You are on page 1of 61

Chikungunya Virus

(CHIKV)
VIRUS
A virus is a small collection of genetic code, either DNA or RNA, surrounded by a protein coat. A
virus cannot replicate alone. Viruses must infect cells and use components of the host cell to
make copies of themselves. Often, they kill the host cell in the process, and cause damage to the
host organism.

-National Human Genome Research Institute


GENE EXPRESSION
The virion RNA is infectious and serves as both genome and viral messenger RNA. The whole
genome is translated into a non-structural polyprotein which is processed by host and viral
proteases. Structural polyprotein is expressed through a subgenomic mRNA.

In alphaviruses, RdRp is expressed by suppression of termination at the end of 10% of nsP


polyproteins. Moreover a ribosomal frameshifting in the 6K region induces the translation of the
TF protein in alphaviruses.

REPLICATION
CYTOPLASMIC

1. Attachment of the viral E glycoprotein to host receptors mediates clathrin-mediated endocytosis of


virus into the host cell.
2. Fusion of virus membrane with host endosomal membrane. RNA genome is released into the
cytoplasm.
3. The positive-sense genomic ssRNA is translated into a polyprotein, which is cleaved into non-
structural proteins necessary for RNA synthesis (replication and transcription).
4. Replication takes place in cytoplasmic viral factories at the surface of endosomes. A dsRNA
genome is synthesized from the genomic ssRNA(+).
5. The dsRNA genome is transcribed/replicated thereby providing viral mRNAs/new ssRNA(+)
genomes.
6. Expression of the subgenomic RNA (sgRNA) gives rise to the structural proteins.
7. Capsid assembly occurs in cytoplasm.
8. The capsid is envelopped by budding at the plasma membrane where the virion exits the cell.
Outline of the Presentation
Introduction
Epidemiology
Pathogen: Chikungunya Virus
Molecular mechanisms
Clinical manifestations
Diagnostics
Treatment/Vaccine development
Biosafety and Prevention
Introduction - Chikungunya
•This virus was first isolated from patients and mosquitoes in the Newala district of Tanzania in
1952–1953
•Chikungunya : derived from a local term in Makonde language for the illness and means ‘that
which bends up’ (or to become contorted) referring to the crippling arthralgia and arthritis
associated with infection with this virus
•Arthropod-borne viral infection
•Caused widespread outbreaks on these two continents, Africa and Asia from the 1960s to the
1980s, before a period of relative quiescence over the following 20 years
•Humans are the primary host during epidemics
Epidemiology
ENDEMIC - refers to the constant presence and/or usual prevalence of a disease or infectious agent
in a population within a geographic area
SPORADIC - refers to a disease that occurs infrequently and irregularly
EPIDEMIC - refers to an increase, often sudden, in the number of cases of a disease above what is
normally expected in that population in that area
OUTBREAK - same definition with epidemic but more limited geographic area
PANDEMIC - refers to an epidemic that has spread over several countries or continents, usually
affecting a large number of people
SYLVATIC CYCLE OR ENZOOTIC CYCLE - the fraction of the pathogen population's lifespan spent
cycling between wild animals and vectors
AUTOCHTHONOUS TRANSMISSION - indigenous rather than descended from migrants or colonists
Epidemiology and Host Range
•The epidemiology of the virus differs in Africa and Asia
•AFRICA: transmitted in savannahs and forests by Aedes mosquitoes
•The most important vertebrate host in the cycle of infection is the non-human primate, e.g.
baboons and Cercopithecus monkeys
•Bushbabies and certain species of bats may also be infected in nature, but their role in viral
maintenance is likely to be of secondary importance
•A. furcifer, A. taylori, A. vittatus, A. fulgens, A. luteocephalus, A. dalzieli, A. vigilax, A.
camptorhyntites and A. africanus. Culex annulirostris and Mansonia uniformis are also
considered competent vectors
•ASIA: primarily from human to human by the vector Aedes aegypti
Epidemiology and Host Range
Transmitted by Aedes aegypti and Aedes albopictus
Blood-borne transmission is possible; cases have been documented among laboratory
personnel handling infected blood and a health care worker drawing blood from an
infected patient
Rare in utero transmission has been documented, mostly during the second trimester
Intrapartum transmission has also been documented when the mother was viremic
around the time of delivery.
 Studies have not found chikungunya virus in human milk
Epidemiology:
DISTRIBUTION AND OUTBREAKS
•Chikungunya virus was first identified in Tanzania in 1952 and for the following ~50 years was isolated and
caused occasional outbreaks in Africa and Asia. Since 2004, chikungunya has spread rapidly and been
identified in over 60 countries throughout Asia, Africa, Europe and the Americas
•2004 – KENYA: spread to surrounding locations in the Indian Ocean. In the two years following, around
500 000 cases were reported; on La Reunion Island, more than 1/3 of the population became infected. The
epidemic then spread from the Indian Ocean to India, where it persisted for a number of years, infecting
almost 1.5 million people. Viremic travelers saw the virus spread to Indonesia, Maldives, Sri Lanka, Myanmar
and Thailand.
•2007 – EUROPE: local transmission was reported for the first time in Europe, in a localized outbreak in north-
eastern Italy where 197 cases recorded. This outbreak confirmed that mosquito-borne outbreaks vectored
by Ae. Albopictus are plausible in Europe. 2010 saw the virus continue to cause illness in South East Asia, and
another outbreak was observed in La Reunion, in the Indian Ocean. Viremic travellers again imported the virus
into Europe, as well as the USA and Taiwan.
Epidemiology:
DISTRIBUTION AND OUTBREAKS
2013 – AMERICAS: he first documented outbreak of chikungunya with autochthonous transmission in the
Americas occurred; it started with two laboratory-confirmed, autochthonous cases were reported in the French
region of the Caribbean island of St Martin and it spread rapidly throughout the region. The European Centre
for Disease Prevention and Control (ECDC) in this same year were reported 72 cases, with France, the UK, and
Germany observing the most cases.
2014 – EUROPE: Europe faced its highest chikungunya burden, with almost 1,500 cases. Again France and the
UK were more affected. France also confirmed 4 cases of locally-acquired chikungunya infection in the south of
the country. Late that year, outbreaks were reported in the Pacific islands including the Cook Islands, Marshall
Islands, Samoa, American Samoa, French Polynesia and Kiribati. More than 1 million suspected cases were also
reported to the Pan American Health Organization (PAHO) regional office that year.
Epidemiology:
DISTRIBUTION AND OUTBREAKS
2015 - ECDC reported a decline in chikungunya cases from the 2014, down to 624 cases. WHO’s African
Regional Office (AFRO) recorded an outbreak in Senegal, representing the first active circulation in the area in
five years. In the Americas, there were 693 489 suspected and 37 480 confirmed cases of chikungunya
reported to the Pan American Health Organization (PAHO) regional office, of which Colombia had the highest
burden with 356 079 suspected cases. This burden in the Americas however, was significantly less than in the
previous year.
2016 - there was a total of 349 936 suspected and 146 914 laboratory-confirmed cases reported to the
PAHO regional office, being half the burden compared to the previous year. Countries reporting most cases
were Brazil, Bolivia and Colombia (with around 300 000 suspected cases between them). Argentina reported
the first evidence of autochthonous transmission of chikungunya, following an outbreak of more than 1 000
suspected cases. In Africa, Kenya reported an outbreak of chikungunya resulting in more than 1 700 suspected
cases, while in Somalia, the town of Mandera was hard hit, with about 80% of the population affected by
chikungunya. Chikungunya cases in India were approaching 65 000. European case reports remained below
500.
Epidemiology:
DISTRIBUTION AND OUTBREAKS
2017 - ECDC reported a total of 10 countries, with 548 cases with chikungunya, of which 84% were
confirmed cases. Italy bore more than 50% of the chikungunya burden. Autochthonous cases were again
reported in Europe (France and Italy) for the first time since 2014.

As in previous years, Asia and the Americas were the regions most affected by chikungunya. Pakistan faced a
persistent outbreak that started the previous year and reported 8 387 cases, while India suffered with 62 000
cases. In the Americas and the Caribbean were reported 185 000 cases; the cases in Brazil accounted for
>90% in the region of the Americas. Chikungunya outbreaks were also reported in Sudan (2018), Yemen
(2019) and more recently in Cambodia and Chad (2020)
Epidemiology:
DISTRIBUTION AND OUTBREAKS
PHILIPPINES: 2018 DATA OF DEPARTMENT OF HEALTH EPIDEMIOLOGY BUREAU
A total 282 chikungunya cases were reported nationwide from January 1 to March 3, 2018 (Figure 1). This is 6%
higher compared to the same time period last year (266).
Epidemiology:
DISTRIBUTION AND OUTBREAKS
Epidemiology:
DISTRIBUTION AND OUTBREAKS
Epidemiology:
DISTRIBUTION AND OUTBREAKS
CHIKUNGUNYA VIRUS
Genus Alphavirus of the Togaviridae Family
Belongs to the Semliki Forest virus antigenic complex that also contains the O’Nyong Nyong,
Mayaro, and Ross River viruses
Enveloped positive-strand RNA virus, with a genome of about 12 kb.
The genome is capped in 5′ and has a polyA tail in the 3′ end
Encodes four nonstructural proteins (nsP1 to nsP4) and five structural proteins (C-E3-E2-6 k-
E1)
 Genetic analysis based on the E1 envelope glycoprotein sequences showed three distinct
lineages: the West African cluster, the East-Central and South African cluster (ECSA), and the
Asian cluster
Electron Micrographs of CHIKV
CHIKUNGUNYA VIRUS
Chikungunya virus is a small (about 60–70 nm-
diameter), spherical, enveloped, positive-strand RNA
virus that is approximately 11 kb in length and
codes for 9 proteins. The genome has two open
reading frames (ORFs): the 5´ORF, translated from
genomic RNA, encodes the nsP1, nsP2, nsP3, and
nsP4 non-structural proteins, and the 3´ORF,
genome is transcribed for subsequent translation
into a polyprotein precursor containing the three
structural proteins PE2 (the precursor of E3 and E2),
E1, and the capsid protein.
International Journal of Technical Innovation in Modern
Engineering & Science (IJTIMES) “Research Symposium
on Advancements in Engineering, Science, Management,
and Technology Volume 5, Special Issue 04, April-2019
VIRAL STRUCTURE: Viral RNA
Viral RNA
•The 5'-proximal two-thirds codes for the nonstructural
proteins required for replication and transcription
•The 3'-proximal one-third codes for the capsid and envelope
proteins
VIRAL STRUCTURE: nsPs and SPs
Protein Size (a.a.) Function
Nonstructural Proteins
nsP1 535 Methylase transferase and guanylytransferase activity that caps viral RNA; sole membrane anchor for replicase complex;
required for initiation of transcription
nsP2 798 N-terminal NTPase, helicase and RNA triphosphatase activities; C-terminal cysteine protease activity responsible for
processing of nonstructural polyproteins; Required for synthesis of subgenomic RNA; Inhibits the transcription of cellular
mRNAs
nsP3 530 Phosphoprotein with unknown functions, but important for minus-strand synthesis; contains macro domain and SH3-
binding regions; likely interacts with host proteins
nsP4 611 RNA-dependent RNA polymerase (RdRp); putative terminal transferase activity
Structural Proteins
Capsid 261 Encapsidates genomic RNA to form nucleocapsid core; carboxyl domain is an autocatalytic serine protease
pE2 487 Intermediate composed of E3 and E2; cleaved by host furin protease
E3 64 N-terminal domain is uncleaved leader peptide for E2; may held shield fusion peptide in E1 during egress
E2 423 Mediates binding to receptors and attachment factors on cell membrane; major target of neutralizing antibodies
6K 61 Leader peptide for E1; putative ion channel; may enhance particle release
TF 76 Transframe protein generated by ribosomal frameshifting; shares N-terminus with 6K; putative ion channel; may enhance
particle release
E1 439 Type II fusion protein; mediates fusion of viral envelope and cellular membrane via fusion peptide
CHIKUNGUNYA VIRUS
CHIKV could be detected in connective tissue (especially in epimysium), in muscle, joint, and
skin fibroblasts, and even in the central nervous system (CNS) in some severe mouse infections
but not in fetal or placental tissues
Cynomolgus macaque model: In this model, at the early stage of the disease, the organs
targeted for CHIKV replication were lymphoid tissues, liver, CNS, joints, and muscle, and the
persistence of CHIKV could be found later in the lymphoid organs, liver, joints, and muscle,
macrophages being the main reservoir for persistent CHIKV infection.
In humans, acute CHIKV infection is characterized by a very early viremia at fever onset that can
increase up to 109 to 1012 RNA copies/mL and lasts up to 12 days.
The most significant event in CHIKV history was the appearance of an adaptive mutation,
an alanine-to-valine substitution at position 226 in the E1 glycoprotein gene (E1:A226V) on
an ESCA-CHIKV strain circulating on Reunion Island after September 2005. It led the
mutated CHIKV to lose cholesterol dependence for growth and enhanced its infectivity,
replication, and transmission by Ae. albopictus, without impairing common vectorial
capability of Ae. aegypti [15, 16]. Similar genetic events occurred independently in India,
Gabon, and Cameroon, suggesting an evolutionary convergence of the virus to this
mosquito and a great subsequent ability for worldwide epidemic expansion [13, 17, 18]. To
date, no difference in virulence between the different strains of CHIKV has been shown in
humans.
A defective cell-mediated immune response (CMI) where CD8+ T-lymphocytes are absent or
inactive has been thought to be the cause of chronic disease and viral persistence.
Oversecretion of toxic chemokines or apoptosis are postulated as causes of cell/tissue
destruction and associated clinical symptoms.[8]
An antibody-dependent enhancement (ADE) mechanism similar to that suggested for dengue
viruses[35] has also been implicated in the pathogenesis.
Molecular Mechanisms of ENTRY to EGRESS
Alphavirus entry is mainly mediated by clathrin-mediated endocytosis (CME) with the requirement of a low
endosomal pH and the integrity of the early endosome compartment to productively infect human cells
CHIKV is capable of productively infecting fibroblasts, keratinocytes, melanocytes, epithelial, and endothelial
cells and to a lesser extent macrophages (Surisseau et al., 2007; Matusali et al., 2019)
CHIKV HAS BROAD TROPISM
Receptors of CHIKV:
MXRA8 / Matrix remodeling-associated protein 8 / Lamitrin
•interacts with envelope protein of CHIKV
•Considered as the first bonafide CHIKV receptor (Zhang et al., 2018, 2019; Basore et al., 2019; Song et al.,
2019; Kim et al., 2020)
•Not present to all permissive cells
Molecular Mechanisms of ENTRY to EGRESS
PHB-1 / Prohibitin 1
•Interacts with chikungunya virus spike glycoprotein E2
CD147
•highly glycosylated protein and broadly expressed on human cell types making it is an interesting target for
binding by pathogens
Molecular Mechanisms of ENTRY to EGRESS
I. Interaction of E2 to the mammalian cell receptor
II. Entry of the virus via CLATHRIN-MEDIATED ENDOCYTOSIS
III. Fusion of the viral envelope and endosomal membrane releases nucleocapsid into the cytosol
IV. Disassembly of the nucleocapsid liberates positive-sense genomic RNA and nonstructural protein (nsP)
translation occurs
V. Four nsPs, together with genomic RNA and presumably host proteins, assemble at the plasma membrane (PM)
and modify it to form viral replication compartments (spherules) containing viral dsRNA. nsP1–4 function as a
replicase and localize to the spherule neck to generate genomic, antigenomic, and subgenomic vRNAs.
VI. Spherule internalization allows formation of large cytopathic vacuoles (CPV-1) that house multiple spherules.
VII. Translation of subgenomic RNA produces the structural polyprotein, and capsid autoproteolysis releases free
capsid into the cytoplasm. Translocation of E3-E2-6K-E1/E2-E2-TK polyproteins into the ER. E2/E1 are
posttranslationally modified, transit the secretory system, and are deposited at the PM.
Molecular Mechanisms of ENTRY to EGRESS
VIII. Interaction of capsid and genomic RNA leads to formation of icosahedral nucleocapsids.
IX. Nucleocapsids assemble with E2/E1 at the PM, resulting in budding of mature progeny
virions.
X. Later in infection, CPV-IIs form, containing hexagonal lattices of E2/E1 and are studded with
nucleocapsids.
XI. CPV-IIs likely serve as transport vehicles and assembly sites for structural proteins, allowing
formation of mature virions and egress.
CLINICAL MANIFESTATIONS
INCUBATION PERIOD IS 2 – 15 DAYS
With a mosquito bite, viral particles are deposited in the skin, from where they eventually reach the lymph nodes and
then the blood flow. They are then distributed to target organs that include joints, muscle, and skin. With less
frequency, the virus can affect the liver or cause encephalopathy, encephalitis, myocarditis, and heart blockage.
Although 5-15% of individuals suffer from asymptomatic infection, the rest generally develop symptoms that progress
from the acute to the chronic phase
I. Acute phase
•The acute phase lasts 3-10 days and is characterized by the abrupt presentation of fever (≥ 39 °C). Other symptoms
follow during the next few days
•Joint pain is reported in 87-98% of the cases
•Joint pain exists in more than one joint and is usually bilateral (symmetrical), occurring mainly in the peripheral joints
(knees, ankles, hands, wrists)
CLINICAL MANIFESTATIONS
•There is myalgia in 46-72% of the cases, affecting arms, thighs, and calves.
•This clinical picture is called chikungunya fever (CHIKF) and tends to be limiting an disabling in relation to the
normal physical activity of individuals
•The exanthema that appears in 40-50% of cases is of the petechial or maculopapular type, mainly affecting
the limbs, trunk, and face, and can cause pruritus. The lesions are transitory and generally appear 2-5 days
after disease onset
•Other symptoms that are less common are diarrhea, vomiting, edema on limbs, bleeding, otitis, and ocular
disease (especially anterior uveitis).
• Some of the more severe manifestations, which are infrequently found, include neurological diseases such
as meningoencephalitis, the Guillain-Barré syndrome, myocarditis, and multiorgan failure.
CLINICAL MANIFESTATIONS
II. Chronic phase
In adults, these conditions can last months or even years after the infection, while they are less
common in children
•Musculoskeletal disorders include arthralgia, inflammatory arthritis, polyarthralgia,
tenosynovitis, enthesitis, and exacerbation of existing rheumatic disease
•Less common symptoms include neuropathy, cerebral disorder, neurosensory deficiency, burning
mouth syndrome, paresthesia, cubital tunnel syndrome, gastrointestinal disorders, exanthema,
pruritus, bursitis, and synovitis
CLINICAL MANIFESTATIONS
CHIKUNGUNYA IN NEONATES
•CHIKF can be accompanied by convulsions, peripheral cyanosis, podalic edema, and epithelial
vesicular lesions that eventually dry and scale.
•Atypical findings: neurological manifestations have been reported that include febrile
convulsions, meningeal syndrome, acute encephalopathy, diplopia, aphasia, acute disseminated
encephalomyelitis, and encephalitis.
•The development of chronic arthralgia and exacerbation of underlying medical conditions is
unusual
CLINICAL MANIFESTATIONS
•CONGENITAL INFECTIONS
•Pregnant women with detectable viremia a few days before childbirth can transmit CHIKV to the
newborn, which can result in a severe neonatal disease, generally encephalopathy followed by
neurodisability.
•Other signs and symptoms observed in newborns are convulsions, thrombocytopenia,
hypotension, ventricular dysfunction, pericarditis, hyperechoic coronary arteries, parenchymal
hemorrhage, and cytotoxic edema.
•It has been demonstrated that maternal-fetal transmission occurs due to contact of the product
with infected maternal blood during childbirth
The infection is initially very rapid and the virus
is typically eliminated 5-7 days after onset of
the fever. During the infection there is a robust
and rapid activation of dendritic, NK/ CD4+ and
CD8+ cells. Persistently high levels of IL-12 are
mainly found in individuals with chronic
symptoms, mostly in persons with rheumatoid
arthritis, as inflammation provoked by CHIKV
infection can cause bone erosion and severe
arthralgia
Dengue Vs. Chinkungunya
LABORATORY FINDINGS
•Lymphopenia (500-1000 cells/mm3)
•moderate thrombocytopenia (100,000-150,000 cells/mm3)
•Leukopenia
•Elevation of hepatic enzyme levels
•Anemia
•Elevated creatinine level
•Hypocalcemia
•Laboratory tests are negative for the rheumatoid factor and the antibodies for cyclic citrullinated
peptides (CCP)
•Radiographic studies show bone erosion, swollen joints and synovitis
DIAGNOSTICS
DIAGNOSTICS
Virus Isolation/Culture - biosafety level (BSL) 3
GOLD STANDARD FOR THE DIAGNOSIS OF CHIKUNGUNYA FEVER IS VIRAL CULTURE
RT-PCR - Highly sensitive and specific; targets envelope and non structural proteins
Serologic Tests
1. ELISA
•Antigen Capture ELISA: Monoclonal antibody (Mab) based Antigen capture ELISA to detect Chikungunya virus antigen
from the mosquitoes
1. IFA
2. PRNT TEST - Plaque Reduction Neutralization Test : Serological detection of neutralizing antibodies
3. RAPID DETECTION TEST KITS - Sandwich Immunoassay principle
Rapid Immunochromatographic Test: Rapid diagnostic test using mouse Monoclonal antibodies that react with
CHIKV E1 proteins
NUCLEIC ACID AMPLIFICATION TESTS
•STANDARD POLYMERASE CHAIN REACTION
•REAL TIME REVERSE TRANSCRIPTASE PCR
oSingle/multiplex format
oSYBR green
•NASBA or NUCLEIC ACID SEQUENCE BASED AMPLIFICATION
oEcl
oMolecular beacons
•LAMP OR LOOP-MEDIATED ISOTHERMAL AMPLIFICATION
PRNT
The PRNT is a specific assay that may be used to document the presence of neutralizing
antibodies specific for a particular arbovirus like Chikungunya virus. Serum samples are
incubated with virus, and if viral neutralizing antibodies are present, they will bind to the virus
and prevent viral infection of cultured cells causing a reduction in the number of plaques
detected. The neutralizing titre of a sample is expressed as the reciprocal of the serum dilution
at which there is a 90% reduction in the number of plaques detected.
TREATMENT
•RIBAVIRIN – Antiviral activity against RNA viruses
•CHLOROQUINE - Inhibits CHIKV infection in cell culture thru effects on endosomal modification
•MONOCLONAL ANTIBODY - Passive transfer of CHIKV immune serum protects against CHIKV-
induced lethality in mouse models
•SUPPORTIVE CARE
VACCINE DEVELOPMENT
•Formalin inactivated (Walter Reed) Phase II
•Live attenuated vaccine Phase II
•Chimeric alpha virus approach Phase I
•Virus like particle in Pre-clinical trials
PREVENTION
1. VECTOR CONTROL
•Reducing the number of breeding sites by eliminating all stagnant water reserves in and near homes;
• Application of larvicidal treatments including biopesticide: Bacillus thuringiensis israelensis
•Control of the adult vector by aerial application of adulticide insecticide (organophosphorus or synthetic pyrethroids)
2. PHYSICAL PROTECTION – Use of repellants, long clothing, mosquito nets
•Wear neutral-coloured (beige, light grey) clothing. If possible, wear long-sleeved, breathable garments.
•If available, pre-soak or spray outer layer clothing and gear with permethrin
•The repellents must contain DEET (N,N-diethyl-3-methylbenzamide), IR3535 [3-(N-acetyl-N-butyl)-aminopropionic acid ethyl
ester], or icaridine [1- piperidinecarboxylic acid, 2-(2-hydroxyethyl)-1-methylpropyl ester]
3. SURVEILLANCE
4. OUTBREAK RESPONSE
5. RESEARCH
BIOSAFETY
•Risk Group 3
•Biosafety level 3 - Research should be conducted using Biosafety Level 3 practices, equipment,
and facility design
•Animal studies may be performed at ABSL-3
•Viability: Susceptible to common disinfectants: 70% ethanol, 1% sodium hydrochloride, 2%
glutaraldehyde, and > 60° C
•PPE Personal protective equipment includes but is not limited to laboratory coats or gowns,
disposable gloves, safety glasses, face shield if risk of splash, and respirator (PAPR or N-95) if
aerosol risk.
RISK GROUP 3
Agents that are associated with serious or lethal human
disease for which preventive or therapeutic interventions
may be available. These agents represent a high risk to an
individual but a low risk to the community.
REFERENCES
• US CDC
• CDC – Division of Vector Borne Infectious Diseases
• DOH – Epidemiology Bureau
• DOH – Research Institute for Tropical Medicine
• Updates on Vector-Borne Viruses: Chikungunya by Dr. Salvacion Gatchalian (2018)
• https://www.iamat.org/country/philippines/risk/chikungunya
• Chikungunya Vaccines in the Pipeline (who.int)
• https://www.uniprot.org/
• The CD147 Protein Complex Is Involved in Entry of Chikungunya Virus and Related Alphaviruses in Human Cells by Caluwe et. al
• Recent Trends in Chikungunya Virus diagnosis by Niranjan and Gupta
• Boston University: https://www.bu.edu/researchsupport/safety/rohp/agent-information-sheets/chikungunya-virus-agent-information-sheet/
• Chikungunya Virus Infection (2011) by Simon et.al. doi: 10.1007/s11908-011-0180-1
• McIntosh, B. M., Sweetnam, J., McGillivray, G. M., & De Sousa, J. (1965). Laboratory transmission of Chikungunya virus byMansonia (Mansonioides)
africana(Theobald). Annals of Tropical Medicine & Parasitology, 59(4), 390–392. doi:10.1080/00034983.1965.1168632
REFERENCES
•Telles, J.-N., Le Roux, K., Grivard, P., Vernet, G., & Michault, A. (2009). Evaluation of real-time nucleic acid
sequence-based amplification for detection of Chikungunya virus in clinical samples. Journal of Medical
Microbiology, 58(9), 1168–1172. doi:10.1099/jmm.0.010736-0
•Wichit, S., Gumpangseth, N., Hamel, R., Yainoy, S., Arikit, S., Punsawad, C., & Missé, D. (2021). Chikungunya and
Zika Viruses: Co-Circulation and the Interplay between Viral Proteins and Host Factors. Pathogens, 10(4), 448.
doi:10.3390/pathogens10040448
•Yap, M. L., Klose, T., Urakami, A., Hasan, S. S., Akahata, W., & Rossmann, M. G. (2017). Structural studies of
Chikungunya virus maturation. Proceedings of the National Academy of Sciences, 114(52), 13703–
13707. doi:10.1073/pnas.1713166114
•Silva, L. A., & Dermody, T. S. (2017). Chikungunya virus: epidemiology, replication, disease mechanisms, and
prospective intervention strategies. Journal of Clinical Investigation, 127(3), 737–749. doi:10.1172/jci84417
•Snyder, J. E., Kulcsar, K. A., Schultz, K. L. W., Riley, C. P., Neary, J. T., Marr, S., … Kuhn, R. J. (2013). Functional
Characterization of the Alphavirus TF Protein. Journal of Virology, 87(15), 8511–8523. doi:10.1128/jvi.00449-13
•Tsetsarkin, K. A., Vanlandingham, D. L., McGee, C. E., & Higgs, S. (2007). A Single Mutation in Chikungunya Virus
Affects Vector Specificity and Epidemic Potential. PLoS Pathogens, 3(12), e201. doi:10.1371/journal.ppat.0030201
•Kendall, C., Khalid, H., Müller, M., Banda, D. H., Kohl, A., Merits, A., … Tuplin, A. (2019). Structural and phenotypic
analysis of Chikungunya virus RNA replication elements. Nucleic Acids Research. doi:10.1093/nar/gkz640
•Changing patterns of chikungunya virus: re-emergence of a zoonotic arbovirus.Powers AM, Logue CHJ Gen Virol.
2007 Sep; 88(Pt 9):2363-2377.

•Fundamentals of Molecular Virology (2nd Edition) by Acheson


•Principles and Practice of Clinical Virology (5th Edition) by Zuckerman et.al.
•Emerging and Reemerging Viral Pathogens Volume 1 and 2 by M.M. Ennaji
•Red Book Atlas of Pediatric Infectious Diseases (3rd Edition) by C. Baker
Immediately refrigerate at 2-
8 degree Celsius while
· Collect in plain or red top tube.
Serology: ELISA IgM awaiting transport
7 days after onset of · Allow to clot
Serum 1 ml Thursday Wednesday 10 days 1,500.00
fever · Centrifuge
Chikungunya (CHIKV) Transport in ice to be
· Separate serum from blood
received at RITM within 72
hours

Immediately refrigerate at 2-8


· Collect in plain or red top tube. degree Celsius while awaiting
Molecular test for 7 days after onset of · Allow to clot transport
Serum 1 ml Tue/Thu Mon/Wed 7 days 4,000.00
Chikungunya (CHIKV) fever · Centrifuge
· Separate serum from blood Transport in ice to be received
at RITM within 72 hours
THANK YOU FOR LISTENING!

You might also like