MV. Persistent arthralgia among Chikungunya patients and asso-
ciated risk factors in Chennai, South India. J Postgrad Med.
2014;60(1):36. doi:10.4103/0022-3859.128795
112. Nimmannitya S, Halstead SB, Cohen SN, Margiotta MR. Dengue
and chikungunya virus infection in man in Thailand, 19621964. I.
Observations on hospitalized patients with hemorrhagic fever. Am
J Trop Med Hyg. 1969;18(6):954971.
113. Myers RM, Carey DE. Concurrent isolation from patient of two
arboviruses, Chikungunya and dengue type 2. Science.
1967;157(3794):13071308.
114. Carey DE, Myers RM, DeRanitz CM, Jadhav M, Reuben R. The
1964 chikungunya epidemic at Vellore, South India, including ob-
servations on concurrent dengue. Trans R Soc Trop Med Hyg.
1969;63(4):434445.
115. Halstead SB, Nimmannitya S, Margiotta MR. Dengue d chi-
kungunya virus infection in man in Thailand, 19621964. II.
Observations on disease in outpatients. Am J Trop Med Hyg.
1969;18(6):972983.
116. Ratsitorahina M, Harisoa J, Ratovonjato J, et al. Outbreak of dengue
and chikungunya fevers, Toamasina, Madagascar, 2006. Emerg Infect
Dis. 2008;14(7):11351137. doi:10.3201/eid1407.071521
117. Lanciotti RS, Kosoy OL, Laven JJ, et al. Chikungunya virus in US
travelers returning from India, 2006. Emerg Infect Dis.
2007;13(5):764767. doi:10.3201/eid1305.070015
118. Laurent P, Le Roux K, Grivard P, et al. Development of a sensitive
realtime reverse transcriptase PCR assay with an internal control
12 of 13
-
MANZOOR
ET AL.
to detect and quantify chikungunya virus. Clin Chem.
2007;53(8):14081414. doi:10.1373/clinchem.2007.086595
119. Panning M, Grywna K, van Esbroeck M, Emmerich P, Drosten C.
Chikungunya fever in travelers returning to Europe from the Indian
Ocean region, 2006. Emerg Infect Dis. 2008;14(3):416422.
doi:10.3201/eid1403.070906
120. Lakshmi V, Neeraja M, Subbalaxmi MV, et al. Clinical features and
molecular diagnosis of Chikungunya fever from South India. Clin
Infect Dis. 2008;46(9):14361442. doi:10.1086/529444
121. An W, Ge N, Cao Y, Sun J, Jin X. Recent progress on chikungunya
virus research. Virol Sin. 2017;32(6):441453. doi:10.1007/s12250-
017-4072-x
122. Couderc T, Khandoudi N, Grandadam M, et al. Prophylaxis and
therapy for Chikungunya virus infection. J Infect Dis.
2009;200(4):516523. doi:10.1086/600381
123. Levitt NH, Ramsburg HH, Hasty SE, Repik PM, Cole FE, Jr., Lupton
HW. Development of an attenuated strain of chikungunya virus for
use in vaccine production. Vaccine. 1986;4(3):157162.
124. GarciaArriaza J, Cepeda V, Hallengard D, et al. A novel poxvirus
based vaccine, MVACHIKV, is highly immunogenic and protects
mice against chikungunya infection. J Virol. 2014;88(6):35273547.
doi:10.1128/JVI.03418-13
125. Smalley C, Erasmus JH, Chesson CB, Beasley DWC. Status of
research and development of vaccines for chikungunya. Vaccine.
2016;34(26):29762981. doi:10.1016/j.vaccine.2016.03.076
126. Plante K, Wang E, Partidos CD, et al. Novel chikungunya vaccine
candidate with an IRESbased attenuation and host range alter-
ation mechanism. PLoS Pathog. 2011;7(7):e1002142. doi:10.1371/
journal.ppat.1002142
127. Bettadapura J, Herrero LJ, Taylor A, Mahalingam S. Approaches to
the treatment of disease induced by chikungunya virus. Indian J
Med Res. 2013;138(5):762765.
128. Brighton SW. Chloroquine phosphate treatment of chronic Chi-
kungunya arthritis. An open pilot study. S Afr Med J.
1984;66(6):217218.
129. Ravichandran R, Manian M. Ribavirin therapy for Chikungunya
arthritis. J Infect Dev Ctries. 2008;2(2):140142.
130. Rulli NE, Rolph MS, Srikiatkhachorn A, Anantapreecha S, Gugliel-
motti A, Mahalingam S. Protection from arthritis and myositis in a
mouse model of acute chikungunya virus disease by bindarit, an
inhibitor of monocyte chemotactic protein1 synthesis. J Infect Dis.
2011;204(7):10261030. doi:10.1093/infdis/jir470
131. Bassetto M, De Burghgraeve T, Delang L, et al. Computer
aided identification, design and synthesis of a novel series of
compounds with selective antiviral activity against chikungu-
nya virus. Antivir Res. 2013;98(1):1218. doi:10.1016/j.
antiviral.2013.01.002
132. White A, Berman S, Lowenthal JP. Comparative immunogenicities
of Chikungunya vaccines propagated in monkey kidney monolayers
and chick embryo suspension cultures. Appl Microbiol.
1972;23(5):951952.
133. Tiwari M, Parida M, Santhosh SR, Khan M, Dash PK, Rao PV.
Assessment of immunogenic potential of Vero adapted formalin
inactivated vaccine derived from novel ECSA genotype of Chi-
kungunya virus. Vaccine. 2009;27(18):25132522. doi:10.1016/j.
vaccine.2009.02.062S0264410X(09)002898
134. Edelman R, Tacket CO, Wasserman SS, Bodison SA, Perry JG,
Mangiafico JA. Phase II safety and immunogenicity study of live
chikungunya virus vaccine TSIGSD218. Am J Trop Med Hyg.
2000;62(6):681685. doi:10.4269/ajtmh.2000.62.681
135. Muthumani K, Lankaraman KM, Laddy DJ, et al. Immunoge-
nicity of novel consensusbased DNA vaccines against Chi-
kungunya virus. Vaccine. 2008;26(40):51285134. doi:10.1016/
j.vaccine.2008.03.060
136. Akahata W, Yang ZY, Andersen H, et al. A viruslike particle vaccine
for epidemic Chikungunya virus protects nonhuman primates against
infection. Nat Med. 2010;16(3):334338. doi:10.1038/nm.2105
137. Enserink M. Massive outbreak draws fresh attention to little
known virus. Am Assoc Advance Sci. 2006;311(5764):1085.
138. Young AR, Locke MC, Cook LE, et al. Dermal and muscle fibroblasts
and skeletal myofibers survive chikungunya virus infection and
harbor persistent RNA. PLoS Pathog. 2019;15(8):e1007993.
139. BonillaAldana D, Dhama K, RodriguezMorales A. Revisiting the
one health approach in the context of COVID19: a look into the
ecology of this emerging disease. Adv Anim Vet Sci.
2020;8(3):234237.
140. Baloch S, Baloch MA, Zheng T, Pei X. The coronavirus disease 2019
(COVID19) pandemic. Tohoku J Exp Med. 2020;250(4):271278.
doi:10.1620/tjem.250.271
141. Costa J, Ferreira EC, Santos C. COVID19, Chikungunya, Dengue
and Zika diseases: an analytical platform based on MALDITOF MS,
IR spectroscopy and RTqPCR for accurate diagnosis and accel-
erate epidemics control. Microorgnisms. 2021;9:708. doi:10.3390/
microorganisms9040708
142. Santana do Rosárioa M. Cristina de Siqueira I. Concerns about
COVID19 and arboviral (chikungunya, dengue, zika) concurrent
outbreaks. Braz J Infect Dis. 2020;24(6):583584. doi:10.1016/j.
bjid.2020.08.008
143. Shragai T, Tesla B, Murdock C, Harrington LC. Zika and chi-
kungunya: mosquitoborne viruses in a changing world. Ann N. Y
Acad Sci. 2017;1399(1):6177. doi:10.1111/nyas.13306
144. LaDeau SL, Allan BF, Leisnham PT, Levy MZ. The ecological foun-
dations of transmission potential and vectorborne disease in ur-
ban landscapes. Funct Ecol. 2015;29:889901. doi:10.1111/1365-
2435.12487
How to cite this article: Manzoor KN, Javed F, Ejaz M, et al.
The global emergence of Chikungunya infection: An integrated
view. Rev Med Virol. 2021;e2287. doi:10.1002/rmv.2287
MANZOOR ET AL.
-
13 of 13
... It was first reported in Tanzania in 1952, and several epidemics were reported in different parts of Africa and Asia. 1 The cases dwindled during 1970s until 2004, when it re-emerged in the Indian Ocean Islands and has expanded to over 60 countries across the globe. 1 In 1963, CHIKV first appeared in India and a few epidemics occurred till 1973. For 32 years, CHIKV was not reported until it re-appeared in 2005-2006 and since then, several outbreaks are being reported in different Indian states 1 across the country. ...
... It was first reported in Tanzania in 1952, and several epidemics were reported in different parts of Africa and Asia. 1 The cases dwindled during 1970s until 2004, when it re-emerged in the Indian Ocean Islands and has expanded to over 60 countries across the globe. 1 In 1963, CHIKV first appeared in India and a few epidemics occurred till 1973. For 32 years, CHIKV was not reported until it re-appeared in 2005-2006 and since then, several outbreaks are being reported in different Indian states 1 across the country. ...
Article
Full-text available
Background Chikungunya disease (CHIKD) is a threat to global health, as it impairs the quality of life of an infected individual ranging from months to years. A systematic evaluation of the serological, virological, and immunological aspects of the circulating viruses and their impact on the host response is imperative for better understanding of the evolving disease dynamics. Methods Serum samples were collected from 196 acute CHIKD patients from ten tertiary care hospitals across India during 2016–2021. Out of 196 patients, paired convalescent samples were collected from 51 patients (one-month post-onset of symptoms). The serum samples were profiled for cytokines and neutralisation capacity. Further, chikungunya virus (CHIKV) was isolated from the acute sera and the replication kinetics of the clinical isolates was evaluated. Findings Serological analysis indicated that neutralisation could be correlated to seroconversion in the convalescent phase but not found significant in acute phase. In the acute phase samples, there was a correlation between elevated serum levels of IFN-γ, IP-10, MCP-1 and MIG and disease severity. During convalescent phase, pro-inflammatory markers such as IL-6, IL-1β, IL-9 and IP-10 were found to be elevated with a corresponding decline in the secretion of anti-inflammatory cytokines such as IL-4 and IL-10, which correlated with persistent arthralgia. Analysis of replication of the clinical isolates revealed that 68.4% of viruses were fast-growing in the Vero cells (cytopathic effect [CPE] observed within 24 h post-infection), and their corresponding acute serum samples showed an elevated secretion of IFN-α, IL-1RA, IL-17F, IL-9, MCP-1 and MIP-1α. Interpretation This study provides an important overview of neutralisation capabilities and cytokine responses along with virus pathogenesis associated with CHIKV infections in India. Funding 10.13039/501100014825Biotechnology Industry Research Assistance Council (BIRAC).
... Management concentrates on alleviating symptoms and providing supportive care. Prevention relies on vector control measures to decrease mosquito populations and minimize human-mosquito contact, as well as raising awareness among communities about personal protection measures (4). The spread of CHIKV highlights the significance of robust surveillance, early detection, effective vector control, and public health education (5). ...
Article
Full-text available
This study addresses critical research gaps in Chikungunya virus (CHIKV) studies in Africa, proposing an AI-integrated approach. The study aims to leverage AI to enhance epidemiological surveillance, vector control, clinical management, community engagement, drug discovery, and data integration, within a One Health framework. The research gaps encompass inadequate real-time surveillance, limited vector knowledge, diagnostic challenges, low community awareness, slow drug development, and fragmented data. The study underscores AI's potential in early outbreak detection through data analysis and predictive modeling. It highlights AI's role in enhancing vector surveillance via image recognition and machine learning. AI-assisted diagnostics aid in accurate case identification. Moreover, AI-driven communication strategies can elevate community engagement. AI expedites drug discovery and vaccine development by predicting potential compounds. Data integration facilitated by AI harmonizes diverse datasets, encouraging interdisciplinary collaboration. The study advocates a One Health approach, recognizing the interdependence of human, animal, and environmental health. The study's insights provide a comprehensive roadmap to address CHIKV research gaps through AI, ultimately advancing public health outcomes in Africa.
... The Chikungunya virus (CHIKV) belongs to the Alphavirus genus within the Togaviridae family. Transmission to humans usually occurs through the bite of an infected mosquito, mainly Aedes aegypti and Aedes albopictus [5][6][7]. Genetic analyses of strains have identified ...
Article
Full-text available
Chikungunya (CHIK) is a re-emerging viral infection endemic in tropical and subtropical areas. While the typical clinical presentation is an acute febrile syndrome, long-term articular complications and even death can occur. This review characterizes the global epidemiological and economic burden of chikungunya. The search included studies published from 2007 to 2022 in MEDLINE, Embase, LILACS, and SciELO for a thorough evaluation of the literature. Rayyan software was used for data analysis, and data were summarized descriptively and reported following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Seventy-six publications were included. Chikungunya is widely distributed in the tropics, including Africa, Asia, South America, and Oceania/the Pacific Islands, and co-circulates with other simultaneous arboviruses such as DENV, ZIKV, and YFV. Chikungunya infection can lead to chronic articular manifestations with a significant impact on the quality of life in the long term. In addition, it generates absenteeism and economic and social losses and can cause fatal infections in vulnerable populations, mainly in high-risk patients with co-morbidities and at the extremes of age. Reported costs associated with CHIKV diseases are substantial and vary by region, age group, and public/private delivery of healthcare services. The chikungunya disease burden includes chronicity, severe infections, increased hospitalization risks, and associated mortality. The disease can impact the economy in several spheres, significantly affecting the health system and national economies. Understanding and measuring the full impact of this re-emerging disease is essential.
Preprint
Chikungunya (CHIKV), o’nyong-nyong (ONNV), and Mayaro (MAYV) viruses are transmitted by mosquitoes and known to cause a debilitating arthritogenic syndrome. These alphaviruses have emerged and re-emerged, leading to outbreaks in tropical and subtropical regions of Asia, South America, and Africa. Despite their prevalence, there persists a critical gap in the availability of sensitive and virus-specific point-of-care (POC) diagnostics. Traditional immunoglobulin-based tests such as enzyme-linked immunosorbent assay (ELISAs) often yield cross-reactive results due to the close genetic relationship between these viruses. Molecular diagnostics such as quantitative polymerase chain reaction (qPCR) offer high sensitivity but are limited by the need for specialized laboratory equipment. Recombinase polymerase amplification (RPA), an isothermal amplification method, presents a promising alternative to qPCR, providing rapid results with minimal equipment requirements. Here, we report the development and validation of three virus-specific RPA-based rapid tests for CHIKV, ONNV, and MAYV. These tests demonstrated both speed and sensitivity, capable of detecting 10 viral copies within 20 minutes of amplification, without exhibiting cross-reactivity to related alphaviruses and other arboviruses. Furthermore, we evaluated the clinical potential of these tests using serum and tissue samples from CHIKV, ONNV, and MAYV-infected mice. Ultimately, findings highlight the significance of these rapid and specific POC diagnostics in improving the early detection and management of these arboviral infections.
Article
Full-text available
Background The global spread of the chikungunya virus (CHIKV) increases the exposure risk for individuals travelling to or living in endemic areas. This Phase 3 study was designed to demonstrate manufacturing consistency between three lots of the single shot live-attenuated CHIKV vaccine VLA1553, and to confirm the promising immunogenicity and safety data obtained in previous trials. Methods This randomized, double-blinded, lot-to-lot consistency, Phase 3 study, assessed immunogenicity and safety of VLA1553 in 408 healthy adults (18–45 years) in 12 sites across the USA. The primary endpoint was a comparison of the geometric mean titre (GMT) ratios of CHIKV-specific neutralizing antibodies between three VLA1553 lots at 28 days post-vaccination. Secondary endpoints included immunogenicity and safety over 6 months post-vaccination. Results GMTs were comparable between the lots meeting the acceptance criteria for equivalence. The average GMT (measured by 50% CHIKV micro plaque neutralization test; μPRNT50) peaked with 2643 at 28 days post-vaccination and decreased to 709 at 6 months post-vaccination. An excellent seroresponse rate (defined as μPRNT50 titre ≥ 150 considered protective) was achieved in 97.8% of participants at 28 days post-vaccination and still persisted in 96% at 6 months after vaccination. Upon VLA1553 immunization, 72.5% of participants experienced adverse events (AEs), without significant differences between lots (related solicited systemic AE: 53.9% of participants; related solicited local AE: 19.4%). Overall, AEs were mostly mild or moderate and resolved without sequela, usually within 3 days. With 3.9% of participants experiencing severe AEs, 2.7% were classified as related, whereas none of the six reported serious adverse events was related to the administration of VLA1553. Conclusions All three lots of VLA1553 recapitulated the safety and immunogenicity profiles of a preceding Phase 3 study, fulfilling pre-defined consistency requirements. These results highlight the manufacturability of VLA1553, a promising vaccine for the prevention of CHIKV disease for those living in or travelling to endemic areas.
Article
Full-text available
Vector-borne viral diseases (VBVDs) continue to pose a considerable public health risk to animals and humans globally. Vectors have integral roles in autochthonous circulation and dissemination of VBVDs worldwide. The interplay of agricultural activities, population expansion, urbanization, host/pathogen evolution, and climate change, all contribute to the continual flux in shaping the epidemiology of VBVDs. In recent decades, VBVDs, once endemic to particular countries, have expanded into new regions such as Iran and its neighbors, increasing the risk of outbreaks and other public health concerns. Both Iran and its neighboring countries are known to host a number of VBVDs that are endemic to these countries or newly circulating. The proximity of Iran to countries hosting regional diseases, along with increased global socioeconomic activities, e.g., international trade and travel, potentially increases the risk for introduction of new VBVDs into Iran. In this review, we examined the epidemiology of numerous VBVDs circulating in Iran, such as Chikungunya virus, Dengue virus, Sindbis virus, West Nile virus, Crimean–Congo hemorrhagic fever virus, Sandfly-borne phleboviruses, and Hantavirus, in relation to their vectors, specifically mosquitoes, ticks, sandflies, and rodents. In addition, we discussed the interplay of factors, e.g., urbanization and climate change on VBVD dissemination patterns and the consequent public health risks in Iran, highlighting the importance of a One Health approach to further surveil and to evolve mitigation strategies.
Article
Aim: This work aimed to investigate the antiviral activity of two 1,4-disubstituted-1,2,3-triazole derivatives (1 and 2) against Chikungunya virus (CHIKV) replication. Materials & methods: Cytotoxicity was analyzed using colorimetric assays and the antiviral potential was evaluated using plaque assays and computational tools. Results: Compound 2 showed antiviral activity against CHIKV 181-25 in BHK-21 and Vero cells. Also, this compound presented a higher activity against CHIKV BRA/RJ/18 in Vero cells, like compound 1. Compound 2 exhibited virucidal activity and inhibited virus entry while compound 1 inhibited virus release. Molecular docking suggested that these derivatives inhibit nsP1 protein while compound 1 may also target capsid protein. Conclusion: Both compounds exhibit promising antiviral activity against CHIKV by blocking different steps of virus replication.
Article
Background: VLA1553 is a live-attenuated vaccine candidate for active immunisation and prevention of disease caused by chikungunya virus. We report safety and immunogenicity data up to day 180 after vaccination with VLA1553. Methods: This double-blind, multicentre, randomised, phase 3 trial was done in 43 professional vaccine trial sites in the USA. Eligible participants were healthy volunteers aged 18 years and older. Patients were excluded if they had history of chikungunya virus infection or immune-mediated or chronic arthritis or arthralgia, known or suspected defect of the immune system, any inactivated vaccine received within 2 weeks before vaccination with VLA1553, or any live vaccine received within 4 weeks before vaccination with VLA1553. Participants were randomised (3:1) to receive VLA1553 or placebo. The primary endpoint was the proportion of baseline negative participants with a seroprotective chikungunya virus antibody level defined as 50% plaque reduction in a micro plaque reduction neutralisation test (μPRNT) with a μPRNT50 titre of at least 150, 28 days after vaccination. The safety analysis included all individuals who received vaccination. Immunogenicity analyses were done in a subset of participants at 12 pre-selected study sites. These participants were required to have no major protocol deviations to be included in the per-protocol population for immunogenicity analyses. This trial is registered at ClinicalTrials.gov, NCT04546724. Findings: Between Sept 17, 2020 and April 10, 2021, 6100 people were screened for eligibility. 1972 people were excluded and 4128 participants were enrolled and randomised (3093 to VLA1553 and 1035 to placebo). 358 participants in the VLA1553 group and 133 participants in the placebo group discontinued before trial end. The per-protocol population for immunogenicity analysis comprised 362 participants (266 in the VLA1553 group and 96 in the placebo group). After a single vaccination, VLA1553 induced seroprotective chikungunya virus neutralising antibody levels in 263 (98·9%) of 266 participants in the VLA1553 group (95% CI 96·7-99·8; p<0·0001) 28 days post-vaccination, independent of age. VLA1553 was generally safe with an adverse event profile similar to other licensed vaccines and equally well tolerated in younger and older adults. Serious adverse events were reported in 46 (1·5%) of 3082 participants exposed to VLA1553 and eight (0·8%) of 1033 participants in the placebo arm. Only two serious adverse events were considered related to VLA1553 treatment (one mild myalgia and one syndrome of inappropriate antidiuretic hormone secretion). Both participants recovered fully. Interpretation: The strong immune response and the generation of seroprotective titres in almost all vaccinated participants suggests that VLA1553 is an excellent candidate for the prevention of disease caused by chikungunya virus. Funding: Valneva, Coalition for Epidemic Preparedness Innovation, and EU Horizon 2020.
Article
Full-text available
COVID-19 and arboviruses (ARBOD) epidemics co-occurrence is a great concern. In tropical and subtropical regions, ARBOD diseases such as chikungunya, dengue, and Zika are frequent. In both COVID-19 and ARBOD cases, an accurate diagnosis of infected patients is crucial to promote adequate treatment and isolation measures in COVID-19 cases. Overlap of clinical symptoms and laboratory parameters between COVID-19 and ARBOD present themselves as an extra challenge during diagnosis. COVID-19 diagnosis is mainly performed by quantitative reverse polymerase chain reaction (RT-qPCR), while ARBOD diagnosis is performed by serology, detection of antigen or antibody, and molecular diagnosis. In this review, the epidemiologic profile of arboviruses and SARS-CoV-2 is analyzed, and potential risks of symptom overlap is addressed. The implementation of an analytical platform based on infrared (IR) spectroscopy, MALDI-TOF mass spectrometry, and RT-qPCR is discussed as an efficient strategy for a fast, robust, reliable, and cost-effective diagnosis system even during the co-occurrence of virus outbreaks. The spectral data of IR spectroscopy and MALDI-TOF MS obtained from COVID-19 infected and recovered patients can be used to build up an integrated spectral database. This approach can enable us to determine quickly the groups that have been exposed and have recovered from COVID-19 or ARBOD, avoiding misdiagnoses.
Article
Full-text available
The present study provides an overview of the coronavirus disease 2019 (COVID-19) outbreak which has rapidly extended globally within a short period. COVID-19 is a highly infectious respiratory disease caused by a new coronavirus known as SARS-CoV-2 (severe acute respiratory syndrome-coronavirus-2). SARS-CoV-2 is different from usual coronaviruses responsible for mild sickness such as common cold among human beings. It is crucial to understand the impact and outcome of this pandemic. We therefore overview the changes in the curves of COVID-19 confirmed cases and fatality rate in China and outside of China from 31st of December 2019 to 25th of March 2020. We also aimed to assess the temporal developments and death rate of COVID-19 in China and worldwide. More than 414,179 confirmed cases of COVID-19 have been reported in 197 countries, including 81,848 cases in China and 332,331 outside of China. Furthermore, 18,440 infected patients died from COVID-19 infection; 3,287 cases were from China and 15,153 fatalities were reported worldwide. Among the worldwide infected cases, 113,802 patients have been recovered and discharged from different hospitals. Effective prevention and control measures should be taken to control the disease. The presented Chinese model (protocol) of disease prevention and control could be utilized in order to curb the pandemic situation.
Article
Full-text available
Chikungunya virus (CHIKV) is an arthritogenic alphavirus that acutely causes fever as well as severe joint and muscle pain. Chronic musculoskeletal pain persists in a substantial fraction of patients for months to years after the initial infection, yet we still have a poor understanding of what promotes chronic disease. While replicating virus has not been detected in joint-associated tissues of patients with persistent arthritis nor in various animal models at convalescent time points, viral RNA is detected months after acute infection. To identify the cells that might contribute to pathogenesis during this chronic phase, we developed a recombinant CHIKV that expresses Cre recombinase (CHIKV-3ʹ-Cre). CHIKV-3ʹ-Cre replicated in myoblasts and fibroblasts, and it induced arthritis during the acute phase in mice. Importantly, it also induced chronic disease, including persistent viral RNA and chronic myositis and synovitis similar to wild-type virus. CHIKV-3ʹ-Cre infection of tdTomato reporter mice resulted in a population of tdTomato+ cells that persisted for at least 112 days. Immunofluorescence and flow cytometric profiling revealed that these tdTomato+ cells predominantly were myofibers and dermal and muscle fibroblasts. Treatment with an antibody against Mxra8, a recently defined host receptor for CHIKV, reduced the number of tdTomato+ cells in the chronic phase and diminished the levels of chronic viral RNA, implicating these tdTomato+ cells as the reservoir of chronic viral RNA. Finally, isolation and flow cytometry-based sorting of the tdTomato+ fibroblasts from the skin and ankle and analysis for viral RNA revealed that the tdTomato+ cells harbor most of the persistent CHIKV RNA at chronic time points. Therefore, this CHIKV-3ʹ-Cre and tdTomato reporter mouse system identifies the cells that survive CHIKV infection in vivo and are enriched for persistent CHIKV RNA. This model represents a useful tool for studying CHIKV pathogenesis in the acute and chronic stages of disease.
Article
Full-text available
Chikungunya virus (CHIKV), an arbovirus belongs to the family Togaviridae and was discovered in Tanzania in year 953-54. In November 2016, an outbreak occurred in Karachi and approximately 30,000 individuals were infected with CHIKV. More than 4,000 cases were confirmed by qualitative reverse transcriptase polymerase chain reaction. However, actual numbers of cases are expected to rise. For the diagnosis of chikungunya virus, several methods including viral culture, detection of viral antigen, anti-CHIKV immunoglobulin M, immunoglobulin G antibodies and viral nucleic acid can be used. The recommended therapies include use of analgesics, antipyretic, antiinflammatory medications like paracetamol and nonsteroidal anti-inflammatory drugs (NSAIDs). In severe cases, where NSAIDs are not effective, disease modifying anti-rheumatic drugs (DMARDs) can also be used. For prevention, mosquito nets and mosquito repellents are vital and must therefore be used effectively.
Article
Full-text available
Chikungunya virus (CHIKV) is an arbovirus transmitted by Aedes mosquitos in tropical and subtropical regions across the world. After decades of sporadic outbreaks, it re-emerged in Africa, Asia, India Ocean and America suddenly, causing major regional epidemics recently and becoming a notable global health problem. Infection by CHIKV results in a spectrum of clinical diseases including an acute self-limiting febrile illness in most individuals, a chronic phase of recurrent join pain in a proportion of patients, and long-term arthralgia for months to years for the unfortunate few. No specific anti-viral drugs or licensed vaccines for CHIKV are available so far. A better understanding of virus-host interactions is essential for the development of therapeutics and vaccines. To this end, we reviewed the existing knowledge on CHIKV’s epidemiology, clinical presentation, molecular virology, diagnostic approaches, host immune response, vaccine development, and available animal models. Such a comprehensive overview, we believe, will shed lights on the promises and challenges in CHIKV vaccine development. Open image in new window
Article
Full-text available
Chikungunya is a severe and debilitating disease. Currently, Brazil is experiencing an epidemic caused by three arboviruses, which has changed the way health professionals have diagnosed and treated infected patients. The difficulty of diagnosis and the lack of a protocol for patient treatment, which fits Brazilian health system models, have made it difficult to manage this disease. It is necessary to implement a multidisciplinary network of patient care, in which primary care units play the main role. This review aims to present current information regarding the clinical aspects and treatment of Chikungunya virus infection.
Article
A comparative study was made of Formalin-inactivated Chikungunya vaccines prepared from the virus propagated in African green monkey kidney monolayers and concentrated chick embryo suspension cultures. The vaccine prepared in the chick embryo suspension cultures was significantly more protective to mice against a live homologous virus challenge and stimulated the production of 4 to 5 times more circulating antibodies than the vaccine prepared with virus grown in African green monkey kidney monolayer cultures.