Abstract
Alphaviruses, members of the enveloped, positive-sense, single-stranded RNA Togaviridae family, represent a reemerging public health threat as mosquito vectors expand into new geographic territories. The Old World alphaviruses, which include chikungunya virus, Ross River virus, and Sindbis virus, tend to cause a clinical syndrome characterized by fever, rash, and arthritis, whereas the New World alphaviruses, which consist of Venezuelan equine encephalitis virus, eastern equine encephalitis virus, and western equine encephalitis virus, induce encephalomyelitis. Following recovery from the acute phase of infection, many patients are left with debilitating persistent joint and neurological complications that can last for years. Clues from human cases and studies using animal models strongly suggest that much of the disease and pathology induced by alphavirus infection, particularly atypical and chronic manifestations, is mediated by the immune system rather than directly by the virus. This review discusses the current understanding of the immunopathogenesis of the arthritogenic and neurotropic alphaviruses accumulated through both natural infection of humans and experimental infection of animals, particularly mice. As treatment following alphavirus infection is currently limited to supportive care, understanding the contribution of the immune system to the disease process is critical to developing safe and effective therapies.
1. Introduction to alphaviruses
The Alphaviruses are a genus in the Togaviridae family, a group of enveloped, positive-sense, single-stranded RNA viruses that are found worldwide (Griffin, 2013) (Fig. 1). Alphaviruses of clinical importance in humans are transmitted by mosquitos, typically species from the Culex and Aedes genera, and represent a re-emerging public health threat as the insect vectors expand into new territories. The viruses circulate in sylvatic cycle between mosquitoes and either birds, nonhuman primates (NHP), or small mammals (Lim et al., 2018). However, promiscuous mosquitoes harboring an alphavirus sometimes feed on humans, creating an urban cycle that can result in massive widespread epidemics. Alphaviruses are divided into two major groups, Old World, and New World, which is based on their geographic distribution and typical clinical disease syndrome induced (Table 1).
Fig. 1.
Geographic range of cases of human disease induced by select Old World (CHIKV, RRV, and SINV) and New World (VEEV, EEEV, and WEEV) alphaviruses. CHIKV = chikungunya virus, RRV = Ross River virus, SINV = Sindbis virus, VEEV = Venezuelan equine encephalitis virus, EEEV = eastern equine encephalitis virus, WEEV = western equine encephalitis virus.
Table 1.
Clinical disease syndromes induced by natural alphavirus infection in humans.
Disease syndrome | Clinical signs and symptoms | Associated alphaviruses |
---|---|---|
Arthritogenic disease |
|
Old World alphaviruses:
|
Neurologic disease |
|
New world alphaviruses
|
The Old World alphaviruses, which include chikungunya virus (CHIKV), Ross River virus (RRV), o’nyong-nyong virus, (ONNV), Mayaro virus (MAYV), and Sindbis virus (SINV), are typically found in Africa, Asia, Europe, and Oceania and induce an arthritogenic disease characterized by fever, rash, and arthralgia (reviewed in Suhrbier et al., 2012). CHIKV was initially identified in 1952 in present-day Tanzania but has been responsible for sporadic epidemics of fever and arthralgia, commonly known as chikungunya fever, across Africa and Asia since the 1960s (Robinson, 1955; Ross, 1956; Zeller et al., 2016). The virus expanded throughout the Indian Ocean region in the mid-2000s, and in a separate outbreak was introduced into the Caribbean in 2013. The virus resulted in over 1 million new cases within a year of arrival in the Americas and has become endemic in Central and South America and the Caribbean (Yactayo et al., 2016; Zeller et al., 2016). Because of ease of human travel, adaptive mutations in CHIKV, and spread of Aedes spp. mosquito vectors, cases of CHIKV infection have now been reported in over 40 countries (Tsetsarkin et al., 2007; Yactayo et al., 2016; Zeller et al., 2016). Closely related to CHIKV, ONNV is responsible for periodic outbreaks of fever and arthralgia across Central and East Africa (Forrester et al., 2012; Rezza et al., 2017). First isolated from mosquitos in Queensland, Australia in 1959, RRV is now endemie in Australia and several Pacific Islands and is responsible for approximately 4000–8000 cases each year in Northern Australia (Endy, 2020; Mylonas et al., 2002). Though part of the Semliki Forest phylogenetic complex that includes RRV and therefore classified as an Old World alphavirus, MAYV is native to and responsible for increasingly frequent outbreaks of arthritogenic disease in Latin America, having first been isolated in Trinidad in 1954 (Anderson et al., 1957; Forrester et al., 2012; Ganjian and Riviere-Cinnamond, 2020). The SINV strain group is the most widely distributed alphavirus, being found across Europe, Asia, Africa and Oceania (Lundström and Pfeffer, 2010). However, most cases of arthritis and arthralgia are reported in Scandinavia, caused by the Ockelbo, Pogosta, or Karelian strains of SINV, or South Africa (Laine et al., 2004; Storm et al., 2014). While clinical disease induced by the Old World alphaviruses is typically self-limiting and rarely causes death, persistent chronic arthralgia and arthritis lasting months after recovery from acute illness is common and can be debilitating (Alla and Combe, 2011).
The New World alphaviruses, which include Venezuelan equine encephalitis virus (VEEV), Eastern equine encephalitis virus (EEEV), and Western equine encephalitis virus (WEEV), are found in North, Central, and South America and typically induce neurological disease (Hollidge et al., 2010). EEEV used to be subdivided into North American and South American variants, but recent ecologic and genetic studies have determined the variants belong to distinct separate lineages (Arrigo et al., 2010). South American EEEV, which readily infects a wide variety of mammals but has only been attributed to about a dozen human cases of encephalitis, has since been renamed Madariaga virus, while members of the North American EEEV lineage have retained the EEEV name (Carrera et al., 2013; Luciani et al., 2015). The sequences of the capsid and 3′ untranslated region of WEEV is similar to EEEV, but the El and E2 glycoprotein sequences are more closely related to the Old World alphavirus SINV, indicating that WEEV resulted from a recombination event (Hahn et al., 1988). VEEV, EEEV, and WEEV typically circulate between mosquitoes and passerine birds in the wild; however, promiscuity by permissive mosquitoes occasionally results in infection of other species, particularly humans and horses (Go et al., 2014). These large mammals are typically dead-end hosts, though VEEV is capable of replicating at high enough titers in the blood to permit continued transmission (Weaver et al., 2004).
Human cases of Venezuelan equine encephalitis (VEE), eastern equine encephalitis (EEE), and western equine encephalitis (WEE) are only sporadically reported in the United States each year, though outbreaks of EEE and VEE over the last few decades have increased and are being diagnosed in previously unreported locations. An outbreak of VEE in the mid-1990s in Venezuela and Columbia affected an estimated 75,000–100,000 people (Weaver et al., 1996), and the number of human cases of EEE in the northeastern United States has markedly increased in the last decade, with states such as Vermont and Maine reporting locally acquired human cases for the first time (Lubelczyk et al., 2013; Molaei et al., 2015). In 2019, the CDC confirmed 38 cases of EEE resulting in 15 deaths were diagnosed in 10 states, more than double the highest number of annual EEE cases in the past decade (Centers for Disease Control and Prevention et al., 2019). Unlike the Old World alphaviruses, mortality is not uncommon following infection with the New World alphaviruses. While most people infected with WEEV remain asymptomatic, EEE carries a substantial death rate, ranging from 30% to 70% of symptomatic individuals, depending on the report (Calisher, 1994; Griffin, 2013; Steele et al., 2007). People who survive the initial clinical illness, especially those infected as infants or children, tend to develop chronic persistent neurological deficits (Bruyn and Lennette, 1953; Earnest et al., 1971; León, 1975; Palmer and Finley, 1956; Villari et al., 1995).
Currently, no treatments beyond symptomatic care are available for patients infected with the alphaviruses (Go et al., 2014). Licensed combination vaccines for EEEV, WEEV, and VEEV are available for horses, but no effective vaccine is approved for non-military use in humans (Bartelloni et al., 1970; Steele et al., 2007). While the explosive increase in CHIKV outbreaks and expansion to the Americas over the past 15 years have created a renewed interest in developing effective vaccines and therapies against the arthritogenic alphaviruses, no candidates have yet been successfully developed (reviewed in Powers, 2018). The reemerging public health concern and propensity to cause chronic debilitating effects presented by the alphaviruses necessitate better understanding of the mechanisms mediating the pathogenesis of infection so that safe and effective vaccines and therapies may be developed.
2. Potential outcomes following alphavirus infection
Cellular infection by an alphavirus can ultimately result in one of three outcomes. First, the virus directly induces death of the cell, typically through a mechanism such as apoptosis. For example, infection of neonatal mouse pups with SINV results in extensive apoptosis of multiple cells in the central nervous system (CNS), including neurons (Labrada et al., 2002). Second, the virus induces death through a secondary, rather than direct, mechanism, such as through effects of the immune system or excitotoxicity. And third, which is the optimal outcome, viral infection is cleared, and the cell survives. However, viral persistence following recovery is becoming increasingly recognized with alphaviruses (Fragkoudis et al., 2018; Hoarau et al., 2010; Levine and Griffin, 1992; Morrison et al., 2011), and reactivation of viral replication and relapse of disease must be controlled by the immune system, re-opening the possibility of pathology being induced as a bystander effect. For example, Semliki Forest virus (SFV) RNA persists in the brains of immunocompetent mice following recovery from acute infection, and suppression of the antibody response several months later results in reactivation of infectious virus production (Fragkoudis et al., 2018). The outcome of infection is dependent on a myriad of factors, including viral species and strain, type of cell infected, and host age, genetics, and immune response.
3. Protective effects of the immune system during Alphavirus infection
While the focus of this review is on virus-induced immune pathology, it is also important to remember that the host immune response plays an essential role in both controlling and clearing alphaviruses, while also mediating long-term protective immunity following either primary infection or vaccination. The type I interferon (IFN) system is essential for protecting against alphavirus replication, and mice that lack a functional IFN system or even specific antiviral interferon stimulated genes exhibit enhanced susceptibility to virus replication and disease (Burdeinick-Kerr et al., 2007; Byrnes et al., 2000; Couderc et al., 2008; Gardner et al., 2012; Partidos et al., 2011; Ryman et al., 2000; Schilte et al., 2010; Wollish et al., 2013). Other components of the innate immune response, such as the complement cascade and Toll-like receptor (TLR) signaling also protect from alphaviruses such as SINV, VEEV, and CHIKV (Brooke et al., 2012; Her et al., 2015; Hirsch et al., 1980; Neighbours et al., 2012; Rudd et al., 2012; Schilte et al., 2010; Wollish et al., 2013).
Adaptive immunity is essential for clearance of alphaviruses from infected tissues and mediating long term protective immunity. While glutamine antagonist-mediated inhibition of lymphocyte proliferation and CNS infiltration results in mitigated clinical disease and pathology in SINV infected mice, viral clearance is also delayed (Baxter et al., 2017; Potter et al., 2015). Virus-specific antibody is responsible for viral clearance from the serum and is a major mediator of protective immunity following either primary alphavirus infection or vaccination (Lum et al., 2013; Mallilankaraman et al., 2011). Virus specific antibody directed against the E2 glycoprotein also mediates non-cytolytic clearance of alphaviruses from the CNS (Levine et al., 1991). T cells also make important contributions to alphavirus protective immunity, with local IFN-γ production by CD4+ and CD8+ T cells playing a cooperative role with anti-SINV antibody in facilitating clearance of SINV from the CNS (Baxter and Griffin, 2020; Binder and Griffin, 2001; Burdeinick-Kerr et al., 2007). CD4+ T cells promote recovery from VEEV-induced CNS disease (Brooke et al., 2010), and CD8+ T cells mediate clearance of RRV from infected muscle tissues (Burrack et al., 2015). CD8+ T cell responses can also mediate vaccine-induced protection against CHIKV (Broeckel et al., 2019), and γδ T cells protect from CHIKV-induced joint disease (Long et al., 2015). Mice lacking functional adaptive immune systems are also prone to developing chronic alphavirus infection (Burrack et al., 2015; Hawman et al., 2013; Lum et al., 2013; Nilaratanakul et al., 2018; Seymour et al., 2015; Teo et al., 2013). Therefore, while understanding how alphavirus-interactions with the immune system promote virus-induced disease, it is also important to keep in mind the beneficial aspect of the immune response when considering immune suppressive strategies for treating alphavirus-induced disease. Table 2 provides a summary of the pathologic and protective effects of different components of the immune response seen during infection with different alphaviruses.
Table 2.
Pathogenic and protective roles of the immune response during alphaviral infection.
4. Natural infection with alphaviruses
4.1. Clinical disease, pathology, and immune response in human patients
4.1.1. Old World alphaviruses
Clinical disease induced by the Old World alphaviruses is variable but most commonly characterized by fever, rash, and polyarthritis (inflammation of multiple joints), and polyarthralgia (pain in multiple joints) (Table 1, reviewed in Suhrbier et al., 2012). The frequency of clinical disease development varies by virus, with most RRV and SINV infections being asymptomatic but as high as 80% of CHIKV and MAYV infections resulting in clinical symptoms (Azevedo et al., 2009; Brummer-Korvenkontio et al., 2002; Endy, 2020; Harley et al., 2001; Suhrbier et al., 2012; Vijayakumar et al., 2011). Following an incubation period ranging from 2 to 10 days, patients generally first present with a fever (Suhrbier et al., 2012). Soon after, polyarthralgia most commonly affecting small joints, such as hands/fingers, wrists, ankles, and knees, develops. Synovial fluid sampled early in the RRV disease process contains high numbers of monocytes and activated macrophages with RRV antigen variably detected by immunofluorescence (Clarris et al., 1975; Fraser et al., 1981). Other clinical symptoms commonly seen during acute infection include headache, myalgia (muscle pain), and a maculopapular rash extending over the trunk, limbs, and face (Suhrbier et al., 2012). Histologically, the CHIKV-induced rash is characterized by edema of the epidermis and dermis and perivascular inflammation consisting of lymphocytes and melanophages in the dermis (Riyaz et al., 2010).
Multiple case reports and series have examined the immune response throughout the course of Old World alphavirus infection, particularly with CHIKV. Numerous pro-inflammatory cytokines and chemokines are reported to be elevated in patients’ serum during the acute phase of infection and include tumor necrosis factor alpha (TNF-α), IFN-α, IFN-γ, interleukin (IL) 1 beta (IL-lβ), IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, IL-17, IL-18, IL-17A, IL-18, IL-27, IL-29, granulocyte-colony stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), CC chemokine ligand 2 (CCL2), CCL3, CCL4, CCL5, CXC chemokine ligand 9 (CXCL9), and CXCL10 (Chaaitanya et al., 2011; Chen et al., 2017; Chirathaworn et al., 2013; Chopra et al., 2014; Chow et al., 2011; Gualberto Cavalcanti et al., 2019; Kelvin et al., 2011; Lohachanakul et al., 2012; Ng et al., 2009; Reddy et al., 2014; Santiago et al., 2015; Tappe et al., 2016; Teng et al., 2015; Venugopalan et al., 2014; Wauquier et al., 2011). Vascular endothelial growth factor and platelet endothelial growth factor, potent angiogenic factors, have also been found elevated in serum starting early in MAYV infection (Santiago et al., 2015; Tappe et al., 2016). During the acute phase of CHIKV infection, peripheral blood mononuclear cells (pBMCs) show elevated NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) and caspase-1 mRNA expression (Chen et al., 2017), and circulating CD8+ T cells express high levels of cytotoxic markers such as granzyme B, perforin, and the degranulation marker CD107A (Dias et al., 2018). Increased levels of pro-inflammatory cytokines, lymphopenia, monocytopenia, and neutrophilia are all associated with more severe CHIKV viremia (Chow et al., 2011; Reddy et al., 2014; Teng et al., 2015). Patients with more severe RRV-induced disease have higher levels of serum mannose binding lectin (MBL), a pattern recognition receptor involved in activation of the complement system (Gunn et al., 2012). During the recovery or convalescent phase of alphavirus infection, multiple cytokines and chemokines in the serum have been reported to stay elevated or even increase further compared to the acute phase, including TNF-α, IFN-γ, IL-1β, IL, 2, IL-5 IL-6, IL-7, IL-8, IL-9, IL-12, IL-10, IL-13, IL-17A, IL-18, GM-CSF, CCL2, CCL4, and CXCL10 (Chirathaworn et al., 2010, 2013; Chopra et al., 2014; Kelvin et al., 2011; Santiago et al., 2015). This indicates that even in individuals who are considered recovered from alphavirus infection, the immune response remains activated.
Death following Old World alphavirus infection is rare but can occur, especially in neonatal or elderly patients with comorbidities (Economopoulou et al., 2009; Tandale et al., 2009). Fatal cases have been associated with respiratory, renal, hepatic and neurological disease, and hypertension and underlying respiratory and cardiovascular disease increase the risk of severe and fatal disease (Chua et al., 2010; Economopoulou et al., 2009; Hoz et al., 2015; Mercado et al., 2018). Postmortem histopathological reports are rarely published due to the low mortality rate associated with arthritogenic alphavirus infection and lack of medical infrastructure in the geographical locations where transmission and disease tend to occur. However, postmortem CHIKV-induced pathology described in case reports includes acute tubulointerstitial nephritis with lymphocytic intertubular inflammation and tubular necrosis, viral pneumonia with pulmonary edema and congestion, hepatocellular necrosis with mixed inflammation, and acute pericarditis with mild mononuclear inflammation (Mercado et al., 2018; Mercado et al., 2016; Reilly et al., 2020). Neurological disease associated with CHIKV infection has the highest mortality rate of potential atypical disease manifestations (Economopoulou et al., 2009; Tandale et al., 2009). Patients who develop neurological complications following CHIKV infection have shown elevated TNF-α, IFN- α, IL-6, IL-8, CCL2, CCL5, CCL17, and CXCL9 in cerebrospinal fluid (CSF) samples compared with patients with non-CHIKV-induced neurological disease (Kashyap et al., 2014). Autopsy of a 73-year-old man who died of CHIKV-associated encephalomyeloradiculitis (inflammation of the brain, spinal cord, and spinal nerve roots) revealed a grossly swollen brain with severe edema and multifocal ischemic changes (Ganesan et al., 2008). Microscopically, demyelination in the subcortical white matter with edema and myelin pallor was noted, and mild perivascular lymphocytic inflammation with phagocytic microglia (gitter cells) was found in the basal ganglia. Pooled risk of neonatal death following maternal-fetal transmission of CHIKV has been reported to be 2.8% (Contopoulos-Ioannidis et al., 2018). Fetal death due to CHIKV infection during early gestation results in no gross malformations, and while babies infected during late gestation are born without any clinical symptoms, within a week of birth, they develop joint edema, petechiae (small red or purple spots due to bleeding in the skin), rash, thrombocytopenia, disseminated intravascular coagulopathy (small blood clots throughout the bloodstream that deplete platelets and clotting factors and block small blood vessels), and encephalopathy with a high rate of persistent disabilities (Gérardin et al., 2008; Touret et al., 2006).
Acute symptoms usually resolve within a week or two, but the accompanying arthralgia and arthritis can last months to years following initial infection, creating significant physical, emotional, mental, and financial disability in affected patients. Multiple reports have described the incidence of persistent arthralgia and arthritis following alphavirus infection, especially with CHIKV, but also RRV, MAYV, and ONNV. Up to 81% of patients develop persistent symptoms, including joint tenderness, swelling, and pain, musculoskeletal stiffness and pain, and fatigue (Bonifay et al., 2018; Borgherini et al., 2008; Chang et al., 2017; Couzigou et al., 2018; Duvignaud et al., 2018; Forechi et al., 2018; Gauri et al., 2016; Gérardin et al., 2011; Huits et al., 2018; Lima et al., 2019; Llagonne-Barets et al., 2016; Murillo-Zamora et al., 2018; Ninla-aesong et al., 2020; Panato et al., 2019; Sánchez et al., 2019; Sissoko et al., 2009; Sosa-Martínez et al., 2018; Theilacker et al., 2013; Tritsch et al., 2019; Watson et al., 2020). Similar to acute clinical disease, small joints such as fingers, hands, wrists, ankles, and knees are more likely to be chronically affected (de Souza et al., 2019; Ninla-aesong et al., 2020; Sánchez et al., 2019). Radiographic and ultrasonographic findings include joint effusions, bone erosions, bone marrow edema, and inflammation of the subcutaneous connective tissues (cellulitis), synovial membranes of a joint (synovitis), and tendons (tendonitis) (Amaral et al., 2020a; Mogami et al., 2017). On synovial biopsy of patients with chronic polyarthritis from CHIKV infection, synovitis with inflammation consisting of macrophages, natural killer (NK) cells, CD4+ T cells, and plasma cells, synovial lining hyperplasia, and vascular proliferation is found (Ganu and Ganu, 2011; Hoarau et al., 2010). Synovial cytology in chronic CHIKV-induced arthritis consists mostly of CD14+ monocytes/macrophages, less than 5% of which stain positive for CHIKV, with smaller numbers of activated NK cells and CD4+ T cells (Hoarau et al., 2010). Mononuclear cells, mostly consisting of CD4+ T cells, have also been detected in synovial effusions from chronic RRV disease cases (Fraser and Becker, 1984). Risk factors that are associated with development of chronic arthralgia or arthritis include being female, older age, smoking, more severe acute disease with involvement of multiple joints, persistent viremia, preexisting co-morbidities such as diabetes mellitus, cardiovascular disease, or joint disease, and dengue virus (DENV) co-infection (Amaral et al., 2020a,b; Badawi et al., 2018; Delgado Enciso et al., 2018; Elsinga et al., 2017, 2018; Heath et al., 2018; Huits et al., 2018; Murillo-Zamora et al., 2018; Ninla-aesong et al., 2020; Patel et al., 2019; Sosa-Martínez et al., 2018; Tritsch et al., 2019). Chronic CHIKV arthritis has also been determined to be a risk factor for acute coronary syndrome (OR = 3.0), possibly because the chronic arthritis may trigger plaque destabilization in patients with concomitant cardiovascular disease (Patel et al., 2019).
Overall, persistent arthralgia and arthritis following alphavirus infection is associated with a pro-inflammatory state. Patients who develop chronic arthralgia or arthritis following alphavirus infection tend to have elevated serum levels of pro-inflammatory cytokines and chemokines, including IFN-α, IFN-γ, TNF-α, IL-1β, IL-6, IL-7, IL-8, IL-10, IL-12, IL-15, IL-17, IL-27, GM-CSF, CCL2, CCL3, CCL4, CXCL9, and CXCL10 (Amaral et al., 2020a; Chaaitanya et al., 2011; Chow et al., 2011; Gualberto Cavalcanti et al., 2019; Hoarau et al., 2010; Kelvin et al., 2011; Ninla-aesong et al., 2019; Reddy et al., 2014; Santiago et al., 2015; Sepúlveda-Delgado et al., 2017). Chronic chikungunya patients with tender joints and arthritis show significantly elevated IL-27 and IL-17A serum levels, respectively (Gualberto Cavalcanti et al., 2019). Compared to patients who recover from CHIKV infection, individuals with persistent arthralgia and arthritis tend to show anemia, elevated platelets (thrombocytosis), and elevated C-reactive protein (Amaral et al., 2020a). pBMCs from chikungunya patients with chronic arthralgia possess more IFN-α mRNA and more TNF-α- and IFN-γ-secreting NKT cells with lower cytotoxic activity compared to recovered patients (Hoarau et al., 2010; Thanapati et al., 2017). Chronic CHIKV patients have a delayed IgG3 response and lower antibody avidity index despite mounting an adequate neutralizing antibody response (Anfasa et al., 2019; Kam et al., 2012). Intensity of T cells responses in chronic chikungunya patients versus recovered chikungunya patients has not been found to differ (Hoarau et al., 2013). Osteoblasts from RRV patients with osteoarthritis show enhanced RRV infectivity and replication associated with increased expression of cytokines that favor bone resorption, including TNF-α, IL-1β, IL-6, and CCL2, and delayed type I IFN signaling (Chen et al., 2014).
4.1.2. New World alphaviruses
Neurological manifestations following VEEV, EEEV, or WEEV infection mainly consist of encephalitis or encephalomyelitis (Table 1). The likelihood of encephalitis development following infection with the New World alphaviruses depends on the viral species but is overall the minority of infections, especially with VEEV and WEEV (Greenlee, 2014). Neurological disease, when present, is similar for VEE, EEE, and WEE, though EEEV infections tend to be more severe. Following a prodromal phase lasting approximately 2–7 days consisting of fever and headache sometimes accompanied by myalgia, signs of irritability, and abdominal pain, onset of encephalitis is typically abrupt and progression rapid (Deresiewicz et al., 1997; Greenlee, 2014; Steele et al., 2007). Neurological manifestations include headache, stupor or coma, seizures, focal weakness, and cranial-nerve palsies. Blood chemistries reveal leukocytosis with a left shift (increased immature cells) and low blood sodium (hyponatremia), and CSF analysis shows elevated protein and increased cells (pleocytosis) initially consisting primarily of neutrophils but evolving primarily to lymphocytes (Deresiewicz et al., 1997; Przelomski et al., 1988; Rozdilsky et al., 1968; Silverman et al., 2013). Changes on brain MRI include symmetric or asymmetric lesions in multiple brain regions, including the basal ganglia, hippocampi, thalami, insular cortex, midbrain, and brainstem (Deresiewicz et al., 1997; Nickerson et al., 2016). Edema and periventricular white matter changes similar to those seen in multiple sclerosis patients have also been seen (Deresiewicz et al., 1997). Prognostic indicators for a poorer outcome include young age, particularly infants and children, shorter prodromal phase (<5 days), severe CSF pleocytosis, and coma (Deresiewicz et al., 1997; Finley et al., 1955; Przelomski et al., 1988; Rozdilsky et al., 1968).
Death, mostly associated with EEE, has mainly been reported to occur during second to third week of hospitalization (Deresiewicz et al., 1997). On postmortem examination, brains tend to be grossly congested and edematous, with diffuse meningoencephalitis found microscopically (Greenlee, 2014). Histopathological changes in the central nervous system (CNS) include perivascular and parenchymal inflammation consisting of mononuclear cells and neutrophils accompanying neuronal loss, especially in the basal ganglia and thalamus (Booss and Esiri, 2003; Lindenberg and Haymaker, 1982; Przelomski et al., 1988; Rozdilsky et al., 1968). Demyelinating lesions and oligodendrocyte death have also been reported, and inflammation of the spinal cord (myelitis) and blood vessels (vasculitis) are frequently present with EEE. Intranuclear or intracytoplasmic viral inclusion bodies are conspicuous by their absence (Rozdilsky et al., 1968; Steele et al., 2007).
Patients who survive the initial course of infection with VEEV, EEEV, and WEEV, especially those infected as infants or children, are often left with persistent neurological sequalae (Silverman et al., 2013). Reports of neurological sequelae are best described in patients who have recovered from WEE and include decreased motor skills and altered gait, intellectual and/or learning disability, facial paralysis, impaired speech, and Parkinson-like symptoms (Bruyn and Lennette, 1953; Earnest et al., 1971; Herzon et al., 1957; Mulder et al., 1951; Palmer and Finley, 1956). Neuropsychological symptoms, including anxiety, depression, paranoia, attention deficit disorder, and schizophrenia-like personality changes have also been associated with WEE (Deaton et al., 1986; Fulton and Burton, 1953; Herzon et al., 1957; Palmer and Finley, 1956). VEE has been associated with recurrent headaches, fatigue, depression, seizures, paralysis, and intellectual disability (Bowen et al., 1976; León, 1975; Rivas et al., 1997). The course of neurological sequelae has been reported to range from a static nonprogressive condition to relapsing-remitting disease to complete recovery and reversion to normal function (Bowen et al., 1976; Palmer and Finley, 1956; Silverman et al., 2013). Regardless, neurological sequalae induced by New World alphavirus infection represent a significant source of physical, emotional, mental, and financial disability for affected patients and their caretakers (Villari et al., 1995).
4.2. Immune-mediated disease induced by alphaviruses
As discussed above, alphaviruses can be directly cytolytic, and direct virus-induced cell killing is likely to contribute to alphavirus-induced disease processes. However, there is also abundant evidence that alphaviruses can interact with the host immune system to cause virus-induced disease. These interactions include evidence that alphavirus-induced inflammation contributes to the pathogenesis of arthralgia and/or myalgia that are characteristic of Old World alphaviruses such as CHIKV and RRV, as well as the role of immune cells in driving pathologic aspects of alphavirus-induced CNS disease. However, alphavirus infections are also associated with autoimmune or inflammatory syndromes, and we will discuss each of these alphavirus-induced disease outcomes in the following section (Table 3).
Table 3.
Immune-mediated conditions induced by alphavirus infection.
Organ system | Condition | Description | alphaviruses |
---|---|---|---|
Musculoskeletal | Chronic polyarthritis | Persistent pain, stiffness, and swelling in and around multiple joints | CHIKV, RRV, MAYV, SINV |
Integumentary | Psoriasis | Chronic disease characterized by raised, red, scaly patches on the skin | CHIKV |
Lichen planus | Inflammatory skin condition characterized by bumps, sores, and rough scaly patches on the skin and mouth that often itch | ||
Type I leprosy reaction | Delayed hypersensitivity to Mycobacterium leprae antigens characterized by increased redness and hardness of existing lesions | ||
Nervous | Guillain Barre Syndrome | Autoimmune disorder where the immune system attacks the myelin sheaths or axons of peripheral nerves | CHIKV |
Acute disseminated encephalomyelitis | Autoimmune disorder characterized by sudden widespread inflammation that damages brain and spinal cord white matter | ||
Transverse myelitis | Autoimmune disorder characterized by sudden transient inflammation of the spinal cord | ||
Ocular | Anterior uveitis | Inflammation of the middle layer of the eye, which includes the iris and ciliary body | CHIKV |
Choroiditis | Inflammation of the choroid, the pigmented vascular layer between the retina and sclera | ||
Panuveitis | Inflammation of all layers of the uvea, which includes the iris, ciliary body, and choroid | ||
Retinitis | Inflammation of the retina, the thin layer of tissue at the back of the eye that converts light to neural signal | ||
Neuroretinitis | Inflammation of the retina defined by optic disc edema and stellate macular exudates | ||
Optic neuritis | Inflammation of the optic nerve |
Persistent arthralgia and arthritis have been reported with high incidence following infection with many of the Old World alphaviruses. Multiple characteristics and findings of alphavirus-induced chronic arthralgia and arthritis suggest the pathogenesis is mediated by the immune system rather than directly by the virus. While some studies have found evidence of CHIKV persistence during chronic infection (Hoarau et al., 2010; Ozden et al., 2007), several other studies evaluating synovial fluid in patients with chronic CHIKV- or RRV-induced arthritis found no evidence of CHIKV RNA via PCR, viral proteins via immunohistochemistry or mass spectrometry, or infectious virus via culture (Chang et al., 2018; Fraser and Becker, 1984; Ganu and Ganu, 2011). Regardless of whether chronic viral replication is driving disease pathogenesis, these results suggest that chronic alphavirus-induced arthralgia may be driven by indirect mechanisms. Patients infected with CHIKV have shown flare-ups of preexisting immune-mediated dermatoses, such as psoriasis, lichen planus, and type I leprosy reactions, indicating viral infection induces strong activation of the immune system (Riyaz et al., 2010). Most patients who develop persistent arthralgia following alphavirus infection are females, who are also far more prone to developing autoimmunities than males (Amaral et al., 2020a,b; de Souza et al., 2019; Heath et al., 2018; Huits et al., 2018; Sosa-Martínez et al., 2018; Tritsch et al., 2019). While treatment of chronic alphavirus-induced arthralgia and arthritis is primarily limited to supportive care anchored by nonsteroidal anti-inflammatory drugs, positive outcomes have been reported following treatment with disease modifying anti-rheumatic drugs. Both sulfasalazine and methotrexate, immunosuppressive drugs used to treat autoimmune conditions such as psoriasis, Crohn’s disease, and rheumatoid arthritis, have been used to inhibit proinflammatory cytokine production and reduce joint swelling, tenderness, and pain in patients with CHIKV- or MAYV-induced persistent arthralgia and arthritis with some success (Amaral et al., 2020a,b; Ganu and Ganu, 2011; Malvy et al., 2009; Poon et al., 2019; Theilacker et al., 2013; Zaid et al., 2018). Together, these findings strongly suggest that persistent arthralgia and arthritis induced by alphaviruses are immune-mediated.
Parallels have been drawn between chronic chikungunya arthritis and rheumatoid arthritis. The two conditions share numerous similarities: (1) both conditions are characterized by symmetrical polyarthritis of the small joints, (2) both conditions are most commonly diagnosed in middle-aged females, (3) symptoms for both conditions include arthralgia, arthritis, myalgia, stiffness, and fatigue, (4) both conditions have similar bloodwork and radiographic findings, and (5) many of the same pro-inflammatory cytokines are elevated in the serum and synovial fluid (Amaral et al., 2020a). Multiple case studies have found some chronic chikungunya arthritis patients meet the diagnostic criteria for rheumatoid arthritis according to the American College of Rheumatology/European League Against Rheumatism 2010 Rheumatoid Arthritis Classification Criteria; this classification system evaluates the joints involved, serologic test results for rheumatoid factor and/or anti-cycle citrullinated peptide antibody, elevated acute phase response as evidenced by abnormal C-reactive protein or erythrocyte sedimentation rate, and symptom duration (Aletaha et al., 2010; Amaral et al., 2020b; Amaral and Schoen, 2018; Bouquillard and Combe, 2009; Huits et al., 2018; Miner et al., 2015; Sepúlveda-Delgado et al., 2017). Using the American College of Rheumatology/European League Against Rheumatism 2010 Rheumatoid Arthritis Classification Criteria, a study found that in North Kerala, India, where chikungunya epidemics are frequent, CHIKV infection is significantly related to rheumatoid arthritis development (OR = 8.73) (Paul and Pariyapurath, 2018). Matrix metalloproteinases 1 and 3, which have been shown to correlate with rheumatoid arthritis disease activity and progression, are elevated in chronic chikungunya arthritis patients compared to healthy controls (Green, 2003; Ninla-aesong et al., 2019). NK cell and T cell phenotypes are similar between rheumatoid arthritis and chronic chikungunya arthritis, with pBMCs from both rheumatoid arthritis and chronic chikungunya arthritis patients having higher percentages of activated and effector CD4+ and CD8+ T cells than control patients (Miner et al., 2015). The frequency of human leukocyte antigen (HLA) alleles HLA-DRB1*01 and HLA-DRB1*04, which are associated with increased risk for developing rheumatoid arthritis (MacGregor et al., 1995; Weyand et al., 1992), is significantly higher in patients with chronic joint manifestations following CHIKV and SINV infection than the general population (Bouquillard and Combe, 2009; Sane et al., 2012). The structural polyproteins of multiple Old World alphaviruses share homology with numerous putative human proteins involved in rheumatoid arthritis and demonstrate similar binding patterns to HLA-DRB1 (Venigalla et al., 2020). These potential mimics are predicted to elicit an efficient B cell response, providing a possible autoimmune mechanism by which alphavirus infections are inducing chronic arthralgia and arthritis.
Instances of the immune response inducing pathology following alphavirus infection in human patients has been demonstrated with atypical neurological manifestations associated with the arthritogenic alphaviruses. In particular, multiple case reports of Guillain-Barre syndrome (GBS) have been reported in the literature. GBS is an autoimmune demyclinating polyneuropathy or axonal neuropathy characterized by acute onset of limb or muscle weakness usually progressing to flaccid paralysis (reviewed in Hughes and Cornblath, 2005). The general pathophysiology of GBS consists of an antibody-mediated attack of the myelin surrounding peripheral nerve axons (acute inflammatory demyelinating polyneuropathy subtype) or the axon itself (acute motor axonal neuropathy and acute motor and sensory axonal neuropathy subtypes) due to molecular mimicry between microbial antigens and axolemmal surface molecules (Bourque et al., 2015; Willison et al., 2016). Most cases are preceded by an infection less than 1 month prior, and the type of infection influences the clinical phenotype and prognosis. While GBS is most commonly associated with Campylobacter jejuni infection (Jacobs et al., 1998), infection with arboviruses, including alphaviruses, are becoming increasingly recognized as contributors to GBS development (Davis et al., 2006; Lebrun et al., 2009; Oehler et al., 2015; Sahu et al., 2014).
The first published association was a 1998 report out of Australia suggesting a connection between Barmah Forest virus infection and GBS (Phan et al., 1998). Since that report though, almost all of the associations between GBS and alphavirus infection have involved the naturally arthritogenic alphavirus CHIKV. The association between GBS and CHIKV was first proposed following an outbreak on Reunion Island in the Indian Ocean in 2006. Four reports collectively described eight patients admitted to hospitals with GBS during the 2006 CHIKV outbreak who tested negative for the typical infectious agents associated with GBS (C. jejuni, cytomegalovirus, Mycoplasma pneumoniae, Epstein-Barr virus, dengue virus) and had serum IgM against CHIKV (Lebrun et al., 2009; Lémant et al., 2008; Tourncbize et al., 2009; Wielanek et al., 2007). More cases of GBS were reported concurrent with a CHIKV outbreak in French Polynesia from October 2014 to March 2015, where an estimated 25% of the population became infected (Aubry and Cao-Lormeau, 2019; Koeltz et al., 2018; Oehler et al., 2015). Antibodies against GM2 and GDI a, gangliosides present in nodes of Ranvier along nerve axons, were each found in two patients from the outbreak, further supporting the hypothesis that CHIKV infection can trigger GBS (Oehler et al., 2015). The French Polynesia outbreak was determined to have originated from an infected individual traveling from the Caribbean (Nhan et al., 2014), where CHIKV had recently spread from Southeast Asia in 2013. Retrospective analysis of severe disease manifestations of CHIKV infections during the initial outbreak in Martinique and Guadeloupe in the French West Indies revealed six patients with GBS (Crosby et al., 2016). A case series evaluation of CHIKV-related GBS in 13 patients in the Caribbean found that 54% of cases were classified as the acute inflammatory demyelinating polyneuropathy subtype and 15% of cases were classified as the acute motor and sensory axonal neuropathy subtype, with the remaining cases classified as more uncommon variants (Balavoine et al., 2017). A case-controlled study evaluating the association between CHIKV infection and GBS in the French West Indies found an odds ratio of 8.3, indicating CHIKV infection was a risk factor for GBS (Stegmann-Planchard et al., 2019).
The association of GBS with arboviral infection became especially apparent during the Zika virus (ZIKV) outbreak in Central and South America from 2015 to 2016. Though humans cases were sporadically described in Africa, Asia, and the Pacific Islands starting in the 1950’s, ZIKV, a flavivirus, was first detected in Brazil in early 2015; by March 2016, over 50,000 suspected cases had been reported, and the approximately 5000 cases of ZIKV-associated congenital microcephaly incited public outcry (Kindhauser et al., 2016; Pinheiro et al., 2016). ZIKV, the closely related flavivirus DENV, and CHIKV, for which local infections have been reported since 2014 (Azevedo et al., 2015), are all transmitted by the same endemic Aedes spp. mosquito vectors (Weaver and Lecuit, 2015). Infection with all three viruses induces fever and rash, making it very difficult to provide an accurate diagnosis via clinical symptoms alone, and many laboratory tests are cross-reactive among the viruses, particularly the flaviviruses ZIKV and DENV (Braga et al., 2017). Reports of GBS in the Americas during the 2015–2016 Zika virus outbreak increased markedly (Capasso et al., 2019), with the inciting agent mosdy thought to be ZIKV, though many patients showed evidence of recent CHIKV or DENV infection as well (Acevedo et al., 2017; Anaya et al., 2017; de Azevedo et al., 2018; Dirlikov et al., 2016; Lima et al., 2019; Malta et al., 2017; Mancera-Páez et al., 2018; Mehta et al., 2018; Styczynski et al., 2017; Vieira et al., 2018; Zambrano et al., 2016). During the ZIKV outbreak, other cases of GBS were reported following CHIKV infection, with no evidence of prior ZIKV infection (Del Carpio-Orantes et al., 2018; Villamil-Gómez et al., 2016). Wachira et al. performed a systematic review evaluating factors reported to trigger GBS prior to and after 2007 and found that both CHIKV and ZIKV were identified in the literature as new etiological agents associated with the autoimmune condition (Wachira et al., 2019). Circulation of multiple arboviruses, particularly CHIKV, ZIKV, and DENV, in affected areas and high frequency of co-infections among patients complicate doctors’ and scientists’ ability to determine which pathogen(s) is responsible for triggering GBS. Further studies evaluating the exact mechanism by which arboviruses trigger GBS and how co-infections contribute to development of the condition are warranted.
Other immune-mediated neurological conditions have been reported as atypical manifestations of CHIKV infection in human patients. Acute disseminated encephalomyelitis (ADEM) is multifocal neurologic and demyclinating syndrome usually triggered by a prior infection whose pathophysiology is thought to be due to T cell activation following formation of antibodies against myelin and other self-antigens in the CNS (reviewed in Gray and Gorelick, 2016). Several cases of ADEM have been reported following CHIKV infection (Anand et al., 2019; Carvalho et al., 2020; Maity et al., 2014; Musthafa et al., 2008; Taraphdar et al., 2015; Teixeira et al., 2019), with a cross-sectional study of CHIKV cases during an outbreak in Pakistan showing an ADEM incidence rate of 5% (Barr et al., 2018). Transverse myelitis, another immune-mediated neurologic condition with variable pathological lesions involving the spinal cord, including demyelination, axonal degeneration, and perivascular infiltration of monocytes and lymphocytes (reviewed in Krishnan et al., 2006), has also been recently documented following CHIKV infection (Choudhary et al., 2016; Farias et al., 2018; González-Galván et al., 2017; Hameed et al., 2019; Kumar et al., 2019; Neri et al., 2018). Multiple case reports and series have described atypical ocular manifestations following CHIKV infection, including anterior uveitis, choroiditis, panuveitis, retinitis, neuroretinitis, and optic neuritis (Chanana et al., 2007; Deeba et al., 2019; Lalitha et al., 2007; Lin et al., 2018; Mahendradas et al., 2008, 2010; Mahesh et al., 2009; Mittal et al., 2007; Murthy et al., 2008; Nair et al., 2012; Neri et al., 2018; Rocha et al., 2018; Rose et al., 2011; Salceanu and Raman, 2018; Scripsema et al., 2015; Ulloa-Padilla et al., 2018). Whether the pathological changes for these ocular conditions are due to a direct viral mechanism or an immune-mediated mechanism are not currently known. However, in most of the reported cases, ocular disease was bilateral, developed after a period of latency following initiation of classical CHIKV symptoms (fever, arthralgia, etc.), and responded to immunosuppressive therapy such as corticosteroids, all suggesting an immune-mediated, rather than direct viral, mechanism (Merle et al., 2018; Rose et al., 2011).
An in-depth systematic review that effectively describes the range of neurological disease induced by CHIKV infection in human patients from CHIKV outbreaks in Reunion Island, Italy, Thailand, the Caribbean, and Pacific Islands was recently published by Cerny et al. (2017). Of 64 neurological cases described, encephalitis was the most common neurological complication diagnosed (27 cases), followed by optic neuropathy (18 cases), neuroretinitis (5 cases), GBS (5 cases), and ADEM (3 cases). Encephalitis cases were more commonly diagnosed in infants and children or elderly patients with co-morbidities, but in contrast, most of the cases involving the peripheral nervous system (PNS) (optic neuropathy, neuroretinitis, GBS) were diagnosed in previously healthy young to middle-aged adults. While encephalitis cases were mainly treated with supportive care and had a high rate of moderate to severe complications (100%) and severe outcomes (47%), cases involving the brainstem, spinal cord, or PNS were treated with immunosuppressive therapy (e.g., steroids, intravenous immunoglobulin), resulting in a good (66%) or partial (33%) response to therapy, and much lower complication (29%) and severe outcome (18%) rates. These findings caused the authors to suggest that based on the treating physicians’ assumptions, CHIKV-induced encephalitis has a direct viral pathomechanism, while neurological conditions involving the brainstem, spinal cord, or PNS has an underlying autoimmune or immunopathogenic mechanism. This hypothesis is further supported by the finding that the mean latency from development of classical CHIKV infection systems (fever, arthralgia, rash, etc.) to development of neurological complications was 3.3 days for encephalitis cases but 16.2 days for brainstem, spinal cord, or PNS cases; furthermore, for several patients who developed PNS disease, the latency period was marked by a complete lack of symptoms. Evaluation of the location of different diagnoses showed that while most cases from the Reunion Island outbreak were thought to have a direct viral pathomechanism (88%), most cases from outbreaks in India were autoimmune conditions involving the PNS (82%). This suggests that genetic factors, whether viral or host, are likely contributing to the development and pathogenesis of immune-mediated neurological disease during CHIKV infection.
5. Alphavirus-induced arthralgia and myalgia
As noted above, arthritogenic alphaviruses, and especially CHIKV, have been associated with several autoimmune neurologic disease states in humans, including GBS. However, the major disease signs associated with arthralgia-associated alphaviruses, which include CHIKV, RRV, and MAYV, are severe acute arthralgia and myalgia, that can progress to chronic disease in a subset of patients (reviewed in Burt et al., 2017). As discussed above, there is evidence for the host inflammatory response contributing to alphavirus-induced arthralgia and myalgia in humans. However, much of the evidence for immune pathology in driving these disease states comes from animal models. CHIKV causes inflammatory arthritis and myositis both in mice and in non-human primates, and these models have been useful systems both for studying the pathogenesis of CHIKV-induced disease and platforms for evaluating CHIKV therapeutics and vaccines (reviewed in Haese et al., 2016). Although most analysis has focuses on CHIKV, mouse models of RRV and MAYV also indicate that host inflammatory responses play a pathologic role in virus-induced myositis and arthritis (de Castro-Jorge et al., 2019; Lidbury et al., 2000).
Components of both the innate and adaptive immune system contribute to alphavirus-induced arthritis and myositis. Monocytes and/or macrophages are a prominent feature of both CHIKV and RRV-induced arthritis or myositis in mouse models, and depletion of these cells results in decreased disease signs (Gardner et al., 2010; Lidbury et al., 2000). Given the importance of monocyte and macrophages in the pathogenesis of CHIKV and RRV, additional studies have evaluated the potential for inhibitors of monocyte recruitment as therapies for alphavirus-induced arthritis. Bindarit, an inhibitor of monocyte chemo tactic proteins (CCL2, CCL8), protects against both CHIKV and RRV-induced disease (Chen et al., 2015; Rulli et al., 2009), which suggest that targeting monocyte chemotactic proteins may be of therapeutic benefit. However, mice that are genetically deficient for CCR2, which acts as a receptor for several chemokines, including CCL2 led to enhanced CHIKV-induced arthritis characterized by neutrophil infiltration into joint tissues (Poo et al., 2014). This suggests that the inflammatory pathways that regulate monocyte recruitment into CHIKV infected tissues is complex and requires further study.
The complement cascade, which plays a protective role in the context of alphavirus-induced CNS disease (Brooke et al., 2012; Hirsch et al., 1980), has also been shown to play a role in alphavirus-induced arthritis and/or myositis. RRV infected mice show high levels of complement deposition in infected skeletal muscle (Morrison et al., 2007). Mice lacking the C3 component of complement, which is essential for activation and amplification of the pathways, show significantly reduced disease signs and muscle damage, yet viral loads and inflammatory cell recruitment into the infected muscle are unchanged. Further analysis found that complement receptor 3 deficiency led to a similar decrease in virus-induced disease and tissue damage, which suggests that complement receptor 3 expression on macrophages or other inflammatory cells may be contributing to inflammatory cell-mediated tissue damage in RRV-infected tissues (Morrison et al., 2008). Studies by Gunn, et al. went on to show that RRV-induced complement activation occurred via the MBL dependent pathway of complement activation and provided evidence that differences in MBL levels may affect RRV-induced disease severity in humans (Gunn et al., 2012). In a follow-up study, Gunn, et al., demonstrated that N-linked glycans on the RRV E2 protein were required for MBL binding, complement activation, and RRV-induced muscle damage (Gunn et al., 2018). It remains to be determined whether the complement cascade’s role in driving virus-induced tissue damage is specific to RRV or if complement plays a larger role in the pathogenesis of other arthralgia associated alphaviruses.
As noted above, adaptive immunity plays an important role in protecting from arthralgia associated alphaviruses, with both antiviral antibody and CD8+ T cells contributing to protection from virus-induced disease. However, there is also evidence that CD4+ T cells contribute to CHIKV-induced disease, since mice lacking CD4+ T cells show a reduction in CHIKV-induced footpad swelling and joint pathology (Teo et al., 2013). Follow up studies by this group showed that expansion of regulatory T cells (Tregs) ameliorates CHIKV-induced disease in the mouse by inhibiting recruitment of pathologic CD4+ T cells into sites of CHIKV infection (Lee et al., 2015). These findings not only provide important insights into the pathogenesis of CHIKV-induced disease, but also have important implications for both the development of CHIKV vaccines and for potential therapeutic approaches for treating CHIKV-induced arthritis. This latter point is illustrated by the findings of Miner et al., where the disease-modifying anti-rheumatic drug cytotoxic T-lymphocyte-associated protein 4 (CTLA4) immunoglobulin, which blocks T cell co-stimulation, had benefit in treating CHIKV-induced arthritis and joint swelling in a mouse model, and this activity was enhanced when CTLA4 immunoglobulin was combined with a potent CHIKV specific monoclonal antibody (Miner et al., 2017).
6. Alphavirus-induced encephalomyelitis
6.1. Venezuelan equine encephalitis virus (VEEV), eastern equine encephalitis virus (EEEV), and western equine encephalitis virus (WEEV)
The neurotropic alphaviruses primarily consist of VEEV, EEEV, and WEEV, and animal models have helped shed light on the pathogenesis and immune response to infection with these viruses. Horses represent a highly permissive aberrant host for natural infection with the equine encephalitides but have also been used to study experimental infection. During infection with VEEV, severity of disease ranges from asymptomatic to rapid death, with clinically affected equids demonstrating fever, lethargy, weakness, and convulsions (Dietz et al., 1978; Gleiser et al., 1962; Monlux and Luedke, 1973; Roberts et al., 1970; Sahu et al., 2003; Walton et al., 1973). Pathological changes are consistent with meningoencephalitis and include perivascular and parenchymal infiltration of lymphocytes, mononuclear cells, and neutrophils with variable presence of glial cell proliferation and/or hypertrophy (gliosis) and clustering around neurons (satellitosis), neuronal damage, and vasculitis in the CNS. EEEV infection has a much higher mortality rate in horses than VEEV, with pathological lesions in the CNS characterized by perivascular cuffing with mononuclear cells, neutrophilic infiltration in the parenchyma, gliosis, neuronophagia, and vascular damage, consistent with diffuse polioencephalomyelitis (inflammation of the gray matter of the brain and spinal cord) (Del Piero et al., 2001; Franklin et al., 2002). WEEV appears to be the least virulent of the equine encephalitides, with most horses developing non-clinical infections (Potter et al., 1977). However, the large size and expense of housing horses precludes most in depth experimental studies evaluating the pathogenesis of VEEV, EEEV, and WEEV infection, necessitating the use of smaller animal models.
While experimental infections have been performed in other animals such as hamsters, rabbits, and guinea pigs, mice and nonhuman primates are considered to be the most relevant animal models for evaluating the pathology and immune response induced by VEEV, EEEV, and WEEV. Experimental infection of mice with virulent strains of VEEV by subcutaneous or footpad inoculation (considered to mimic natural infection by mosquito bite) results in CNS invasion via axonal transport up olfactory neurons following initial infection of dermal dendritic cells, replication in the draining lymph nodes, and viremic dissemination throughout the body (Charles et al., 1995; Davis et al., 1994; MacDonald and Johnston, 2000; Ryzhikov et al., 1991; Steele et al., 1998, 2006; Vogel et al., 1996). Aerosol or intranasal infection leads to a direct infection of olfactory neuroepithelium, resulting in infection of the brain as early as 16 h post infection (HPI) (Bocan et al., 2019; Cain et al., 2017; Ryzhikov et al., 1991; Steele et al., 1998; Vogel et al., 1996). Once in the CNS, VEEV rapidly disseminates, primarily targeting neurons in both the brain and spinal cord, though macrophages may also be infected (Jackson et al., 1991; Steele et al., 1998; Vogel et al., 1996). Histopathological changes are consistent with encephalomyelitis and characterized by thick perivascular cuffs and parenchymal infiltration, initially of neutrophils but then evolving to mostly mononuclear cells, as well as an increase in astrocytes (astrocytosis) (Ryzhikov et al., 1991; Schoneboom et al., 2000a; Sharma et al., 2008; Sharma and Maheshwari, 2009; Steele et al., 1998, 2007). Neuronal damage has been reported to occur through both apoptosis and necrosis (Bocan et al., 2019; Jackson and Rossiter, 1997; Schoncboom et al., 2000a; Steele et al., 1998; Steele and Twenhafel, 2010).
Infection of NHPs with VEEV via the intraperitoneal route results in transient fever and viremia within the first 3 days post infection (DPI) and infection of the brain by 6 DPI (Gleiser et al., 1962). Histopathological changes are similar but generally less severe than those seen in mouse models and include multifocal perivascular cuffs composed mostly of lymphocytes, gliosis, and rare evidence of neuronal damage associated with microglial clusters (glial nodules). Experimental intranasal infection with VEEV results in lesions in the olfactory bulbs by 48 HPI but is otherwise results in similar, though slightly more severe, pathological lesions in the CNS (Danes et al., 1973). Most experimental VEEV infection studies in NHP’s are conducted in the context of vaccine research using aerosol exposure. Infection of the CNS following challenge by this route is variable but induces lesions similar to those seen in humans when present, such as perivascular cuffing and gliosis (Fine et al., 2008; Steele and Twenhafel, 2010).
Unlike VEEV, EEEV does not initially replicate in dendritic cells or macrophages (Gardner et al., 2008; Levitt et al., 1979) but instead in fibroblasts, skeletal muscle, and osteoblasts (Vogel et al., 2005). Also unlike with the mouse model of VEEV infection, neuroinvasion by EEEV occurs via the hematogenous route rather than via axonal transport into the olfactory bulb (Vogel et al., 2005). EEEV can be detected in the brains of mice infected subcutaneously as early as 24 HPI, and multifocal positive EEEV antigen staining seen in multiple regions of the brain supports hematogenous spread. Similar to VEEV, neurons represent the main infected cell, though glial cells may also be infected (Roy et al., 2009; Vogel et al., 2005). Pathological changes in the brain include diffuse neuronal necrosis but fairly mild infiltration of neutrophils and eosinophils. Though not a known natural route of infection, aerosol challenge with EEEV has been studied mainly in the context of biodefense, as many strains of EEEV are classified as Select Agents. In this challenge model, infection has been demonstrated to first occur in the olfactory bulb and then disperse throughout the brain via transneuronal spread, similar to aerosol models of VEEV infection (Honnold et al., 2015a; Roy et al., 2009; Steele and Twenhafel, 2010). Histopathological findings include fulminant meningoencephalitis with abundant neuronal vacuolation, degeneration, and necrosis but few inflammatory infiltrates (Honnold et al., 2015b; Phelps et al., 2019). An aerosol challenge guinea pig model of EEE has shown a similar route of viral entry into the CNS, and pathological changes in the brain consist of marked neuronal necrosis and both perivascular and parenchymal inflammation (Roy et al., 2009). When inoculated with EEEV subcutaneously, hamsters have been shown to be exquisitely sensitive to infection, with virus detected in lung, liver, muscle, heart, kidney, spleen early in infection and titers increasing in the brain from 3 DPI to death at 5 or 6 DPI (Paessler et al., 2004). Similar to humans infected with EEEV, hamsters demonstrate fulminant vasculitis and subependymal hemorrhages with mixed inflammation in the brain. Neuronal death is mainly attributed to virus-mediated apoptosis (Honnold et al., 2015b; Vogel et al., 2005).
NHPs have also been used to evaluate the pathology induced by EEEV infection. Direct inoculation of virus into the brains of juvenile rhesus macaques results in severe encephalitis characterized by neuronal necrosis and loss with relatively mild inflammation (Nathanson et al., 1969). Aerosol challenge of cynomolgus macaques with virulent EEEV results in severe meningoencephalomyelitis, characterized by diffuse neuronal necrosis, perivascular cuffing with mononuclear cells and neutrophils, gliosis, satellitosis, and vasculitis (Reed et al., 2007; Roy et al., 2013; Steele and Twenhafel, 2010). Perivascular spaces are often expanded by edema or hemorrhage, and viral antigen is detected in multiple brain regions, primarily targeting neurons. Marmosets, small callitrichid monkeys native to Central and South America, have also been evaluated as potential models for EEE, and infection with EEEV induces meningoencephalitis with perivascular hemorrhages, similar to what is seen in human patients (Adams et al., 2008; Porter et al., 2017). When comparing the pathology induced by VEEV versus EEEV in different animal models, despite both viruses primarily targeting neurons, EEEV infection induces markedly more severe neuronal damage and death. However, these pathological changes are associated with relatively little inflammation, whereas VEEV infection of the brain results in massive immune cell infiltration, but only minimal damage to neurons.
Generally considered the least neurovirulent of the New World alphaviruses, animal studies of WEEV infection have shown variable pathology. Mice infected with more virulent WEEV strains demonstrate high mortality via multiple inoculation routes and develop severe meningoencephalitis characterized by neuronal degeneration and necrosis, edema, and mild infiltration of mononuclear cells (Aguilar, 1970; Logue et al., 2009; Phelps et al., 2017). Infection with the less virulent strains demonstrate more perivascular inflammation but minimal neuronal damage (Logue et al., 2009). Following recovery from WEEV, CD-1 mice demonstrate persistent gliosis (Bande et al., 2019). High mortality in WEEV-infected neonatal mice due to severe inflammation and/or necrosis in peripheral tissues such as bone marrow, skeletal muscle, and peripheral nerves without any CNS involvement (Aguilar, 1970) is similar to disease seen in some reports of hamsters infected with VEEV (Gorelkin and Jahrling, 1975; Jackson et al., 1991; Jahrling and Scherer, 1973; Walker et al., 1976). Challenge experiments via subcutaneous, intranasal, and aerosol routes in mice indicate WEEV invades the brain via axonal transport up the olfactory tract similar to VEEV (Bantle et al., 2019; Phelps et al., 2017; Phillips et al., 2013, 2016). Experimental infection of hamsters with WEEV demonstrates that, similar to the other equine encephalitides, neurons are the preferred cell for infection in the CNS (Julander et al., 2007), though infection of microglia has also been demonstrated in an aerosol challenge model of WEEV in cynomolgus macaques (Reed et al., 2005).
The immune response has been shown to play a major role in the pathogenesis of VEEV infection. Following subcutaneous inoculation with VEEV, mice show upregulation of proinflammatory genes Ifng, Il6, Il12, Il17, and Tnf, chemokine genes Cxcl9, Cxcl10, Cxcl11, Cxcl13, Ccl2, Ccl3, Ccl5,and Ccl12, and genes associated with TLR signaling, including Tlr1, Tlr2, Tlr3, Tlr7, and Tlr9, correlating with blood brain barrier compromise and infiltration of neutrophils and mononuclear cells into the brain (Grieder et al., 1997; Gupta et al., 2017; Sharma et al., 2008; Sharma and Maheshwari, 2009). Infection of multiple strains of mice with the V3000 strain of VEEV results in induction of proinflammatory cytokines, including inducible nitric oxide synthase, IL-1β, IL-6, IL-12, and TNF-α (Schoneboom et al., 1999, 2000a, 2000b). Mice administered magnesium or cadmium demonstrate more rapid development and severe histopathological CNS lesions concurrent with upregulation of proinflammatory cytokines following infection with both VEEV and SFV (Seth et al., 2003). Treatment of BALB/c mice with IFN-α prior to VEEV challenge results in improved survival, reduced TNF-α production by macrophages, and delayed activation of the adaptive immune response (Lukaszewski and Brooks, 2000). Inhibition of GSK-3β, a serine/threonine protein kinase with immunomodulatory capabilities, protects C3H mice from VEEV TC83-induced encephalitis (Kehn-Hall et al., 2012).
The adaptive immune response also contributes to pathological changes in the CNS during VEEV infection. Severe combined immunodeficiency (SCID) mice, which lack both T cells and B cells, show improved survival times compared to wild-type mice despite demonstrating higher viral titers (Charles et al., 2001). Nude mice, which lack T cells, infected with VEEV do not develop the inflammatory demyelinating spinal cord lesions seen in wild-type (WT) counterparts, and mice administered anti-thymocyte serum, which depletes T cells, have delayed mortality, indicating T cells play a role in inducing VEEV pathology (Dal Canto and Rabinowitz, 1981; Woodman et al., 1975). However, the role and contributions of CD4+ versus CD8+ T cells or other immune cells, such as macrophages, to immune-mediated damage during VEEV infection has not yet been elucidated.
As inflammation in the brain is not a key aspect of brain pathology during EEEV infection, less is understood about the role the immune system plays in the pathogenesis of EEE. However, mice infected via the intranasal or aerosol route with EEEV do demonstrate upregulation of proinflammatory cytokines IFN-γ, CCL4, CCL5, CXCL9, and G-CSF (Honnold et al., 2015a). While few articles describing the local immune response to WEEV have been published, increases in pro-inflammatory cytokines such as IFN-γ, TNF-α, IL-12, CCL2, and CXCL10 have also been demonstrated at 3 DPI in the brains of WEEV MCM strain-infected CD-1 mice before death at 4–5 DPI (Logue et al., 2010; Phillips et al., 2013). Significant differences in clinical outcomes among various inbred mouse strains following WEEV infection, with C57BL/6 and BALB/c mouse strains showing high susceptibility and BALB/cBy and DBA/2 mouse strains being more resistant to severe disease, suggest that some of these disparities in neurovirulence due to host genetics are likely due to differences in the immune response (Blakely et al., 2015).
6.2. Sindbis virus (SINV)
Numerous extensive studies have been conducted using the naturally arthritogenic alphavirus SINV to study the neuropathogenesis of alphavirus infection. SINV is neurotropic in mice but, unlike most strains of the equine encephalitides, can be handled under biosafety level 2 conditions and therefore represents a more accessible model of alphavirus encephalomyelitis. Disease severity is dependent on the strain of SINV used; for example, while both viruses effectively disseminate throughout the CNS, AR339, the pro-totypic strain of SINV, is fatal in neonatal mice but nonvirulent in weanling mice, while neuroadapted SINV (NSV) induces ascending paralysis and 100% mortality in susceptible mice of all ages (Griffin, 1976; Griffin and Johnson, 1977; Jackson et al., 1987; Taylor et al., 1955). Similar to VEEV and WEEV, neuroinvasion occurs via transaxonal transport from the neuro-epithelium to the olfactory bulb, after which SINV disseminates caudally throughout the brain and to the brainstem and spinal cord (Lee et al., 2013; Passoni et al., 2017; Thach et al., 2000). Ascending paralysis seen with SINV NSV infection is due to infection and degeneration of motor neurons in the lumbar spinal cord (Havert et al., 2000; Jackson et al., 1988). In brains of C57BL/6 mice infected with SINV, marked loss of neurons, particularly in the CA regions of hippocampus, is a key feature (Baxter et al., 2018; Kimura and Griffin, 2003). Perivascular cuffing and parenchymal infiltration of mononuclear cells is also considerable (Baxter et al., 2018; Irani and Griffin, 1991; Kimura and Griffin, 2003; Moench and Griffin, 1984; Stohlman et al., 1998). Hydrocephalus and dilation of the lateral ventricles may also be seen (Baxter and Griffin, 2020; Kimura and Griffin, 2003). Mice infected with the nonfatal TE strain of SINV demonstrate clinical disease characterized by gait deficits and exhibit behavioral changes concurrent to the development of these pathological changes in the brain (Baxter and Griffin, 2016; Baxter et al., 2018; Potter et al., 2015). In particular, hippocampal-dependent memory changes persist beyond the period of recovery from clinical disease and virus clearance (Potter et al., 2015). The mechanism of neuronal damage is dependent on the type of neuron, with evidence of both apoptosis and necrosis demonstrated (Havert et al., 2000; Kerr et al., 2002; Lewis et al., 1996; Nargi-Aizenman and Griffin, 2001).
Studies have shown that the immune system, particularly T cells, play the predominant role in inducing CNS pathology during SINV infection. During nonfatal SINV infection, inflammatory cells begin to infiltrate the CNS around 3 DPI, peaking at 7–10 DPI (Moench and Griffin, 1984). While NK cells peak early in infection, at 7 DPI, CD8+ T cells and macrophages predominate, though numbers of CD4+ T cells rapidly increase, becoming the main immune cell type by 10 DPI (Baxter and Griffin, 2020; Irani and Griffin, 1991, 1996; Metcalf and Griffin, 2011; Moench and Griffin, 1984). Onset of neurological disease and mortality in mice infected with SINV coincides with clearance of infectious virus and infiltration of immune cells into the brain (Griffin and Hardwick, 1997; Jackson et al., 1987; Kulcsar et al., 2014). SCID mice, which are deficient in both mature T and B cells, show 100% survival and minimal neuropathology following SINV NSV infection and do not develop neurological disease following SINV AR339 infection despite supporting high levels of virus replication (Levine et al., 1991; Wesselingh et al., 1994). Impaired proliferation and infiltration of lymphocytes into the brain of SINV-infected mice treated with 6-diazo-5-oxo-1-norleucine, a glutamine antagonist that arrests lymphocytes in the G1 phase of the cell cycle (Wang et al., 2011), leads to reduced mortality, weight loss, and development of neurological disease and cognitive deficits (Baxter et al., 2017; Manivannan et al., 2016; Potter et al., 2015). Mice deficient in multiple components of the cellular adaptive immune response, including TCRα, TCRβδ, β2m, TAPI, and CD4, but not CD8, show reduced mortality following SINV NSV infection, indicating T cells, especially CD4+ T cells, play a major role in inducing CNS pathology (Kimura and Griffin, 2000; Rowell and Griffin, 2002). In particular, Th17 cells expressing granzyme B, IL-22, and GM-CSF infiltrate the CNS during the development of SINV NSV-induced paralysis and fatal neurological disease (Kulcsar et al., 2014). These pathogenic Th17 cells are regulated by IL-10, with Il10−/− mice developing more severe weight loss and neurological disease and increased mortality concurrent with more CD4+ T cells skewed towards a Th1 and Th17 response but away from a Th2 and Treg response in the brain (Kulcsar et al., 2014; Kulcsar and Griffin, 2016; Martin and Griffin, 2018). BALB/c mice, which are more resistant to fatal encephalomyelitis than C57BL/6 mice, demonstrate fewer Th17 cells and more IL-10-dependcnt Tregs in the brain following SINV NSV infection (Kulcsar et al., 2015). These studies together demonstrate the importance of the T cell response in inducing neuropathology in the mouse model of alphavirus encephalomyelitis.
Proinflammatory cytokines have been shown to play a role in the immunopathogenesis in the SINV mouse model of alphavirus encephalomyelitis. Following infection with multiple strains of SINV, mouse brains show uprcgulation of proinflammatory cytokine gene expression, including Tnf, Csf2, Il1b, Il4, Il6, Il12b, Il17a, Il23, Lif, and Ifng (Baxter et al., 2018; Baxter and Griffin, 2016; Kulcsar et al., 2015; Lee et al., 2013; Martin and Griffin, 2018; Rowell and Griffin, 1999; Wesselingh et al., 1994). Mice deficient in interferon regulatory factors 3 and 7, who show increased disease severity and mortality respectively, have upregulated brain expression of multiple proinflammatory cytokines that correlate with increased severity of inflammation (Irf3−/− mice), numbers of CD4+ (Irf3−/− mice) and CD8+ (Irf7−/− mice) T cells, and TUNEL-positive cells mice) in the brain and spinal cord compared to WT mice following infection with the generally nonfatal TE strain of SINV (Schultz et al., 2019). IL-1β deficient mice are less susceptible to fatal encephalitis induced by SINV NSV, despite showing comparable histopathology and levels of apoptosis as WT mice (Liang et al., 1999). TNF-α deficient mice infected with SINV NSV show improved survival and reduced neuronal death compared to WT mice (Carmen et al., 2009), and TNF-receptor-1-deficient mice show reduced mortality and delayed development of paralysis compared to WT mice infected with the neurovirulent SVNI strain of SINV (Sarid et al., 2001). IFN-γ, the sole type II IFN, is known to contribute to the clearance of SINV in cooperation with anti-SINV antibody (Baxter and Griffin, 2016; Binder and Griffin, 2001; Burdeinick-Kerr et al., 2007; Lee et al., 2013) but is also involved in inducing pathology. WT mice lose more weight following SINV TE infection compared to Ifng−/− and Ifngr1−/− mice due to changes in feed intake indirectly related to levels of TNF-α in the brain (Baxter and Griffin, 2016). During the height of clinical disease at 7 DPI, Ifng−/− and Ifngr1−/ mice show reduced inflammation in the brain and spinal cord and are less likely to demonstrate dilatation of the lateral ventricles compared to WT mice (Baxter and Griffin, 2020). Mice with intact IFN-γ signaling have more macrophages and degranulating NK cells in the brain but fewer CD8+ T cells producing granzyme B and perforin and show delayed initiation of viral RNA clearance compared to Ifng−/− and Ifngr1−/− mice (Baxter and Griffin, 2020; Lee et al., 2013). Upregulation of proinflammatory cytokines induced during alphavirus encephalomyelitis likely promotes the infiltration of inflammatory cells that then induce pathological changes and neuronal damage in the CNS.
Another indirect mechanism by which neurons may die during alphavirus infection of the CNS is excitotoxicity. In neuronal excitotoxicity, excess amounts of the neurotransmitter glutamate accumulate in the synaptic cleft, leading to overstimulating of a-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA), N-methyl-d-aspartate, and kainate-type receptors (reviewed in Sattler and Tymianski, 2001). Influx of metal ions, particularly Ca2+, into the neuron through channels gated by these receptors leads to metabolic destruction of the cell (Lee et al., 1999; Nicotera and Orrenius, 1998). Glutamate excitotoxicity has been shown to play a role in the pathogenesis of SINV infection, with neuronal infection by multiple strains of SINV resulting in downregulation of glutamate transporter 1, a receptor primarily expressed on astrocytes responsible for preventing glutamate excitotoxicity by taking up the majority of glutamate from the synaptic cleft (Baxter et al., 2017; Carmen et al., 2009; Darman et al., 2004; Nargi-Aizenman and Griffin, 2001; Prow and Irani, 2007). Treatment of mice with either an N-methyl-d-aspartate receptor antagonist or AMPA receptor antagonist results in fewer necrotic and apoptotic neurons in the hippocampus following infection with SINV NSV (Nargi-Aizenman et al., 2004). Furthermore, spinal cord neuronal damage and paralysis is prevented, and mortality is reduced in mice treated with the AMPA receptor antagonist in a viral replication-independent manner (Greene et al., 2008; Nargi-Aizenman et al., 2004). However, inhibition of glutamate excitotoxicity either by AMPA receptor or glutamine antagonist treatment also results in reduced proliferation and impaired infiltration of immune cells in the draining lymph nodes and brain, respectively, during SINV infection (Baxter et al., 2017; Greene et al., 2008; Manivannan et al., 2016; Potter et al., 2015). Furthermore, mice deficient in TNF-α or IL-1β do not demonstrate glutamate transporter 1 downregulation in the brain following SINV NSV infection like WT mice do (Carmen et al., 2009; Prow and Irani, 2007). These findings indicate that multiple pathways involved in excitotoxicity also affect the immune response and emphasize the interconnectedness of different mechanisms known to induce neuronal damage during alphavirus encephalomyelitis.
6.3. Semliki forest virus (SFV)
SFV, an Old World alphavirus antigcnically related to RRV and MAYV, is generally considered to be non-pathogenic in humans, only occasionally causing fever, myalgia, and arthralgia (Mathiot et al., 1990). However, a case of fatal encephalitis due to laboratory acquired SFV infection has been reported (Willems et al., 1979), and infection of mice with multiple strains of SFV by both direct and indirect routes consistently results in demyelinating meningoencephalomyelitis (Gates et al., 1984; Kelly et al., 1982). While some SFV strains produce acute fatal encephalitis, the well-studied avirulent A7(74) strain of SFV induces a transient viral infection that is cleared by 8 DPI in susceptible immunocompetent strains, such as BALB/c mice (Amor et al., 1996; Berger, 1980; Kelly et al., 1982; Morris et al., 1997). Perivascular cuffs composed of CD8+ T cells, B cells, and macrophages are found throughout the brain starting during the first week of infection, but beginning around 10–15 DPI, mice develop demyelinating lesions in the white matter of the cerebellum, corpus callosum, and brainstem (Amor et al., 1996; Berger, 1980; Kelly et al., 1982; Morris et al., 1997; Subak-Sharpe et al., 1993). Following infection and subsequent SFV clearance, focal areas of vacuolation in the myelin are filled with necrotic cells and surrounded by mononuclear cells, particularly lymphocytes extending cytoplasmic projections to astrocytes and macrophages (Kelly et al., 1982; Sheahan et al., 1983).
Similar to EEEV, following peripheral inoculation, SFV primarily enters the CNS via hematogenous spread by infecting vascular endothelial cells (Erälinna et al., 1996; Pathak and Webb, 1974; Soilu-Hänninen et al., 1994), though intranasal infection has shown that neuroinvasion via the olfactory bulb is also possible (Keogh et al., 2003; Oliver and Fazakerley, 1998). Upregulation of intercellular adhesion molecule 1 and fibrinogen in the CNS during SFV infection indicates that blood brain barrier integrity is also compromised (Erälinna et al., 1996; Soilu-Hänninen et al., 1994). While neurons are susceptible to SFV infection in an host age-dependent manner similar to SINV (Fazakerley et al., 1993; Fragkoudis et al., 2009; Oliver et al., 1997), unlike SINV and the other New World alphaviruses, oligodendrocytes represent the primary targeted cell in the CNS (Fazakerley et al., 2006; Fragkoudis et al., 2009; Sheahan et al., 1983). In optic nerves of SFV-infected BALB/c mice, oligodendrocytes develop swellings and vacuolations along internodal myelin sheaths, appearing more like immature cells (Butt et al., 1996); this suggests SFV infection induces the oligodendrocytes to dedifferentiate rather than die, and therefore the cells are capable of remyelination.
Numerous studies have shown that demyelination following SFV infection is primarily immune mediated. Mice lacking both T and B cells through genetics (SCID mice), irradiation, or cyclophosphamide administration show reduced and/or delayed pathology despite robust viral replication (Amor et al., 1996; Berger, 1980; Fazakerley and Webb, 1987a). Nude mice do not develop the perivascular inflammation or demyelinating lesions of immunocompetent mice (Amor et al., 1996; Fazakerley et al., 1988, 2006; Gates et al., 1984), and transfer of T cells, but not hyperimmune serum, back into nude or SCID mice results in demyelination (Amor et al., 1996; Fazakerley et al., 1983; Fazakerley and Webb, 1987a, 1987b). Depletion of CD8+ T cells, but not CD4+ T cells, reduces inflammation and eliminates demyelinating lesions, suggesting that CD8+ T cells are primarily responsible for the CNS damage (Subak-Sharpe et al., 1993). Production of MHC class I, which is expressed on oligodendrocytes and makes them a target for cytotoxic T cells (Fazakerley et al., 1993), is upregulated early in SFV infection (M. M. Morris et al., 1997), and interactions between lymphocytes and oligodendrocytes have been observed by electron microscopy at 7 DPI before demyelinating lesions are seen starting at 10 DPI (Pathak et al., 1983). While CNS expression of cytokines IL-1α, IL-1β, IL-10, and TGFβ increases early in infection, TNF-α, IL-2, and GM-CSF expression increases when cellular infiltrates are at their peak; IFN-γ and IL-6 first become apparent at 10 DPI immediately prior to the demyelination phase of disease, suggesting these cytokines may play a role in induction of CNS pathology (Blackman and Morris, 1984; McKimmic et al., 2005; Morris et al., 1997). Mice that lack the IFN-γ receptor, and therefore have deficient type II IFN signaling, demonstrate higher levels of neuronal necrosis associated with increased viral antigen and more macrophages and B cells in the brain, but comparable amounts of demyelination, following SFV infection (Keogh et al., 2003). Nude mice, which have impaired antibody class switching and therefore produce only antiviral IgM, μMT mice, which lack the ability to produce antibody, and SCID mice all demonstrate delayed viral clearance and/or viral persistence (Amor et al., 1996; Fazakerley et al., 2006; Fazakerley and Webb, 1987b; Fragkoudis et al., 2008). However, administration of polyclonal or monoclonal SFV antibody quickly reduces infectious virus to undetectable levels in SCID mice, indicating that, similar to SINV, anti-SFV antibody plays a major role in SFV clearance in the CNS (Amor et al., 1996; Fragkoudis et al., 2018).
SFV A7(74) has been shown to react immunologically with immune sera against several antigens in the CNS, including galactocerebroside (found on oligodendrocytes), glucocerebroside, total ganglioside, and GT1b ganglio-side (found on neurons), indicating the possibility of SFV infection creating an autoimmune state against cells present in the CNS (Webb et al., 1984). Administering anti-SFV hyperimmune serum, but not normal mouse serum, results in CNS tissue degeneration, but no demyelination or inflammatory response, in both SFV-infected and mock-infected control mice, suggesting autoantibodies play a role in pathology (Fazakerley and Webb, 1987b). C57BL/6 mice immunized with SFV proteins show lymphocyte proliferation against peptides on the SFV E2 glycoprotein and myelin oligodendrocyte glycoprotein, inducing SFV-likc demyelination; this suggests that SFV infection creates a molecular mimicry response against myelin oligodendrocyte glycoprotein, leading to autoimmune demyelination (Mokhtarian et al., 1999). Because of this, SFV has been used in mouse models of experimental autoimmune encephalomyelitis to evaluate the pathogenesis of autoimmune conditions such as multiple sclerosis (Wu et al., 1988). In C57BL/6 mice infected with SFV A7(74), CNS inflammation and virus clearance occur from 6 to 10 DPI, demyelination occurs from 15 to 21 DPI, and remyelination follows, with brains showing almost no lesions by 35 DPI (Mokhtarian et al., 2003). In this model, CD8+ T cell numbers peak between 8 and 15 DPI, concurrent with initial myelin injury, but B cells and microglia represent the predominant inflammatory cells present during the late period of demyelination at 21–28 DPI. γδ T cells and antibody against the SFV epitope, E2 Th peptidc2, have been shown to contribute to the remyelination process (Mokhtarian et al., 2012; Safavi et al., 2011) via an unknown mechanism. The C57BL/6 experimental autoimmune encephalomyelitis model using SFV highlights the multifaceted role the immune response plays in inducing pathology and the challenges that must be overcome in understanding and addressing immunc-mcdiatcd damage following alphavirus infection.
7. Conclusion and future directions
The increase in geographic expansion and frequency of outbreaks of disease induced by alphaviruses necessitates the development of effective therapies that not only curb the initial course of infection but address and/or prevent the development of chronic disease. Thanks to information assembled from reports of human cases and experimental infection of animals, we now know the immune system is responsible for a significant amount of clinical disease following alphavirus infection. Infection by alphaviruses promotes a proinflammatory state both systemically and in the tissues targeted by the viruses, and development of clinical disease coincides with the infiltration of immune cells, particularly T cells. Furthermore, blockage or inhibition of the inflammatory response helps mitigate the development and severity of clinical disease following alphavirus infection. However, the immune response is also responsible for restriction of viral replication, clearance of infectious virus and viral RNA, and prevention of persistent viral RNA reactivation and relapse of clinical disease. Some components of the immune response, such as TNF-α, IFN-γ, and CCL2, have been reported to be both pathogenic and protective during alphavirus infection. This double-edged sword highlights the complicated balance the immune response must navigate when responding to infection by an alphavirus, and understanding this interplay between pathology and protection is critical when considering potential treatments that modulate the immune response.
Many aspects of the role the immune response plays in development of clinical disease remain to be understood. For example, the pathomechanism of chronic disease following alphavirus infection, particularly with CHIKV, is currently unknown. Despite reports of debilitating pain, swelling, and stiffness in patients suffering from chronic chikungunya arthritis, damage to affected joints is not usually severe, raising the question of what unseen factors are mediating clinical symptoms. The recent increase in detection of CHIKV-associated GBS cases, especially in light of similar co-circulating arboviruses in affected regions, warrants further evaluation of the role of molecular mimicry in development of autoimmune disease following alphavirus infection. Understanding the effect of host genetics on the immune response and the role played in the range of disease severity and development of atypical manifestations is also in its infancy. Disparate manifestations of CHIKV-induced neurological disease in various geographical regions suggest that the diverse immune systems of the human populations among affected areas are likely responding to infection and mediating the course of disease in different manners. Robust animal models that effectively replicate the course of infection and development of disease and pathology, particularly chronic or atypical disease manifestations, will greatly assist in answering these questions, and critical evaluation, characterization, and modification of these models should be ongoing. Further illumination of the immunopathogenesis of alphavirus infection will help lead to therapies that effectively target the specific components of the immune response liable for tissue damage and pathology, leading to better disease outcomes and improved quality of life in affected individuals.
Acknowledgments
V.K.B. is supported by the National Institutes of Health (NIH) grants K01 OD026529 and U19 AI100625. M.T.H. is supported by NIH grants U19 AI100625 and U19 AI142759.
References
- Acevedo N, Waggoner J, Rodriguez M, Rivera L, Landivar J, Pinsky B, Zambrano H, 2017. Zika virus, Chikungunya virus, and Dengue virus in cerebrospinal fluid from adults with neurological manifestations, Guayaquil, Ecuador. Front. Microbiol 8, 42. 10.3389/fmicb.2017.00042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adams AP, Aronson JF, Tardif SD, Patterson JL, Brasky KM, Geiger R, Garza l., de M, Carrion R, Weaver SC, 2008. Common marmosets (Callithrix jacchus) as a nonhuman primate model to assess the virulence of eastern equine encephalitis virus strains. J. Virol 82, 9035–9042. 10.1128/JVI.00674-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aguilar MJ, 1970. Pathological changes in brain and other target organs of infant and weanling mice after infection with non-neuroadapted Western equine encephalitis virus. Infect. Immun 2, 533–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham CO, Birnbaum NS, Burmester GR, Bykerk VP, Cohen MD, Combe B, Costenbader KH, Dougados M, Emery P, Ferraccioli G, Hazes JMW, Hobbs K, Huizinga TWJ, Kavanaugh A, Kay J, Kvien TK, Laing T, Mease P, Ménard HA, Moreland LW, Naden RL, Pincus T, Smolen JS, Stanislawska-Biernat E, Symmons D, Tak PP, Upchurch KS, Vencovský J, Wolfe F, Hawker G, 2010. 2010 Rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheum. 62, 2569–2581. 10.1002/art.27584. [DOI] [PubMed] [Google Scholar]
- Alla SAO, Combe B, 2011. Arthritis after infection with chikungunya virus. Best Pract. Res. Cl. Rh 25, 337–346. 10.1016/j.berh.2011.03.005. [DOI] [PubMed] [Google Scholar]
- Amaral JK, Schoen RT, 2018. A case report of chikungunya fever, rheumatoid arthritis, and Felty’s Syndrome. Rheumatol. Ther 5, 293–301. 10.1007/s40744-018-0103-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amaral JK, Bilsborrow JB, Schoen RT, 2020a. Chronic Chikungunya Arthritis and Rheumatoid Arthritis: what they have in common. Am. J. Med 133, e91–e97. 10.1016/j.amjmed.2019.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amaral JK, Bingham CO, Schoen RT, 2020b. Successful methotrexate treatment of chronic chikungunya arthritis. J. Clin. Rheumatol 26, 119–124. 10.1097/RHU.0000000000000943. [DOI] [PubMed] [Google Scholar]
- Amor S, Scallan MF, Morris MM, Dyson H, Fazakerley JK, 1996. Role ofimmune responses in protection and pathogenesis during Semliki Forest virus encephalitis. J. Gen. Virol 77, 281–291. 10.1099/0022-1317-77-2-281. [DOI] [PubMed] [Google Scholar]
- Anand KS, Agrawal AK, Garg J, Dhamija RK, Mahajan RK, 2019. Spectrum of neurological complications in chikungunya fever: experience at a tertiary care centre and review of literature. Trop. Doct 49,79–84. 10.1177/0049475518825219. [DOI] [PubMed] [Google Scholar]
- Anaya J-M, Rodríguez Y, Monsalve DM, Vega D, Ojeda E, González-Bravo D, Rodríguez-Jiménez M, Pinto-Díaz CA, Chaparro P, Gunturiz ML, Ansari AA, Gershwin ME, Molano-González N, Ramirez-Santana C, Acosta-Ampudia Y, 2017. A comprehensive analysis and immunobiology of autoimmune neurological syndromes during the Zika virus outbreak in Cúcuta, Colombia. J. Autoimmun 77, 123–138. 10.1016/j.jaut.2016.12.007. [DOI] [PubMed] [Google Scholar]
- Anderson CR, Downs WG, Wattley GH, Ahin NW, Reese AA, 1957. Mayaro virus: a new human disease agent. II. Isolation from blood of patients in Trinidad, B.W.I. Am. J. Trop. Med. Hyg 6, 1012–1016. [DOI] [PubMed] [Google Scholar]
- Anfasa F, Lim SM, Fekken S, Wever R, Osterhaus ADME, Martina BEE, 2019. Characterization of antibody response in patients with acute and chronic chikungunya virus disease. J. Clin. Virol 117, 68–72. 10.1016/j.jcv.2019.06.001. [DOI] [PubMed] [Google Scholar]
- Arrigo NC, Adams AP, Weaver SC, 2010. Evolutionary patterns of eastern equine encephalitis virus in North versus South America suggest ecological differences and taxonomic revision. J. Virol 84, 1014–1025. 10.1128/JVI.01586-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aubry M, Cao-Lormeau VM, 2019. History of arthropod-borne virus infections in French Polynesia. New Microbes New Infect. 29, 100513. 10.1016/j.nmni.2019.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Azevedo MB, Coutinho MSC, Silva MAD, Arduini DB, Lima JDV, Monteiro R, Mendes BNB, Lemos MCF, Noronha CP, Saraceni V, 2018. Neurologie manifestations in emerging arboviral diseases in Rio de Janeiro City, Brazil, 2015–2016. Rev. Soc. Bras. Med. Trop 51, 347–351. 10.1590/0037-8682-0327-2017. [DOI] [PubMed] [Google Scholar]
- Azevedo RDSDS, Oliveira CS, Vasconcelos PFDC, 2015. Chikungunya risk for Brazil. Rev. Saude Publica 49,58. 10.1590/S0034-8910.2015049006219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Azevedo RSS, Silva EVP, Carvalho VL, Rodrigues SG, Nunes-Neto JP, Monteiro H, Peixoto VS, Chiang JO, Nunes MRT, Vasconcelos PFC, 2009. Mayaro fever virus, Brazilian Amazon. Emerg. Infect. Dis 15, 1830–1832. 10.3201/eid1511.090461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Badawi A, Ryoo SG, Vasileva D, Yaghoubi S, 2018. Prevalence of chronic comorbidities in chikungunya: a systematic review and meta-analysis. Int. J. Infect. Dis 67, 107–113. 10.1016/j.ijid.2017.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balavoine S, Pircher M, Hoen B, Herrmann-Storck C, Najioullah F, Madeux B, Signate A, Valentino R, Lannuzel A, Saint Louis M, Cassadou S, Cabie A, Schepers K, 2017. Guillain-Barré syndrome and chikungunya: description of all cases diagnosed during the 2014 outbreak in the french West Indies. Am. J. Trop. Med. Hyg 97, 356–360. 10.4269/ajtmh.15-0753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bantle CM, Phillips AT, Smeyne RJ, Rocha SM, Olson KE, Tjalkens RB, 2019. Infection with mosquito-bome alphavirus induces selective loss of dopaminergic neurons, neuroinflammation and widespread protein aggregation. NPJ Parkinsons Dis. 5, 20. 10.1038/s41531-019-0090-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barr KL, Khan E, Farooqi JQ, Imtiaz K, Prakoso D, Malik F, Lednicky JA, Long MT, 2018. Evidence of chikungunya virus disease in Pakistan since 2015 with patients demonstrating involvement of the central nervous system. Front. Public Health 6, 186. 10.3389/fpubh.2018.00186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartelloni PJ, McKinney RW, Duffy TP, Cole FE, 1970. An inactivated eastern equine encephalomyelitis vaccine propagated in chick-embryo cell culture. II. Clinical and serologic responses in man. Am. J. Trop. Med. Hyg 19, 123–126. 10.4269/ajtmh.1970.19.123. [DOI] [PubMed] [Google Scholar]
- Baxter VK, Glowinski R, Braxton AM, Potter MC, Slusher BS, Griffin DE, 2017. Glutamine antagonist-mediated immune suppression decreases pathology but delays virus clearance in mice during nonfatal alphavirus encephalomyelitis. Virology 508, 134–149. 10.1016/j.virol.2017.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baxter VK, Griffin DE, 2016. Interferon gamma modulation of disease manifestation and the local antibody response to alphavirus encephalomyelitis. J. Gen. Virol 97, 2908–2925. 10.1099/jgv.0.000613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baxter VK, Griffin DE, 2020. Interferon-gamma modulation of the local T cell response to alphavirus encephalomyelitis. Viruses 12, 113–125. 10.3390/v12010113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baxter VK, Troisi EM, Pate NM, Zhao JN, Griffin DE, 2018. Death and gastrointestinal bleeding complicate encephalomyelitis in mice with delayed appearance of CNS IgM after intranasal alphavirus infection. J. Gen. Virol 99, 309–320. 10.1099/jgv.0.001005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berger ML, 1980. Humoral and cell-mediated immune mechanisms in the production of pathology in avirulent Semliki Forest virus encephalitis. Infect. Immun 30, 244–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Binder GK, Griffin DE, 2001. Interferon-gamma-mediated site-specific clearance of alphavirus from CNS neurons. Science 293, 303–306. 10.1126/science.1059742. [DOI] [PubMed] [Google Scholar]
- Blackman MJ, Morris AG, 1984. Gamma interferon production and cytotoxicity of spleen cells from mice infected with Semliki Forest virus. J. Gen. Virol 65, 955–961. 10.1099/0022-1317-65-5-955. [DOI] [PubMed] [Google Scholar]
- Blakely PK, Delekta PC, Miller DJ, Irani DN, 2015. Manipulation of host factors optimizes the pathogenesis of western equine encephalitis virus infections in mice for antiviral drug development. J. Neurovirol 21, 43–55. 10.1007/s13365-014-0297-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bocan TM, Stafford RG, Brown JL, Akuoku Frimpong J, Basuli F, Hollidge BS, Zhang X, Raju N, Swenson RE, Smith DR, 2019. Characterization of brain inflammation, apoptosis, hypoxia, blood-brain barrier integrity and metabolism in Venezuelan Equine Encephalitis Virus (VEEV TC-83) exposed mice by in vivo positron emission tomography imaging. Viruses 11, 1052. 10.3390/v11111052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonifay T, Lienne J-F, Bagoée C, Santa F, Vesin G, Walter G, Nacher M, Vaserman N, Djossou F, Epelboin L, 2018. Prevalence and risk factors of post chi-kungunya rheumatic musculoskeletal disorders: a prospective follow-up study in French Guiana. Eur. J. Clin. Microbiol. Infect. Dis 37, 2159–2164. 10.1007/s10096-018-3353-0. [DOI] [PubMed] [Google Scholar]
- Booss J, Esiri MM, 2003. The Arboviruses. In: Viral Encephalitis in Humans. American Society for Microbiology, pp. 163–189. [Google Scholar]
- Borgherini G, Poubeau P, Jossaume A, Gouix A, Cotte L, Michault A, Arvin Berod C, Paganin F, 2008. Persistent arthralgia associated with chikungunya virus: a study of 88 adult patients on reunion island. Clin. Infect. Dis 47, 469–475. 10.1086/590003. [DOI] [PubMed] [Google Scholar]
- Bouquillard É, Combe B, 2009. A report of 21 cases of rheumatoid arthritis following Chikungunya fever. A mean follow-up of two years. Joint Bone Spine 76, 654–657. 10.1016/j.jbspin.2009.08.005. [DOI] [PubMed] [Google Scholar]
- Bourque PR, Chardon JW, Massie R, 2015. Autoimmune peripheral neuropathies. Clin. Chim. Acta 449, 37–42. 10.1016/j.cca.2015.02.039. [DOI] [PubMed] [Google Scholar]
- Bowen GS, Fashinell TR, Dean PB, Gregg MB, 1976. Clinical aspects of human Venezuelan equine encephalitis in Texas. Bull. Pan Am. Health Organ 10, 46–57. [PubMed] [Google Scholar]
- Braga JU, Bressan C, Dalvi APR, Calvet GA, Daumas RP, Rodrigues N, Wakimoto M, Nogueira RMR, Nielsen-Saines K, Brito C, Bispo de Filippis AM, Brasil P, 2017. Accuracy of Zika virus disease case definition during simultaneous Dengue and Chikungunya epidemics. PLoS One 12, e0179725. 10.1371/joumal.pone.0179725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Broeckel RM, Haese N, Ando T, Dmitriev I, Kreklywich CN, Powers J, Denton M, Smith P, Morrison TE, Heise M, DeFilippis V, Messaoudi I, Curiel DT, Streblow DN, 2019. Vaccine-induced skewing of T cell responses protects against chikungunya virus disease. Front. Immunol 10, 2563. 10.3389/fimmu.2019.02563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brooke CB, Deming DJ, Whitmore AC, White LJ, Johnston RE, 2010. T cells facilitate recovery from Venezuelan equine encephalitis virus-induced encephalomyelitis in the absence of antibody. J. Virol 84, 4556–4568. 10.1128/JVI.02545-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brooke CB, Schäfer A, Matsushima OK, White LJ, Johnston RE, 2012. Early activation of the host complement system is required to restrict central nervous system invasion and limit neuropathology during Venezuelan equine encephalitis virus infection. J. Gen. Virol 93, 797–806. 10.1099/vir.0.038281-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brummer-Korvenkontio M, Vapalahti O, Kuusisto P, Saikku P, Manni T, Koskela P, Nygren T, Brummer-Korvenkontio H, Vaheri A, 2002. Epidemiology of Sindbis virus infections in Finland 1981–96: possible factors explaining a peculiar disease pattern. Epidemiol. Infect 129, 335–345. 10.1017/s0950268802007409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bruyn HB, Lennette EH, 1953. Western equine encephalitis in infants; a report on three cases with sequelae. Calif. Med 79, 362–366. [PMC free article] [PubMed] [Google Scholar]
- Burdeinick-Kerr R, Wind J, Griffin DE, 2007. Synergistic roles of antibody and interferon in noncytolytic clearance of Sindbis virus from different regions of the central nervous system. J. Virol 81, 5628–5636. 10.1128/JVI.01152-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burrack KS, Montgomery SA, Homann D, Morrison TE, 2015. CD8+ T cells control Ross River virus infection in musculoskeletal tissues of infected mice. J. Immunol 194, 678–689. 10.4049/jimmunol.1401833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burt FJ, Chen W, Miner JJ, Lenschow DJ, Merits A, Schnetder E, Kohl A, Rudd PA, Taylor A, Herrero LJ, Zaid A, Ng LFP, Mahalingam S, 2017. Chikungunya virus: an update on the biology and pathogenesis of this emerging pathogen. Lancet Infect. Dis 17, e107–e117. 10.1016/S1473-3099(16)30385-1. [DOI] [PubMed] [Google Scholar]
- Butt AM, Tutton MG, Kirvell SL, Amor S, Jenkins HG, 1996. Morphology of oligodendrocytes during demyelination in optic nerves of mice infected with Semliki Forest virus. Neuropathol. Appi. Neurobiol 22, 540–547. 10.1111/j.1365-2990.1996.tb01133.x. [DOI] [PubMed] [Google Scholar]
- Byrnes AP, Durbin JE, Griffin DE, 2000. Control of Sindbis virus infection by antibody in interferon-deficient mice. J. Virol 74, 3905–3908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cain MD, Salimi H, Gong Y, Yang L, Hamilton SL, Heffeman JR, Hou J, Miller MJ, Klein RS, 2017. Virus entry and replication in the brain precedes blood-brain barrier disruption during intranasal alphavirus infection. J. Neuroimmunol 308, 118–130. 10.1016/j.jneuroim.2017.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calisher CH, 1994. Medically important arboviruses of the United States and Canada. Clin. Microbiol. Rev 7, 89–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Capasso A, Ompad DC, Vieira DL, Wilder-Smith A, Tozan Y, 2019. Incidence of Guillain-Barré Syndrome (GBS) in Latin America and the Caribbean before and during the 2015–2016 Zika virus epidemic: A systematic review and meta-analysis. PLoS Negl. Trop. Dis 13, e0007622. 10.1371/journal.pntd.0007622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carmen J, Rothstein JD, Kerr DA, 2009. Tumor necrosis factor-α modulates glutamate transport in the CNS and is a critical determinant of outcome from viral encephalomyelitis. Brain Res. 1263, 143–154. 10.1016/j.brainres.2009.01.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carrera J-P, Forrester N, Wang E, Vittor AY, Haddow AD, López-Vergès S, Abadía I, Castaño E, Sosa N, Báez C, Estripeaut D, Díaz Y, Beltrán D, Cisneros J, Cedeño HG, Travassos da Rosa AP, Hernandez H, Martínez-Torres AO, Tesh RB, Weaver SC, 2013. Eastern Equine Encephalitis in Latin America. N. Engl. J. Med 369, 732–744. 10.1056/NEJMoa1212628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carvalho K, Biancardi AL, Provenzano G, Moraes H, 2020. Acute disseminated encephalomyelitis (ADEM) associated with mosquito-bome diseases: chikungunya virus X yellow fever immunization. Rev. Soc. Bras. Med. Trop 53, e20190160. 10.1590/0037-8682-0160-2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Centers for Disease Control and Prevention, National Center for Emerging Zoonotic Infectious Diseases, Division of Vector-Borne Diseases, 2019. Eastern Equine Encephalitis [WWW Document]. Centers for Disease Control and Prevention, https://www.cdc.gov/easternequineencephalitis/tech/epi.html. accessed 2.14.20. [Google Scholar]
- Cerny T, Schwarz M, Schwarz U, Lemant J, Gérardin P, Keller E, 2017. The range of neurological complications in Chikungunya fever. Neurocrit. Care 27, 447–457. 10.1007/s12028-017-0413-8. [DOI] [PubMed] [Google Scholar]
- Chaaitanya IK, Muruganandam N, Sundaram SG, Kawalekar O, Sugunan AP, Manimunda SP, Ghosal SR, Muthumani K, Vijayachari P, 2011. Role of proinflammatory cytokines and chemokines in chronic arthropathy in CHIKV infection. Viral Immunol. 24, 265–271. 10.1089/vim.2010.0123. [DOI] [PubMed] [Google Scholar]
- Chanana B, Azad RV, Nair S, 2007. Bilateral macular choroiditis following Chikungunya virus infection. Eye 21, 1020–1021. 10.1038/sj.eye.6702863. [DOI] [PubMed] [Google Scholar]
- Chang AY, Encinales L, Porras A, Pachecho N, Reid SP, Martins KAO, Pacheco S, Bravo E, Navarno M, Rico A, Amdur R, Kamalopathy P, Firestein GS, Bethony JM, Simon GL, 2017. Frequency of chronic joint pain following chikungunya infection: a Colombian cohort study. Arthritis Rheumatol. 70, 578–584. 10.1002/art.40384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang AY, Martins KAO, Encinales L, Reid SP, Acuña M, Encinales C, Matranga CB, Pacheco N, Cure C, Shukla B, Ruiz Arteta T, Amdur R, Cazares LH, Gregory M, Ward MD, Porras A, Rico-Mendoza A, Dong L, Kenny T, Brueggemann E, Downey LG, Kamalapathy P, Lichtenberger P, Falls O, Simon GL, Bethony JM, Firestein GS, 2018. Chikungunya arthritis mechanisms in the Americas. Arthritis Rheumatol. 70, 585–593. 10.1002/art.40383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Charles PC, Trgovcich J, Davis NL, Johnston RE, 2001. Immunopathogenesis and immune modulation of Venezuelan equine encephalitis virus-induced disease in the mouse. Virology 284, 190–202. 10.1006/viro.2001.0878. [DOI] [PubMed] [Google Scholar]
- Charles PC, Walters E, Margolis F, Johnston RE, 1995. Mechanism of neuroinvasion of Venezuelan equine encephalitis virus in the mouse. Virology 208, 662–671. 10.1006/viro.1995.1197. [DOI] [PubMed] [Google Scholar]
- Chen W, Foo S-S, Li RW, Smith PN, Mahalingam S, 2014. Osteoblasts from osteoarthritis patients show enhanced susceptibility to Ross River virus infection associated with delayed type I interferon responses. Virol. J 11, 189. 10.1186/s12985-014-0189-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen W, Foo S-S, Taylor A, Lulla A, Merits A, Hueston L, Forwood MR, Walsh NC, Sims NA, Herrero LJ, Mahalingam S, 2015. Bindarit, an inhibitor of monocyte chemotactic protein synthesis, protects against bone loss induced by chikungunya virus infection. J. Virol 89, 581–593. 10.1128/JVI.02034-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen W, Foo S-S, Zaid A, Teng T-S, Herrero LJ, Wolf S, Tharmarajah K, Vu LD, van Vreden C, Taylor A, Freitas JR, Li RW, Woodruff TM, Gordon R, Ojcius DM, Nakaya HI, Kanneganti T-D, O’Neill LAJ, Robertson AAB, King NJ, Suhrbier A, Cooper MA, Ng LFP, Mahalingam S, 2017. Specific inhibition of NLRP3 in chikungunya disease reveals a role for inflammasomes in alphavirus-induced inflammation. Nat. Microbiol 2, 1435–1445. 10.1038/s41564-017-0015-4. [DOI] [PubMed] [Google Scholar]
- Chirathaworn C, Poovorawan Y, Lertmaharit S, Wuttirattanakowit N, 2013. Cytokine levels in patients with chikungunya virus infection. Asian Pac. J. Trop. Med 6, 631–634. 10.1016/S1995-7645(13)60108-X. [DOI] [PubMed] [Google Scholar]
- Chirathaworn C, Rianthavorn P, Wuttirattanakowit N, Poovorawan Y, 2010. Serum IL-18 and IL-18BP levels in patients with Chikungunya virus infection. Viral Immunol. 23, 113–117. 10.1089/vim.2009.0077. [DOI] [PubMed] [Google Scholar]
- Chopra A, Saluja M, Venugopalan A, 2014. Effectiveness of chloroquine and inflammatory cytokine response in patients with early persistent musculoskeletal pain and arthritis following chikungunya virus infection. Arthritis Rheumatol. 66, 319–326. 10.1002/art.38221. [DOI] [PubMed] [Google Scholar]
- Choudhary N, Makhija P, Puri V, Khwaja GA, Duggal A, 2016. An unusual case of myelitis with myositis. J. Clin. Diagn. Res 10, 19–20. 10.7860/JCDR/2016/13095.7777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chow A, Her Z, Ong EKS, Chen J-M, Dimatatac F, Kwek DJC, Barkham T, Yang H, Renia L, Leo Y-S, Ng LFP, 2011. Persistent arthralgia induced by Chikungunya virus infection is associated with interleukin-6 and granulocyte macrophage colony-stimulating factor. J. Infect. Dis 203, 149–157. 10.1093/infdis/jiq042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chua HH, Abdul Rashid K, Law WC, Hamizah A, Chem YK, Khairul AH, Chua KB, 2010. A fatal case of chikungunya virus infection with liver involvement. Med. J. Malaysia 65, 83–84. [PubMed] [Google Scholar]
- Clarris BJ, Doherty RL, Fraser JRE, French EL, Muirden KD, 1975. Epidemic polyarthritis: a cytological, virological, and immunochemical study. Aust. N. Z. J. Med 5, 450–457. [DOI] [PubMed] [Google Scholar]
- Contopoulos-Ioannidis D, Newman-Lindsay S, Chow C, Labeaud AD, 2018. Mother-to-child transmission of Chikungunya virus: a systematic review and metaanalysis. PLoS Negl. Trop. Dis 12, e0006510. 10.1371/joumal.pntd.0006510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Couderc T, Chrétien F, Schilte C, Disson O, Brigitte M, Guivel-Benhassine F, Touret Y, Barau G, Cayet N, Schuffenecker I, Desprès P, Arenzana-Seisdedos F, Michault A, Albert ML, Lecuit M, 2008. A mouse model for Chikungunya: young age and inefficient type-I interferon signaling are risk factors for severe disease. PLoS Pathog. 4, e29. 10.1371/journal.ppat.0040029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Couzigou B, Criquet-Hayot A, Javelle E, Tignac S, Mota E, Rigaud F, Alain A, Troisgros O, Pierre-Francois S, Abel S, Banydeen R, Cabie A, 2018. Occurrence of chronic stage chikungunya in the general population ofmartinique during the first 2014 epidemic: a prospective epidemiological study. Am. J. Trop. Med. Hyg 99, 182–190. 10.4269/ajtmh.17-0543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crosby L, Perreau C, Madeux B, Cossic J, Armand C, Herrmann-Storke C, Najioullah F, Valentino R, Thiéry G, 2016. Severe manifestations of chikungunya virus in critically ill patients during the 2013–2014 Caribbean outbreak. Int. J. Infect. Dis 48, 78–80. 10.1016/j.ijid.2016.05.010. [DOI] [PubMed] [Google Scholar]
- Dal Canto MC, Rabinowitz SG, 1981. Central nervous system demyelination in Venezuelan equine encephalomyelitis infection. J. Neurol. Sci 49, 397–418. 10.1016/0022-510x(81)90030-7. [DOI] [PubMed] [Google Scholar]
- Danes L, Rychterova V, Kufner J, Hruskova J, 1973. The role of the olfactory route on infection of the respiratory tract with Venezuelan equine encephalomyelitis virus in normal and operated Macaca rhesus monkeys. II. Results of histological examination. Acta Virol. 17, 57–60. [PubMed] [Google Scholar]
- Darman J, Backovic S, Dike S, Maragakis NJ, Krishnan C, Rothstein JD, Irani DN, Kerr DA, 2004. Viral-induced spinal motor neuron death is non-cell-autonomous and involves glutamate excitotoxicity. J. Neurosci 24, 7566–7575. 10.1523/JNEUROSCI.2002-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis LE, DeBiasi R, Goade DE, Haaland KY, Harrington JA, Hamar JB, Pergam SA, King MK, DeMasters BK, Tyler KL, 2006. West Nile virus neuroinvasive disease. Ann. Neurol 60, 286–300. 10.1002/ana.20959. [DOI] [PubMed] [Google Scholar]
- Davis NL, Grieder FB, Smith JF, Greenwald GF, Valenski ML, Sellon DC, Charles PC, Johnston RE, 1994. A molecular genetic approach to the study of Venezuelan equine encephalitis virus pathogenesis. Arch. Virol. Suppl 9, 99–109. 10.1007/978-3-7091-9326-6_11. [DOI] [PubMed] [Google Scholar]
- de Castro-Jorge LA, de Carvalho RVH, Klein TM, Hiroki CH, Lopes AH, Guimaräes RM, Fumagalh MJ, Floriano VG, Agostinho MR, Slhessarenko RD, Ramalho FS, Cunha TM, Cunha FQ, da Fonseca BAL, Zamboni DS, 2019. The NLRP3 inflammasome is involved with the pathogenesis of Mayaro virus. PLoS Pathog. 15, e1007934. 10.1371/joumal.ppat.1007934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Souza CG, da Costa JF, de Sousa Dantas D, de Abreu Freitas RP, Lopes JM, Okano AH, 2019. Evaluation of pain, functional capacity and kinesiophobia in women in the chronic stage of chikungunya virus infection: a cross-sectional study in northeastern Brazil. Acta Trop. 199, 104853. 10.1016/j.actatropica.2018.12.008. [DOI] [PubMed] [Google Scholar]
- Deaton AV, Craft S, Skenazy J, 1986. Enduring psychiatric and neuropsychologic sequelae in the post-encephalitis patient. Int. J. Psychiatry Med 16, 275–280. 10.2190/rv2q-ukfw-uy9l-5d8h. [DOI] [PubMed] [Google Scholar]
- Deeba IM, Hasan MM, Mosabbir AA, Siam MHB, Islam MS, Raheem E, Hossain MS, 2019. Manifestations of atypical symptoms of chikungunya during the Dhaka outbreak (2017) in Bangladesh. Am. J. Trop. Med. Hyg 100, 1545–1548. 10.4269/ajtmh.19-0122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Del Carpio-Orantes L, Pola-Ramirez MR, Garcia-Mendez S, Mata-Miranda MP, Perfecto-Arroyo MA, Solis-Sanchez I, Trujillo-Ortega BM, Gonzalez-Flores EE, 2018. The most frequent causative agents of Guillain-Barre syndrome in a Mexican general hospital. Rev. Neurol 67, 203–209. [PubMed] [Google Scholar]
- Del Piero F, Wilkins PA, Dubovi EJ, Biolatti B, Cantile C, 2001. Clinical, pathologic, immunohistochemical, and virologie findings of eastern equine encephalomyelitis in two horses. Vet. Pathol 38, 451–456. [DOI] [PubMed] [Google Scholar]
- Delgado Enciso I, Paz Michel B, Melnikov V, Guzmán-Esquivel J, Espinoza Gomez F, Soriano-Hemandez AD, Rodriguez-Sanchez IP, Martinez-Fierro ML, Ceja Espíritu G, Olmedo-Buenrostro BA, Galvan-Salazar HR, Delgado-Enciso OG, Delgado Enciso J, Lopez-Lemus UA, Montes-Galindo DA, 2018. Smoking and female sex as key risk factors associated with severe arthralgia in acute and chronic phases of Chikungunya virus infection. Exp. Ther. Med 15, 2634–2642. 10.3892/etm.2017.5668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deresiewicz RL, Thaler SJ, Hsu L, Zamani AA, 1997. Clinical and neuroradiographic manifestations of eastern equine encephalitis. N. Engl. J. Med 336, 1867–1874. 10.1056/NEJM199706263362604. [DOI] [PubMed] [Google Scholar]
- Dias C.N.d.S., Gois BM, Lima VS, Guerra-Gomes IC, Araújo JMG, Gomes J.d. A.S., Araújo DAM, Medeiros IA, de Azevedo F.d.L.A.A., Veras RC, Janebro DI, Amaral IPGD, Keesen TSL, 2018. Human CD8 T-cell activation in acute and chronic chikungunya infection. Immunology 155, 499–504. 10.1111/imm.12992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dietz WH, Alvarez O, Martin DH, Walton TE, Ackerman LJ, Johnson KM, 1978. Enzootic and epizootic Venezuelan equine encephalomyelitis virus in horses infected by peripheral and intrathecal routes. J. Infect. Dis 137, 227–237. 10.1093/infdis/137.3.227. [DOI] [PubMed] [Google Scholar]
- Dirlikov E, Major CG, Mayshack M, Medina N, Matos D, Ryff KR, Torres-Aponte J, Alkis R, Munoz-Jordan J, Colon-Sanchez C, Sahnas JL, Pastula DM, Garcia M, Segarra MO, Malave G, Thomas DL, Rodríguez-Vega GM, Luciano CA, Sejvar J, Sharp TM, Rivera-Garcia B, 2016. Guillain-Barré Syndrome during ongoing Zika Virus Transmission - Puerto Rico, January 1-July 31, 2016. Morb. Mortal. Wkly. Rep 65, 910–914. 10.15585/mmwr.mm6534el. [DOI] [PubMed] [Google Scholar]
- Duvignaud A, Fianu A, Bertolotti A, Jaubert J, Michault A, Poubeau P, Fred A, Méchain M, Gauzere BA, Favier F, Malvy D, Gérardin P, 2018. Rheumatism and chronic fatigue, the two facets of post-chikungunya disease: the TELECHIK cohort study on Reunion island. Epidemiol. Infect 146, 633–641. 10.1017/S0950268818000031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Earnest MP, Goolishian HA, Calverley JR, Hayes RO, Hill HR, 1971. Neurologic, intellectual, and psychologic sequelae following western encephalitis. A follow-up study of 35 cases. Neurology 21, 969–974. [DOI] [PubMed] [Google Scholar]
- Economopoulou A, Dominguez M, Helynck B, Sissoko D, Wichmann O, Quenel P, Germonneau P, Quatresous I, 2009. Atypical Chikungunya virus infections: clinical manifestations, mortality and risk factors for severe disease during the 2005–2006 outbreak on Réunion. Epidemiol. Infect 137, 534–541. 10.1017/S0950268808001167. [DOI] [PubMed] [Google Scholar]
- Elsinga J, Gerstenbluth I, van der Ploeg S, Halabi Y, Lourents NT, Burgerhof JG, van der Veen HT, Bailey A, Grobusch MP, Tami A, 2017. Long-term chikungunya sequelae in curaçao: burden, determinants, and a novel classification tool. J. Infect. Dis 216, 573–581. 10.1093/infdis/jix312. [DOI] [PubMed] [Google Scholar]
- Elsinga J, Halabi Y, Gerstenbluth I, Tami A, Grobusch MP, 2018. Consequences of a recent past dengue infection for acute and long-term chikungunya outcome: a retrospective cohort study in Curaçao. Travel Med. Infect. Dis 23, 34–43. 10.1016/j.tmaid.2018.03.008. [DOI] [PubMed] [Google Scholar]
- Endy TP, 2020. Viral febrile illnesses and emerging pathogens. In: Ryan ET, Hill DR, Solomon T, Endy TP, Aronson N (Eds.), Hunters Tropical Medicine and Emerging Infectious Disease, pp. 325–360. 10.1016/B978-0-323-55512-8.00036-3. [DOI] [Google Scholar]
- Erälinna JP, Soilu-Hänninen M, Röyttä M, Hukkanen V, Salmi AA, Salonen R, 1996. Blood-brain barrier breakdown and increased intercellular adhesion molecule (ICAM-1/CD54) expression after Semliki Forest (A7) virus infection facilitates the development of experimental allergic encephalomyelitis. J. Neuroimmunol 66, 103–114. 10.1016/0165-5728(96)00031-8. [DOI] [PubMed] [Google Scholar]
- Farias LABG, Ferragut JM, Pires Neto RDJ, 2018. Encephalitis and transverse myelitis in dengue and chikungunya coinfection. Rev. Soc. Bras. Med. Trop 51, 403. 10.1590/0037-8682-0259-2017. [DOI] [PubMed] [Google Scholar]
- Fazakerley JK, Amor S, Webb HE, 1983. Reconstitution of Semliki forest virus infected mice, induces immune mediated pathological changes in the CNS. Clin. Exp. Immunol 52, 115–120. [PMC free article] [PubMed] [Google Scholar]
- Fazakerley JK, Cotterill CL, Lee G, Graham A, 2006. Virus tropism, distribution, persistence and pathology in the corpus callosum of the Semliki Forest virus-infected mouse brain: a novel system to study virus-oligodendrocyte interactions. Neuropathol. Appl. Neurobiol 32, 397–409. 10.1111/j.1365-2990.2006.00739.x. [DOI] [PubMed] [Google Scholar]
- Fazakerley JK, Khalili-Shirazi A, Webb HE, 1988. Semliki Forest virus (A7[74]) infection of adult mice induces an immune-mediated demyelinating encephalomyelitis. Ann. N. Y. Acad. Sci 540, 672–673. 10.1111/j.1749-6632.1988.tb27208.x. [DOI] [PubMed] [Google Scholar]
- Fazakerley JK, Pathak S, Scallan M, Amor S, Dyson H, 1993. Replication of the A7(74) strain of Semliki Forest virus is restricted in neurons. Virology 195, 627–637. 10.1006/viro.1993.1414. [DOI] [PubMed] [Google Scholar]
- Fazakerley JK, Webb HE, 1987a. Semliki Forest virus induced, immune mediated demyelination: the effect of irradiation. Br. J. Exp. Pathol 68, 101–113. [PMC free article] [PubMed] [Google Scholar]
- Fazakerley JK, Webb HE, 1987b. Semliki Forest virus-induced, immune-mediated demyelination: adoptive transfer studies and viral persistence in nude mice. J. Gen. Virol 68, 377–385. 10.1099/0022-1317-68-2-377. [DOI] [PubMed] [Google Scholar]
- Fine DL, Roberts BA, Terpening SJ, Mott J, Vasconcelos D, House RV, 2008. Neurovirulence evaluation of Venezuelan equine encephalitis (VEE) vaccine candidate V3526 in nonhuman primates. Vaccine 26, 3497–3506. 10.1016/j.vaccine.2008.04.044. [DOI] [PubMed] [Google Scholar]
- Finley KH, Longshore WA, Palmer RJ, Cook RE, Riggs N, 1955. Western equine and St. Louis encephalitis; preliminary report of a clinical follow-up study in California. Neurology 5, 223–235. [DOI] [PubMed] [Google Scholar]
- Forechi L, Silveira-Nunes G, Barbosa MA, Barbosa ÉG, Santos DLD, Vieira ER, Barbosa AC, 2018. Pain, balance, grip strength and gait parameters of older adults with and without post-chikungunya chronic arthralgia. Trop. Med. Int. Health 23, 1394–1400. 10.1111/tmi.13154. [DOI] [PubMed] [Google Scholar]
- Forrester NL, Palacios G, Tesh RB, Savji N, Guzman H, Sherman M, Weaver SC, Lipkin WI, 2012. Genome-scale phylogeny of the alphavirus genus suggests a marine origin. J. Virol 86, 2729–2738. 10.1128/JVI.05591-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fragkoudis R, Ballany CM, Boyd A, Fazakerley JK, 2008. In Semliki Forest virus encephalitis, antibody rapidly clears infectious virus and is required to eliminate viral material from the brain, but is not required to generate lesions of demyelination. J. Gen. Virol 89, 2565–2568. 10.1099/vir.0.2008/002238-0. [DOI] [PubMed] [Google Scholar]
- Fragkoudis R, Dixon-Ballany CM, Zagrajek AK, Kedzierski L, Fazakerley JK, 2018. Following acute encephalitis, Semliki Forest virus is undetectable in the brain by infec-tivity assays but functional virus RNA capable of generating infectious virus persists for life. Viruses 10, 273. 10.3390/v10050273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fragkoudis R, Tamberg N, Siu R, Kiiver K, Kohl A, Merits A, Fazakerley JK, 2009. Neurons and oligodendrocytes in the mouse brain differ in their ability to replicate Semliki Forest virus. J. Neurovirol 15, 57–70. 10.1080/13550280802482583. [DOI] [PubMed] [Google Scholar]
- Franklin RP, Kinde H, Jay MT, Kramer LD, Green E-GN, Chiles RE, Osdund E, Husted S, Smith J, Parker MD, 2002. Eastern equine encephalomyelitis virus infection in a horse from California. Emerg. Infect. Dis 8, 283–288. 10.3201/eid0803.010199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fraser JR, Becker GJ, 1984. Mononuclear cell types in chronic synovial effusions of Ross River virus disease. Aust. N. Z. J. Med 14, 505–506. 10.1111/j.1445-5994.1984.tb03629.x. [DOI] [PubMed] [Google Scholar]
- Fraser JR, Cunningham AL, Clarris BJ, Aaskov JG, Leach R, 1981. Cytology of synovial effusions in epidemic polyarthritis. Aust. N. Z. J. Med 11, 168–173. [DOI] [PubMed] [Google Scholar]
- Fulton JS, Burton AN, 1953. After effects of western equine encephalomyelitis infection in man. Can. Med. Assoc. J 69, 268–272. [PMC free article] [PubMed] [Google Scholar]
- Ganesan K, Diwan A, Shankar SK, Desai SB, Sainani GS, Katrak SM, 2008. Chikungunya encephalomyeloradiculitis: report of 2 cases with neuroimaging and 1 case with autopsy findings. Am. J. Neuroradiol 29, 1636–1637. 10.3174/ajnr.A1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ganjian N, Riviere-Cinnamond A, 2020. Mayaro virus in Latin America and the Caribbean. Rev. Panam Salud Publica 44, e14. 10.26633/RPSP.2020.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ganu MA, Ganu AS, 2011. Post-chikungunya chronic arthritis—our experience with DMARDs over two year follow up. J. Assoc. Physicians India 59, 83–86. [PubMed] [Google Scholar]
- Gardner CL, Burke CW, Higgs ST, Klimstra WB, Ryman KD, 2012. Interferon-alpha/beta deficiency greatly exacerbates arthritogenic disease in mice infected with wild-type chikungunya virus but not with the cell culture-adapted live-attenuated 181/25 vaccine candidate. Virology 425, 103–112. 10.1016/j.virol.2011.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gardner CL, Burke CW, Tesfay MZ, Glass PJ, Klimstra WB, Ryman KD, 2008. Eastern and Venezuelan equine encephalitis viruses differ in their ability to infect dendritic cells and macrophages: impact of altered cell tropism on pathogenesis. J. Virol 82, 10634–10646. 10.1128/JVI.01323-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gardner J, Anraku I, Le TT, Larcher T, Major L, Roques P, Schroder WA, Higgs S, Suhrbier A, 2010. Chikungunya virus Arthritis in adult wild-type mice. J. Virol 84, 8021–8032. 10.1128/JVI.02603-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gates MC, Sheahan BJ, Atkins GJ, 1984. The pathogenicity of the M9 mutant of Semliki Forest virus in immune-compromised mice. J. Gen. Virol 65, 73–80. 10.1099/0022-1317-65-l-73. [DOI] [PubMed] [Google Scholar]
- Gauri LA, Thaned A, Fatima Q, Yadav H, Singh A, Jaipal HP, Chaudhary A, 2016. Clinical spectrum of chikungunya in Bikaner (North Western India) in 2006 and follow up of patients for five years. J. Assoc. Physicians India 64, 22–25. [PubMed] [Google Scholar]
- Gérardin P, Barau G, Michault A, Bintner M, Randrianaivo H, Choker G, Lenglet Y, Touret Y, Bouveret A, Grivard P, Le Roux K, Blanc S, Schuffenecker I, Couderc T, Arenzana-Seisdedos F, Lecuit M, Robillard P-Y, 2008. Multidisciplinary prospective study of mother-to-child chikungunya virus infections on the island of La Réunion. PLoS Med. 5, e60. 10.1371/joumal.pmed.0050060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gérardin P, Fianu A, Malvy D, Mussard C, Boussaïd K, Rollot O, Michault A, Gauzere BA, Bréart G, Favier F, 2011. Perceived morbidity and community burden after a Chikungunya outbreak: the TELECHIK survey, a population-based cohort study. BMC Med. 9, 5. 10.1186/1741-7015-9-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gleiser CA, Gochenour WS, Berge TO, TIGERTT WD, 1962. The comparative pathology of experimental Venezuelan equine encephalomyelitis infection in different animal hosts. J. Infect. Dis 110, 80–97. 10.1093/infdis/110.1.80. [DOI] [PubMed] [Google Scholar]
- Go YY, Balasuriya UBR, Lee C-K, 2014. Zoonotic encephalitides caused by arboviruses: transmission and epidemiology of alphaviruses and flaviviruses. Clin. Exp. Vaccine Res 3, 58–77. 10.7774/cevr.2014.3.l.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- González-Galván LM, Arredondo-Ruiz P, Chac-Lezama G, Molina-Villena AA, Saenz-Castro M, Perez-Garcia JA, Alonso-Martinez D, Ramirez-Valadez C, Pena-Perez CA, 2017. Transverse myelitis associated to chikungunya virus infection. Med. Int. Mex 33, 109–115. [Google Scholar]
- Gorelkin L, Jahrling PB, 1975. Virus-initiated septic shock. Acute death of Venezuelan encephalitis virus-infected hamsters. Lab. Invest 32, 78–85. [PubMed] [Google Scholar]
- Gray MP, Gorelick MH, 2016. Acute disseminated encephalomyelitis. Pediatr. Emerg. Care 32, 395–400. 10.1097/PEC.0000000000000825. [DOI] [PubMed] [Google Scholar]
- Green MJ, 2003. Serum MMP-3 and MMP-1 and progression of joint damage in early rheumatoid arthritis. Rheumatology 42, 83–88. 10.1093/rheumatology/keg037. [DOI] [PubMed] [Google Scholar]
- Greene IP, Lee E-Y, Prow N, Ngwang B, Griffin DE, 2008. Protection from fatal viral encephalomyelitis: AMPA receptor antagonists have a direct effect on the inflammatory response to infection. Proc. Natl. Acad. Sci. U. S. A 105, 3575–3580. 10.1073/pnas.0712390105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greenlee JE, 2014. The equine encephalitides. Handb. Clin. Neurol 123, 417–432. 10.1016/B978-0-444-53488-0.00019-5. [DOI] [PubMed] [Google Scholar]
- Grieder FB, Davis BK, Zhou XD, Chen SJ, Finkelman FD, Gause WC, 1997. Kinetics of cytokine expression and regulation of host protection following infection with molecularly cloned Venezuelan equine encephalitis virus. Virology 233, 302–312. 10.1006/viro.1997.8617. [DOI] [PubMed] [Google Scholar]
- Griffin DE, 1976. Role of the immune response in age-dependent resistance of mice to encephalitis due to Sindbis virus. J. Infect. Dis 133, 456–464. [DOI] [PubMed] [Google Scholar]
- Griffin DE, 2013. Alphaviruses. In: Knipe DM, Howley PM (Eds.), Fields Virology. Lippincott Williams & Wilkins, Philadelphia, pp. 652–686. [Google Scholar]
- Griffin DE, Hardwick JM, 1997. Regulators of apoptosis on the road to persistent alphavirus infection. Annu. Rev. Microbiol 51, 565–592. 10.1146/annurev.micro.51.1.565. [DOI] [PubMed] [Google Scholar]
- Griffin DE, Johnson RT, 1977. Role of the immune response in recovery from Sindbis virus encephalitis in mice. J. Immunol 118, 1070–1075. [PubMed] [Google Scholar]
- Gualberto Cavalcanti N, MeloVilar K, Branco Pinto Duarte AL, de Melo Rêgo MJB, Cristiny Pereira M, da Rocha Pitta I, Diniz Lopes Marques C, Caldino da Rocha Pitta M, 2019. IL-27 in patients with Chikungunya fever: a possible chronicity biomarker? Acta Trop. 196, 48–51. 10.1016/j.actatropica.2019.05.005. [DOI] [PubMed] [Google Scholar]
- Gunn BM, Jones JE, Shabman RS, Whitmore AC, Sarkar S, Blevins LK, Morrison TE, Heise MT, 2018. Ross River virus envelope glycans contribute to disease through activation of the host complement system. Virology 515, 250–260. 10.1016/j.virol.2017.12.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gunn BM, Morrison TE, Whitmore AC, Blevins LK, Hueston L, Fraser RJ, Herrero LJ, Ramirez R, Smith PN, Mahalingam S, Heise MT, 2012. Mannose binding lectin is required for alphavirus-induced arthritis/myositis. PLoS Pathog. 8, e1002586. 10.1371/journal.ppat.1002586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gupta P, Sharma A, Han J, Yang A, Bhomia M, Knollmann-Ritschel B, Puri RK, Maheshwari RK, 2017. Differential host gene responses from infection with neuro-virulent and partially-neurovirulent strains of Venezuelan equine encephalitis virus. BMC Infect. Dis 17, 309. 10.1186/s12879-017-2355-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haese NN, Broeckel RM, Hawman DW, Heise MT, Morrison TE, Streblow DN, 2016. Animal models of chikungunya virus infection and disease. J. Infect. Dis 214, S482–S487. 10.1093/infdis/jiw284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hahn CS, Lustig S, Strauss EG, Strauss JH, 1988. Western equine encephalitis virus is a recombinant virus. Proc. Natl. Acad. Sci. U. S. A 85, 5997–6001. 10.1073/pnas.85.16.5997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hameed S, Memon M, Imtiaz H, Kanwar D, 2019. Longitudinally extensive transverse myelitis with seropositive chikungunya. BMJ Case Rep. 12. 10.1136/bcr-2019-231745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harley D, Sleigh A, Ritchie S, 2001. Ross River virus transmission, infection, and disease: a cross-disciplinary review. Clin. Microbiol. Rev 14, 909–932. 10.1128/CMR.14.4.909-932.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Havert MB, Schofield B, Griffin DE, Irani DN, 2000. Activation of divergent neuronal cell death pathways in different target cell populations during neuroadapted sindbis virus infection of mice. J. Virol 74, 5352–5356. 10.1128/JVI.74.11.5352-5356.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hawman DW, Stoermer KA, Montgomery SA, Pal P, Öko L, Diamond MS, Morrison TE, 2013. Chronic joint disease caused by persistent chikungunya virus infection is controlled by the adaptive immune response. J. Virol 87, 13878–13888. 10.1128/JVI.02666-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heath CJ, Lowther J, Noël TP, Mark-George I, Boothroyd DB, Mitchell G, MacPherson C, Desiree LaBeaud A, 2018. The identification of risk factors for chronic Chikungunya Arthralgia in Grenada, West Indies: a cross-sectional cohort study. Open Forum Infect. Dis 5, ofx234. 10.1093/ofid/ofx234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Her Z, Teng T-S, Tan JJL, Teo T-H, Kam Y-W, Lum F-M, Lee WWL, Gabriel C, Melchiotti R, Andiappan AK, Lulla V, Lulla A, Win MK, Chow A, Biswas SK, Leo Y-S, Lecuit M, Merits A, Renia L, Ng LFP, 2015. Loss of TLR3 aggravates CHIKV replication and pathology due to an altered virus-specific neutralizing antibody response. EMBO Mol. Med 7, 24–41. 10.15252/emmm.201404459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herzon H, Shelton JT, Bruyn HB, 1957. Sequelae of western equine and other arthropod-borne encephalitides. Neurology 7, 535–548. 10.1212/wnl.7.8.535. [DOI] [PubMed] [Google Scholar]
- Hirsch RL, Griffin DE, Winkelstein JA, 1980. Role of complement in viral infections: participation of terminal complement components (C5 to C9) in recovery of mice from Sindbis virus infection. Infect. Immun 30, 899–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoarau JJ, Gay F, Pellé O, Samri A, Jaffar-Bandjee MC, Gasque P, Autran B, 2013. Identical strength of the T cell responses against E2, nsP1 and capsid CHIKV proteins in recovered and chronic patients after the epidemics of 2005–2006 in La Reunion Island. PLoS One 8, e84695. 10.1371/joumal.pone.0084695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoarau JJ, Jaffar-Bandjee MC, Krejbich-Trotot P, Das T, Li-Pat-Yuen G, Dassa B, Denizot M, Guichard E, Ribera A, Henni T, Tallet F, Moiton MP, Gauzere BA, Bruniquet S, Jaffar Bandjee Z, Morbidelli P, Martigny G, Jolivet M, Gay F, Grandadam M, Tolou H, Vieillard V, Debré P, Autran B, Gasque P, 2010. Persistent chronic inflammation and infection by Chikungunya arthritogenic alphavirus in spite of a robust host immune response. J. Immunol 184, 5914–5927. 10.4049/jimmunol.0900255. [DOI] [PubMed] [Google Scholar]
- Hollidge BS, González-Scarano F, Soldan SS, 2010. Arboviral encephalitides: transmission, emergence, and pathogenesis. J. Neuroimmune Pharmacol 5, 428–442. 10.1007/s11481-010-9234-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Honnold SP, Mossel EG, Bakken RR, Fisher D, Lind CM, Cohen JW, Eccleston LT, Spurgers KB, Erwin-Cohen R, Bradfute SB, Maheshwari RK, Glass PJ, 2015a. Eastern equine encephalitis virus in mice I: clinical course and outcome are dependent on route of exposure. Virol. J 12, 152. 10.1186/s12985-015-0386-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Honnold SP, Mossel EC, Bakken RR, Lind CM, Cohen JW, Eccleston LT, Spurgers KB, Erwin-Cohen R, Glass PJ, Maheshwari RK, 2015b. Eastern equine encephalitis virus in mice II: pathogenesis is dependent on route of exposure. Virol. J 12, 154. 10.1186/s12985-015-0385-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoz J.M.d.L., Bayona B, Viloria S, Accini JL, Juan-Vergara HS, Viasus D, 2015. Fatal cases of Chikungunya virus infection in Colombia: diagnostic and treatment challenges. J. Clin. Virol 69, 27–29. 10.1016/j.jcv.2015.05.021. [DOI] [PubMed] [Google Scholar]
- Hughes RA, Cornblath DR, 2005. Guillain-Barré syndrome. Lancet 366, 1653–1666. 10.1016/S0140-6736(05)67665-9. [DOI] [PubMed] [Google Scholar]
- Huits R, De Kort J, Van Den Berg R, Chong L, Tsoumanis A, Eggermont K, Bartholomeeusen K, Ariën KK, Jacobs J, Van Esbroeck M, Bottieau E, Cnops L, 2018. Chikungunya virus infection in Aruba: diagnosis, clinical features and predictors of post-chikungunya chronic polyarthralgia. PLoS One 13, e0196630. 10.1371/journal.pone.0196630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Irani DN, Griffin DE, 1991. Isolation of brain parenchymal lymphocytes for flow cytometric analysis. Application to acute viral encephalitis. J. Immunol. Methods 139, 223–231. [DOI] [PubMed] [Google Scholar]
- Irani DN, Griffin DE, 1996. Regulation of lymphocyte homing into the brain during viral encephalitis at various stages of infection. J. Immunol 156, 3850–3857. [PubMed] [Google Scholar]
- Jackson AC, Moench TR, Griffin DE, Johnson RT, 1987. The pathogenesis of spinal cord involvement in the encephalomyelitis of mice caused by neuroadapted Sindbis virus infection. Lab. Invest 56, 418–423. [PubMed] [Google Scholar]
- Jackson AC, Moench TR, Trapp BD, Griffin DE, 1988. Basis of neurovirulence in Sindbis virus encephalomyelitis of mice. Lab. Invest 58, 503–509. [PubMed] [Google Scholar]
- Jackson AC, Rossiter JP, 1997. Apoptotic cell death is an important cause of neuronal injury in experimental Venezuelan equine encephalitis virus infection of mice. Acta Neuropathol. 93, 349–353. 10.1007/s004010050626. [DOI] [PubMed] [Google Scholar]
- Jackson AC, SenGupta SK, Smith JF, 1991. Pathogenesis of Venezuelan equine encephalitis virus infection in mice and hamsters. Vet. Pathol 28, 410–418. 10.1177/030098589102800509. [DOI] [PubMed] [Google Scholar]
- Jacobs BC, Rothbarth PH, van der Meché FGA, Herbrink P, Schmitz PIM, de Klerk MA, van Doorn PA, 1998. The spectrum of antecedent infections in Guillain-Barré syndrome: a case-control study. Neurology 51, 1110–1115. 10.1212/WNL.51.4.1110. [DOI] [PubMed] [Google Scholar]
- Jahrling PB, Scherer WF, 1973. Histopathology and distribution of viral antigens in hamsters infected with virulent and benign Venezuelan encephalitis viruses. Am. J. Pathol 72, 25–38. [PMC free article] [PubMed] [Google Scholar]
- Julander JG, Siddharthan V, Blatt LM, Schafer K, Sidwell RW, Morrey JD, 2007. Effect of exogenous interferon and an interferon inducer on western equine encephalitis virus disease in a hamster model. Virology 360, 454–460. 10.1016/j.virol.2006.10.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kam Y-W, Simarmata D, Chow A, Her Z, Teng T-S, Ong EKS, Renia L, Leo Y-S, Ng LFP, 2012. Early appearance of neutralizing immunoglobulin G3 antibodies is associated with chikungunya virus clearance and long-term clinical protection. J. Infect. Dis 205, 1147–1154. 10.1093/infdis/jis033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kashyap RS, Morey S, Bhullar S, Baheti N, Chandak N, Purohit H, Taori G, Daginawala H, 2014. Determination of Toll-like receptor-induced cytokine profiles in the blood and cerebrospinal fluid of Chikungunya patients. Neuroimmunomodulation 21, 338–346. 10.1159/000358240. [DOI] [PubMed] [Google Scholar]
- Kehn-Hall K, Narayanan A, Lundberg L, Sampey G, Pinkham C, Guendel I, Van Duyne R, Senina S, Schultz KL, Stavale E, Aman MJ, Bailey C, Kashanchi F, 2012. Modulation of GSK-3β activity in Venezuelan equine encephalitis virus infection. PLoS One 7, e34761. 10.1371/journal.pone.0034761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelly WR, Blakemore WF, Jagelman S, Webb HE, 1982. Demyelination induced in mice by avirulent Semliki Forest virus. II. An ultrastructural study of focal demyelination in the brain. Neuropathol. Appi. Neurobiol 8, 43–53. 10.1111/j.1365-2990.1982.tb00256.x. [DOI] [PubMed] [Google Scholar]
- Kelvin AA, Banner D, Silvi G, Moro ML, Spataro N, Gaibani P, Cavrini F, Pierro A, Rossini G, Cameron MJ, Bermejo-Martin JF, Paquette SG, Xu L, Danesh A, Farooqui A, Borghetto I, Kelvin DJ, Sambri V, Rubino S, 2011. Inflammatory cytokine expression is associated with chikungunya virus resolution and symptom severity. PLoS Negl. Trop. Dis 5, e1279. 10.1371/joumal.pntd.0001279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keogh B, Atkins GJ, Mills K, Sheahan BJ, 2003. Role ofinterferon-γ and nitric oxide in the neuropathogenesis of avirulent Semliki Forest virus infection. Neuropathol. Appi. Neurobiol 29, 553–562. 10.1046/j.0305-1846.2003.00492.x. [DOI] [PubMed] [Google Scholar]
- Kerr DA, Larsen T, Cook SH, Fannjiang Y-R, Choi E, Griffin DE, Hardwick JM, Irani DN, 2002. BCL-2 and BAX protect adult mice from lethal Sindbis virus infection but do not protect spinal cord motor neurons or prevent paralysis. J. Virol 76, 10393–10400. 10.1128/jvi.76.20.10393-10400.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kimura T, Griffin DE, 2000. The role of CD8(+) T cells and major histocompatibility complex class I expression in the central nervous system of mice infected with neuro-virulent Sindbis virus. J. Virol 74, 6117—6125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kimura T, Griffin DE, 2003. Extensive immune-mediated hippocampal damage in mice surviving infection with neuroadapted Sindbis virus. Virology 311, 28–39. 10.1016/S0042-6822(03)00110-7. [DOI] [PubMed] [Google Scholar]
- Kindhauser MK, Allen T, Frank V, Santhana RS, Dye C, 2016. Zika: the origin and spread ofa mosquito-bome virus. Bull. World Health Organ 94, 675–686C. 10.2471/BLT.16.171082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koeltz A, Lastère S, Jean-Baptiste S, 2018. Intensive care admissions for severe chikungunya virus infection, french Polynesia. Emerg. Infect. Dis 24, 794–796. 10.3201/eid2404.161536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krishnan C, Kaplin AI, Pardo CA, Kerr DA, Keswani SC, 2006. Demyelinating disorders: update on transverse myelitis. Curr. Neurol. Neurosci. Rep 6, 236–243. 10.1007/s11910-006-0011-l. [DOI] [PubMed] [Google Scholar]
- Kulcsar KA, Baxter VK, Abraham R, Nelson A, Griffin DE, 2015. Distinct immune responses in resistant and susceptible strains of mice during neurovirulent alphavirus encephalomyehtis. J. Virol 89, 8280–8291. 10.1128/JVI.00173-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kulcsar KA, Baxter VK, Greene IP, Griffin DE, 2014. Interleukin-10 modulation of pathogenic Th17 cells during fatal alphavirus encephalomyehtis. Proc. Nad. Acad. Sci. U. S. A 111, 16053–16058. 10.1073/pnas.1418966111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kulcsar KA, Griffin DE, 2016. T cell-derived interleukin-10 is an important regulator of the Th17 response during lethal alphavirus encephalomyehtis. J. Neuroimmunol 295–296, 60–67. 10.1016/j.jneuroim.2016.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar R, Rajvanshi P, Khosla H, Arora S, 2019. Neuro-chikungunya: acute transverse myelopathy associated with chikungunya virus infection. J. Assoc. Physicians India 67, 84–85. [PubMed] [Google Scholar]
- Labrada L, Liang XH, Zheng W, Johnston C, Levine B, 2002. Age-dependent resistance to lethal alphavirus encephalitis in mice: analysis of gene expression in the central nervous system and identification of a novel interferon-inducible protective gene, mouse ISG12. J. Virol 76, 11688–11703. 10.1128/jvi.76.22.11688-11703.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laine M, Luukkainen R, Toivanen A, 2004. Sindbis viruses and other alphaviruses as cause of human arthritic disease. J. Intern. Med 256, 457–471. 10.1111/j.1365-2796.2004.01413.x. [DOI] [PubMed] [Google Scholar]
- Lalitha P, Rathinam S, Banushree K, Maheshkumar S, Vijayakumar R, Sathe P, 2007. Ocular involvement associated with an epidemic outbreak of chikungunya virus infection. Am. J. Ophthalmol 144, 552–556. 10.1016/j.ajo.2007.06.002. [DOI] [PubMed] [Google Scholar]
- Lebrun G, Chadda K, Reboux A-H, Martinet O, Gauzere BA, 2009. Guillain-Barré syndrome after chikungunya infection. Emerg. Infect. Dis 15, 495–496. 10.3201/eid1503.071482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee E-Y, Schultz KLW, Griffin DE, 2013. Mice deficient in interferon-gamma or interferon-gamma receptor 1 have distinct inflammatory responses to acute viral encephalomyehtis. PLoS One 8, e76412. 10.1371/journal.pone.0076412.g010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee JM, Zipfel GJ, Choi DW, 1999. The changing landscape ofischaemic brain injury mechanisms. Nature 399, A7–14. [DOI] [PubMed] [Google Scholar]
- Lee WWL, Teo T-H, Her Z, Lum F-M, Kam Y-W, Haase D, Renia L, Rötzschke O, Ng LFP, 2015. Expanding regulatory T cells alleviates chikungunya virus-induced pathology in mice. J. Virol 89, 7893–7904. 10.1128/JVI.00998-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levine B, Griffin DE, 1992. Persistence of viral RNA in mouse brains after recovery from acute alphavirus encephalitis. J. Virol 66, 6429–6435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levine B, Hardwick JM, Trapp BD, Crawford TO, Bollinger RC, Griffin DE, 1991. Antibody-mediated clearance of alphavirus infection from neurons. Science 254, 856–860. [DOI] [PubMed] [Google Scholar]
- Levitt NH, Miller HV, Edelman R, 1979. Interaction of alphaviruses with human peripheral leukocytes: in vitro replication of Venezuelan equine encephalomyehtis virus in monocyte cultures. Infect. Immun 24, 642–646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lewis J, Wesselingh SL, Griffin DE, Hardwick JM, 1996. Alphavirus-induced apo-ptosis in mouse brains correlates with neurovirulence. J. Virol 70, 1828–1835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lémant J, Boisson V, Winer A, Thibault L, André H, Tixier F, Lemercier M, Antok E, Cresta MP, Grivard P, Besnard M, Rollot O, Favier F, Huerre M, Campinos JL, Michault A, 2008. Serious acute chikungunya virus infection requiring intensive care during the Reunion Island outbreak in 2005–2006. Crit. Care Med 36, 2536–2541. 10.1097/CCM.0b013e318183f2d2. [DOI] [PubMed] [Google Scholar]
- León CA, 1975. Sequelae of Venezuelan equine encephalitis in humans: a four year follow-up. Int. J. Epidemiol 4, 131–140. [DOI] [PubMed] [Google Scholar]
- Liang XH, Goldman JE, Jiang HH, Levine B, 1999. Resistance of interleukin-1beta-deficient mice to fatal Sindbis virus encephalitis. J. Virol 73, 2563–2567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lidbury BA, Simeonovic C, Maxwell GE, Marshall ID, Hapel AJ, 2000. Macrophage-induced muscle pathology results in morbidity and mortality for Ross River virus-infected mice. J. Infect. Dis 181, 27–34. 10.1086/315164. [DOI] [PubMed] [Google Scholar]
- Lim EXY, Lee WS, Madzokere ET, Herrero LJ, 2018. Mosquitoes as suitable vectors for alphaviruses. Viruses 10, 84. 10.3390/v10020084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lima M.E.d.S., Bachur TPR, Aragão GF, 2019. Guillain-Barre syndrome and its correlation with dengue, Zika and chikungunya viruses infection based on a literature review of reported cases in Brazil. Acta Trop. 197, 105064. 10.1016/j.actatropica.2019.105064. [DOI] [PubMed] [Google Scholar]
- Lin J, Chen RWS, Hazan A, Weiss M, 2018. Chikungunya virus infection manifesting as intermediate uveitis. Ocul. Immunol. Inflamm 26, 680–682. 10.1080/09273948.2016.1263340. [DOI] [PubMed] [Google Scholar]
- Lindenberg R, Haymaker W, 1982. Tissue reactions in the gray matter of the central nervous system. In: Haymaker W, Adams RD (Eds.), Histology and Histopathology of the Nervous System. Charles C Thomas, Springfield, IL, pp. 973–1275. [Google Scholar]
- Llagonne-Barets M, Icard V, Leparc-Goffart I, Prat C, Perpoint T, André P, Ramière C, 2016. A case of Mayaro virus infection imported from French Guiana. J. Clin. Virol 77, 66–68. 10.1016/j.jcv.2016.02.013. [DOI] [PubMed] [Google Scholar]
- Logue CH, Bosio CF, Weite T, Keene KM, Ledermann JP, Phillips A, Sheahan BJ, Pierro DJ, Marienee N, Brault AC, Bosio CM, Singh AJ, Powers AM, Olson KE, 2009. Virulence variation among isolates of western equine encephalitis virus in an outbred mouse model. J. Gen. Virol 90, 1848–1858. 10.1099/vir.0.008656-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Logue CH, Phillips AT, Mossel EG, Ledermann JP, Welte T, Dow SW, Olson KE, Powers AM, 2010. Treatment with cationic liposome-DNA complexes (CLDCs) protects mice from lethal Western equine encephalitis virus (WEEV) challenge. Antiviral Res. 87, 195–203. 10.1016/j.antiviral.2010.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lohachanakul J, Phuklia W, Thannagith M, Thonsakulprasert T, Ubol S, 2012. High concentrations of circulating interleukin-6 and monocyte chemotactic protein-1 with low concentrations of interleukin-8 were associated with severe chikungunya fever during the 2009–2010 outbreak in Thailand. Microbiol. Immunol 56, 134–138. 10.1111/j.1348-0421.2011.00417.x. [DOI] [PubMed] [Google Scholar]
- Long KM, Ferris MT, Whitmore AC, Montgomery SA, Thurlow LR, McGee CE, Rodriguez CA, Lim JK, Heise MT, 2015. γδ T cells play a protective role in chikungunya virus-induced disease. J. Virol 90, 433–443. 10.1128/JVI.02159-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lubelczyk C, Mutebi JP, Robinson S, Elias SP, Smith LB, Juris SA, Foss K, Lichtenwalner A, Shively KJ, Hoenig DE, Webber L, Sears S, Smith RP, 2013. An epizootic of eastern equine encephalitis virus, maine, USA in 2009: outbreak description and entomological studies. Am. J. Trop. Med. Hyg 88, 95–102. 10.4269/ajtmh.2012.11-0358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luciani K, Abadía I, Martinez-Torres AO, Cisneros J, Guerra I, Garcia M, Estripeaut D, Carrera JP, 2015. Madariaga virus infection associated with a case of acute disseminated encephalomyelitis. Am. J. Trop. Med. Hyg 92, 1130–1132. 10.4269/ajtmh.14-0845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lukaszewski RA, Brooks TJ, 2000. Pegylated alpha interferon is an effective treatment for virulent Venezuelan equine encephalitis virus and has profound effects on the host immune response to infection. J. Virol 74, 5006–5015. 10.1128/jvi.74.11.5006-5015.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lum F-M, Teo T-H, Lee WWL, Kam Y-W, Renia L, Ng LFP, 2013. An essential role of antibodies in the control of Chikungunya virus infection. J. Immunol 190, 6295–6302. 10.4049/jimmunol.1300304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lundström JO, Pfeffer M, 2010. Phylogeographic structure and evolutionary history of Sindbis virus. Vector Borne Zoonotic Dis. 10, 889–907. 10.1089/vbz.2009.0069. [DOI] [PubMed] [Google Scholar]
- MacDonald GH, Johnston RE, 2000. Role of dendritic cell targeting in Venezuelan equine encephalitis virus pathogenesis. J. Virol 74, 914–922. 10.1128/jvi.74.2.914-922.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MacGregor A, Oilier W, Thomson W, Jawaheer D, Silman A, 1995. HLA-DRB1*0401/0404 genotype and rheumatoid arthritis: increased association in men, young age at onset, and disease severity. J. Rheumatol 22, 1032–1036. [PubMed] [Google Scholar]
- Mahendradas P, Ranganna SK, Shetty R, Balu R, Narayana KM, Babu RB, Shetty BK, 2008. Ocular manifestations associated with chikungunya. Ophthalmology 115, 287–291. 10.1016/j.ophtha.2007.03.085. [DOI] [PubMed] [Google Scholar]
- Mahendradas P, Shetty R, Malathi J, Madhavan HN, 2010. Chikungunya virus iridocyclitis in Fuchs’ heterochromic iridocyclitis. Indian J. Ophthalmol 58, 545–547. 10.4103/0301-4738.71707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahesh G, Giridhar A, Shedbele A, Kumar R, Saikumar SJ, 2009. A case of bilateral presumed chikungunya neuroretinitis. Indian J. Ophthalmol 57, 148–150. 10.4103/0301-4738.45508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maity P, Roy P, Basu A, Das B, Ghosh US, 2014. A case of ADEM following Chikungunya fever. J. Assoc. Physicians India 62, 441–442. [PubMed] [Google Scholar]
- Mallilankaraman K, Shedlock DJ, Bao H, Kawalekar OU, Fagone P, Ramanathan AA, Ferraro B, Stabenow J, Vijayachari P, Sundaram SG, Muruganandam N, Sarangan G, Srikanth P, Khan AS, Lewis MG, Kim JJ, Sardesai NY, Muthumani K, Weiner DB, 2011. A DNA vaccine against chikungunya virus is protective in mice and induces neutralizing antibodies in mice and nonhuman primates. PLoS Negl. Trop. Dis 5, e928. 10.1371/journal.pntd.0000928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malta JMAS, Vargas A, Leite PLE, Percio J, Coelho GE, Ferraro AHA, Cordeiro T.M.d.O., Dias J.d.S., Saad E, 2017. Guillain-Barré syndrome and other neurological manifestations possibly related to Zika virus infection in municipalities from Bahia, Brazil, 2015. Epidemiol. Serv. Saude 26, 9–18. 10.5123/S1679-49742017000100002. [DOI] [PubMed] [Google Scholar]
- Malvy D, Ezzedine K, Mamani-Matsuda M, Autran B, Tolou H, Receveur M-C, Pistone T, Rambert J, Moynet D, Mossalayi D, 2009. Destructive arthritis in a patient with chikungunya virus infection with persistent specific IgM antibodies. BMC Infect. Dis 9, 200. 10.1186/1471-2334-9-200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mancera-Páez O, Román GC, Pardo-Turriago R, Rodríguez Y, Anaya J-M, 2018. Concurrent Guillain-Barré syndrome, transverse myelitis and encephalitis post-Zika: A case report and review of the pathogenic role of multiple arboviral immunity. J. Neurol. Sci 395, 47–53. 10.1016/j.jns.2018.09.028. [DOI] [PubMed] [Google Scholar]
- Manivannan S, Baxter VK, Schultz KLW, Slusher BS, Griffin DE, 2016. Protective effects of glutamine antagonist 6-Diazo-5-Oxo-l-norleucine in mice with alphavirus encephalomyelitis. J. Virol 90, 9251–9262. 10.1128/JVI.01045-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martin NM, Griffin DE, 2018. Interleukin-10 modulation of virus clearance and disease in mice with alphaviral encephalomyelitis. J. Virol 92, 1002. 10.1128/JVI.01517-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mathiot CC, Grimaud G, Garry P, Bouquety JC, Mada A, Daguisy AM, Georges AJ, 1990. An outbreak of human Semliki Forest virus infections in Central African Republic. Am. J. Trop. Med. Hyg 42, 386–393. 10.4269/ajtmh.1990.42.386. [DOI] [PubMed] [Google Scholar]
- McKimmie CS, Johnson N, Fooks AR, Fazakerley JK, 2005. Viruses selectively upregulate Toll-like receptors in the central nervous system. Biochem. Biophys. Res. Commun 336, 925–933. 10.1016/j.bbrc.2005.08.209. [DOI] [PubMed] [Google Scholar]
- Mehta R, Soares CN, Medialdea-Carrera R, Ellul M, da Silva MTT, Rosala-Hallas A, Jardim MR, Burnside G, Pamplona L, Bhojak M, Manohar R, da Silva GAM, Adriano MV, Brasil P, Nogueira RMR, Santos DCC, Turtle L, de Sequeira PC, Brown DW, Griffiths MJ, de Filippis AMB, Solomon T, 2018. The spectrum of neurological disease associated with Zika and chikungunya viruses in adults in Rio de Janeiro, Brazil: A case series. PLoS Negl. Trop. Dis 12, e0006212. 10.1371/journal.pntd.0006212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mercado M, Acosta-Reyes J, Parra E, Guzmán L, Beltrán M, Gasque P, Mejía-García C, Viasus D, 2018. Renal involvement in fatal cases of chikungunya virus infection. J. Clin. Virol 103, 16–18. 10.1016/j.jcv.2018.03.009. [DOI] [PubMed] [Google Scholar]
- Mercado M, Acosta-Reyes J, Parra E, Pardo L, Rico A, Campo A, Navarro E, Viasus D, 2016. Clinical and histopathological features of fatal cases with dengue and chikungunya virus co-infection in Colombia, 2014 to 2015. Euro Surveill. 21 10.2807/1560-7917.ES.2016.21.22.30244. [DOI] [PubMed] [Google Scholar]
- Merle H, Donnio A, Jean-Charles A, Guyomarch J, Hage R, Najioullah F, Césaire R, Cabié A, 2018. Ocular manifestations of emerging arboviruses: dengue fever, Chikungunya, Zika virus, West Nile virus, and yellow fever. J. Fr. Ophtalmol 41, e235–e243. 10.1016/j.jfo.2018.05.002. [DOI] [PubMed] [Google Scholar]
- Metcalf TU, Griffin DE, 2011. Alphavirus-induced encephalomyelitis: antibody-secreting cells and viral clearance from the nervous system. J. Virol 85, 11490–11501. 10.1128/JVI.05379-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miner JJ, Aw Yeang HX, Fox JM, Taffner S, Maikova ON, Oh ST, Kim AHJ, Diamond MS, Lenschow DJ, Yokoyama WM, 2015. Chikungunya viral arthritis in the United States: a mimic of seronegative rheumatoid arthritis. Arthritis Rheumatol. 67, 1214–1220. 10.1002/art.39027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miner JJ, Cook LE, Hong JP, Smith AM, Richner JM, Shimak RM, Young AR, Monte K, Poddar S, Crowe JE, Lenschow DJ, Diamond MS, 2017. Therapy with CTLA4-Ig and an antiviral monoclonal antibody controls chikungunya virus arthritis. Sci. Transl. Med 9. 10.1126/scitranslmed.aah3438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mittal A, Mittal S, Bharati MJ, Ramakrishnan R, Saravanan S, Sathe PS, 2007. Optic neuritis associated with chikungunya virus infection in South India. Arch. Ophthalmol 125, 1381–1386. 10.1001/archopht.125.10.1381. [DOI] [PubMed] [Google Scholar]
- Moench TR, Griffin DE, 1984. Immunocytochemical identification and quantitation of the mononuclear cells in the cerebrospinal fluid, meninges, and brain during acute viral meningoencephalitis. J. Exp. Med 159, 77–88. 10.1084/jem.159.l.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mogami R, Pereira Vaz JL, de Fatima Barcelos Chagas Y, de Abreu MM, Torezani RS, de Almeida Vieira A, Junqueira Filho EA, Barbosa YB, Carvalho ACP, Lopes AJ, 2017. Ultrasonography of hands and wrists in the diagnosis of complications of chikungunya fever. J. Ultrasound Med 37, 511–520. 10.1002/jum.14344. [DOI] [PubMed] [Google Scholar]
- Mokhtarian F, Huan C-M, Roman C, Raine CS, 2003. Semliki Forest virus-induced demyelination and remyelination—involvement of B cells and anti-myelin antibodies. J. Neuroimmunol 137, 19–31. 10.1016/s0165-5728(03)00039-0. [DOI] [PubMed] [Google Scholar]
- Mokhtarian F, Safavi F, Sarafraz-Yazdi E, 2012. Immunization with a peptide of Semliki Forest virus promotes remyelination in experimental autoimmune encephalomyelitis. Brain Res. 1488, 92–103. 10.1016/j.brainres.2012.09.038. [DOI] [PubMed] [Google Scholar]
- Mokhtarian F, Zhang Z, Shi Y, Gonzales E, Sobel RA, 1999. Molecular mimicry between a viral peptide and a myelin oligodendrocyte glycoprotein peptide induces autoimmune demyelinating disease in mice. J. Neuroimmunol 95, 43–54. 10.1016/s0165-5728(98)00254-9. [DOI] [PubMed] [Google Scholar]
- Molaei G, Armstrong PM, Graham AC, Kramer LD, Andreadis TG, 2015. Insights into the recent emergence and expansion of eastern equine encephalitis virus in a new focus in the Northern New England USA. Parasit. Vectors 8, 516. 10.1186/s13071-015-1145-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Monlux WS, Luedke AJ, 1973. Brain and spinal cord lesions in horses inoculated with Venezuelan equine encephalomyelitis virus (epidemic American and Trinidad strains). Am. J. Vet. Res 34, 465–473. [PubMed] [Google Scholar]
- Morris MM, Dyson H, Baker D, Harbige LS, Fazakerley JK, Amor S, 1997. Characterization of the cellular and cytokine response in the central nervous system following Semliki Forest virus infection. J. Neuroimmunol 74, 185–197. 10.1016/s0165-5728(96)00786-2. [DOI] [PubMed] [Google Scholar]
- Morrison TE, Fraser RJ, Smith PN, Mahalingam S, Heise MT, 2007. Complement contributes to inflammatory tissue destruction in a mouse model of Ross River virus-induced disease. J. Virol 81, 5132–5143. 10.1128/JVI.02799-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morrison TE, Oko L, Montgomery SA, Whitmore AC, Lotstein AR, Gunn BM, Elmore SA, Heise MT, 2011. A mouse model of chikungunya virus-induced musculoskeletal inflammatory disease: evidence of arthritis, tenosynovitis, myositis, and persistence. Am. J. Pathol 178, 32–40. 10.1016/j.ajpath.2010.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morrison TE, Simmons JD, Heise MT, 2008. Complement receptor 3 promotes severe ross river virus-induced disease. J. Virol 82, 11263–11272. 10.1128/JVI.01352-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mulder DW, Parrott M, Thaler M, 1951. Sequelae of western equine encephalitis. Neurology 1, 318–327. 10.1212/wnl.l.7-8.318. [DOI] [PubMed] [Google Scholar]
- Murillo-Zamora E, Mendoza-Cano O, Trujillo-Hemández B, Guzmán-Esquivel J, Higareda-Almaraz E, Higareda-Almaraz MA, Sánchez-Piña RA, Lugo-Radillo A, 2018. Persistent arthralgia and related risks factors: a cohort study at 12 months from laboratory-confirmed chikungunya infection. Arch. Med. Res 49, 65–73. 10.1016/j.arcmed.2018.04.008. [DOI] [PubMed] [Google Scholar]
- Murthy KR, Venkataraman N, Satish V, Babu K, 2008. Bilateral retinitis following chikungunya fever. Indian J. Ophthalmol 56, 329–331. [PMC free article] [PubMed] [Google Scholar]
- Musthafa AKM, Abdurahiman P, Jose J, 2008. Case of ADEM following Chikungunya fever. J. Assoc. Physicians India 56, 473. [PubMed] [Google Scholar]
- Mylonas AD, Brown AM, Carthew TL, McGrath B, Purdie DM, Pandeya N, Vecchio PC, Collins LG, Gardner ID, de Looze FJ, Reymond EJ, Suhrbier A, 2002. Natural history of Ross River virus-induced epidemic polyarthritis. Med. J. Aust 177, 356–360. [DOI] [PubMed] [Google Scholar]
- Nair AG, Biswas J, Bhende MP, 2012. A case of bilateral Chikungunya neuroretinitis. J. Ophthalmic Inflamm. Infect 2, 39–40. 10.1007/s12348-011-0038-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nargi-Aizenman JL, Griffin DE, 2001. Sindbis virus-induced neuronal death is both necrotic and apoptotic and is ameliorated by N-methyl-D-aspartate receptor antagonists. J. Virol 75, 7114–7121. 10.1128/JVI.75.15.7114-7121.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nargi-Aizenman JL, Havert MB, Zhang M, Irani DN, Rothstein JD, Griffin DE, 2004. Glutamate receptor antagonists protect from virus-induced neural degeneration. Ann. Neurol 55, 541–549. 10.1002/ana.20033. [DOI] [PubMed] [Google Scholar]
- Nathanson N, Stolley PD, Boolukos PJ, 1969. Eastern equine encephalitis. Distribution of central nervous system lesions in man and Rhesus monkey. J. Comp. Pathol 79, 109–115. 10.1016/0021-9975(69)90034-6. [DOI] [PubMed] [Google Scholar]
- Neighbours LM, Long K, Whitmore AC, Heise MT, 2012. Myd88-dependent toll-like receptor 7 signaling mediates protection from severe Ross River virus-induced disease in mice. J. Virol 86, 10675–10685. 10.1128/JVI.00601-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neri V, Bacelar O, Farinhas JG, Franga LC, Dantas FF, Leon SA, 2018. Optic neuritis and acute transverse myelitis after chikungunya virus infection. Int. J. Infect. Dis 73, S179. 10.1016/j.ijid.2018.04.3820. [DOI] [Google Scholar]
- Ng LFP, Chow A, Sun Y-J, Kwek DJC, Lim P-L, Dimatatac F, Ng L-C, Ooi E-E, Choo K-H, Her Z, Kourilsky P, Leo Y-S, 2009. IL-1beta, IL-6, and RANTES as biomarkers of Chikungunya severity. PLoS One 4, e4261. 10.1371/journal.pone.0004261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nhan T-X, Claverie A, Roche C, Teissier A, Colleuil M, Baudet J-M, Cao-Lormeau V-M, Musso D, 2014. Chikungunya virus imported into French Polynesia, 2014. Emerg. Infect. Dis 20, 1773–1774. 10.3201/eid2010.141060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nickerson JP, Kannabiran S, Burbank HN, 2016. MRI findings in eastern equine encephalitis: the “parenthesis” sign. Clin. Imaging 40, 222–223. 10.1016/j.clinimag.2015.10.012. [DOI] [PubMed] [Google Scholar]
- Nicotera P, Orrenius S, 1998. The role of calcium in apoptosis. Cell Calcium 23,173–180. [DOI] [PubMed] [Google Scholar]
- Nilaratanakul V, Chen J, Tran O, Baxter VK, Traisi EM, Yeh JX, Griffin DE, 2018. Germline IgM is sufficient, but not required, for antibody-mediated alphavirus clearance from the central nervous system. J. Virol 92, e02081–17. 10.1128/JVI.02081-17. JVI.02081-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ninla-aesong P, Mitamun W, Noipha K, 2019. Proinflammatory cytokines and chemokines as biomarkers of persistent arthralgia and severe disease after chikungunya virus infection: a 5-year follow-up study in Southern Thailand. Viral Immunol. 32, 442–452. 10.1089/vim.2019.0064. [DOI] [PubMed] [Google Scholar]
- Ninla-aesong P, Mitamun W, Noipha K, 2020. Long-term persistence of chikungunya virus-associated manifestations and anti-chikungunya virus antibody in southern Thailand: 5 years after an outbreak in 2008–2009. Viral Immunol. 33, 86–93. 10.1089/vim.2019.0168. [DOI] [PubMed] [Google Scholar]
- Oehler E, Fournier E, Leparc-Goffart I, Larre P, Cubizolle S, Sookhareea C, Lastère S, Ghawche F, 2015. Increase in cases of Guillain-Barré syndrome during a Chikungunya outbreak, French Polynesia, 2014 to 2015. Euro Surveill. 20, 30079. 10.2807/1560-7917.ES.2015.20.48.30079. [DOI] [PubMed] [Google Scholar]
- Oliver KR, Fazakerley JK, 1998. Transneuronal spread of Semliki Forest virus in the developing mouse olfactory system is determined by neuronal maturity. Neuroscience 82, 867–877. 10.1016/s0306-4522(97)00309-6. [DOI] [PubMed] [Google Scholar]
- Oliver KR, Scallan MF, Dyson H, Fazakerley JK, 1997. Susceptibility to a neurotropic virus and its changing distribution in the developing brain is a function of CNS maturity. J. Neurovirol 3, 38–48. 10.3109/13550289709015791. [DOI] [PubMed] [Google Scholar]
- Ozden S, Huerre M, Riviere JP, Coffey LL, Afonso PV, Mouly V, de Monredon J, Roger J-C, El Amrani M, Yvin J-L, Jaffar M-C, Frenkiel M-P, Sourisseau M, Schwartz O, Gulter-Browne G, Despres P, Gessain A, Ceccaldi P-E, 2007. Human muscle satellite cells as targets of chikungunya virus infection. PLoS One 2, e527. 10.1371/journal.pone.0000527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paessler S, Aguilar P, Anishchenko M, Wang H-Q, Aronson J, Campbell G, Cararra A-S, Weaver SC, 2004. The hamster as an animal model for eastern equine encephalitis—and its use in studies of virus entrance into the brain. J. Infect. Dis 189, 2072–2076. 10.1086/383246. [DOI] [PubMed] [Google Scholar]
- Palmer RJ, Finley KH, 1956. Sequelae of encephalitis; report of a study after the California epidemic. Calif. Med 84, 98–100. [PMC free article] [PubMed] [Google Scholar]
- Panato CS, Figueredo ED, Bassi D, Felipe IMA, Firmo WDCA, Rêgo AS, Silva F.d.M.A.M., 2019. Evaluation of functional disability after Chikungunya infection. Rev. Soc. Bras. Med. Trop 52, e20190112. 10.1590/0037-8682-0112-2019. [DOI] [PubMed] [Google Scholar]
- Partidos CD, Weger J, Brewoo J, Seymour R, Borland EM, Ledermann JP, Powers AM, Weaver SC, Stinchcomb DT, Osorio JE, 2011. Probing the attenuation and protective efficacy of a candidate chikungunya virus vaccine in mice with compromised interferon (IFN) signaling. Vaccine 29, 3067–3073. 10.1016/j.vaccine.2011.01.076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Passoni G, Langevin C, Palha N, Mounce BC, Briolat V, Affaticati P, De Job E, Joly J-S, Vignuzzi M, Saleh M-C, Herbomel P, Boudinot P, Levraud J-P, 2017. Imaging of viral neuroinvasion in the zebrafish reveals that Sindbis and chikungunya viruses favour different entry routes. Dis. Model. Mech 10, 847–857. 10.1242/dmm.029231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patel DM, Patel MV, Sharma KH, Patel GR, Patel MB, Shah VV, 2019. Post chikungunya chronic arthritis: systemic inflammatory status triggering acute coronary syndrome. Curr. Rheumatol. Rev 15, 229–233. 10.2174/1573397114666180816112948. [DOI] [PubMed] [Google Scholar]
- Pathak S, Illavia SJ, Webb HE, 1983. The identification and role of cells involved in CNS demyelination in mice after Semliki Forest virus infection: an ultrastructural study. Prog. Brain Res 59, 237–254. 10.1016/S0079-6123(08)63869-8. [DOI] [PubMed] [Google Scholar]
- Pathak S, Webb HE, 1974. Possible mechanisms for the transport of Semliki forest virus into and within mouse brain. An electron-microscopic study. J. Neurol. Sci 23, 175–184. 10.1016/0022-510x(74)90221-4. [DOI] [PubMed] [Google Scholar]
- Paul B, Pariyapurath R, 2018. Risk factor assessment of rheumatoid arthritis in North Kerala. Eur.J. Rheumatol 5, 184–190. 10.5152/eurjrheum.2018.17111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phan TG, Somerville E, Hueston L, Dwyer DE, 1998. Probable association between Barmah Forest virus infection and Guillain-Barré syndrome. Med. J. Aust 168, 422–423. [PubMed] [Google Scholar]
- Phelps AL, O’Brien LM, Eastaugh LS, Davies C, Lever MS, Ennis J, Zeitlin L, Nunez A, Ulaeto DO, 2017. Susceptibility and lethality of Western equine encephalitis virus in Balb/c mice when infected by the aerosol route. Viruses 9, 163. 10.3390/v9070163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phelps AL, O’Brien LM, Eastaugh LS, Davies C, Lever MS, Ennis J, Zeitlin L, Nunez A, Ulaeto DO, 2019. Aerosol infection of Balb/c mice with eastern equine encephalitis virus; susceptibility and lethality. Virol. J 16, 2. 10.1186/s12985-018-1103-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phillips AT, Rico AB, Stauft CB, Hammond SL, Aboellail TA, Tjalkens RB, Olson KE, 2016. Entry Sites of Venezuelan and Western Equine Encephalitis Viruses in the mouse central nervous system following peripheral infection. J. Virol 90, 5785–5796. 10.1128/JVI.03219-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phillips AT, Stauft CB, Aboellail TA, Toth AM, Jarvis DL, Powers AM, Olson KE, 2013. Bioluminescent imaging and histopathologic characterization of WEEV neuroinvasion in outbred CD-1 mice. PLoS One 8, e53462. 10.1371/journal.pone.0053462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pinheiro TJ, Guimarães LF, Silva MTT, Soares CN, 2016. Neurological manifestations of Chikungunya and Zika infections. Arq. Neuropsiquiatr 74, 937–943. 10.1590/0004-282X20160138. [DOI] [PubMed] [Google Scholar]
- Poo YS, Nakaya H, Gardner J, Larcher T, Schroder WA, Le TT, Major LD, Suhrbier A, 2014. CCR2 deficiency promotes exacerbated chronic erosive neutrophil-dominated chikungunya virus arthritis. J. Virol 88, 6862–6872. 10.1128/JVI.03364-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poon AN, Simon GL, Chang AY, 2019. Treatment of chronic chikungunya with methotrexate. J. Clin. Rheumatol 00, 1–2. 10.1097/RHU.0000000000000998. [DOI] [PubMed] [Google Scholar]
- Porter AI, Erwin-Cohen RA, Twenhafel N, Chance T, Yee SB, Kern SJ, Norwood D, Hartman LJ, Parker MD, Glass PJ, DaSilva L, 2017. Characterization and pathogenesis of aerosolized eastern equine encephalitis in the common marmoset (Callithrix jacchus). Virol. J 14, 25. 10.1186/s12985-017-0687-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Potter MC, Baxter VK, Mathey RW, Alt J, Rojas C, Griffin DE, Slusher BS, 2015. Neurological sequelae induced by alphavirus infection of the CNS are attenuated by treatment with the glutamine antagonist 6-diazo-5-oxo-l-norleucine. J. Neurovirol 21, 159–173. 10.1007/s13365-015-0314-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Potter ME, Currier RW, Pearson JE, Harris JC, Parker RL, 1977. Western equine encephalomyelitis in horses in the Northern Red River Valley, 1975. J. Am. Vet. Med. Assoc 170, 1396–1399. [PubMed] [Google Scholar]
- Powers AM, 2018. Vaccine and therapeutic options to control chikungunya virus. Clin. Microbiol. Rev 31, e00104–16. 10.1128/CMR.00104-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prow NA, Irani DN, 2007. The opioid receptor antagonist, naloxone, protects spinal motor neurons in a murine model of alphavirus encephalomyelitis. Exp. Neurol 205, 461–470. 10.1016/j.expneurol.2007.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Przelomski MM, O’Rourke E, Grady GF, Berardi VP, Markley HG, 1988. Eastern equine encephalitis in Massachusetts: a report of 16 cases, 1970–1984. Neurology 38, 736–739. 10.1212/wnl.38.5-736. [DOI] [PubMed] [Google Scholar]
- Reddy V, Mani RS, Desai A, Ravi V, 2014. Correlation of plasma viral loads and presence of Chikungunya IgM antibodies with cytokine/chemokine levels during acute Chikungunya virus infection. J. Med. Virol 86, 1393–1401. 10.1002/jmv.23875. [DOI] [PubMed] [Google Scholar]
- Reed DS, Lackemeyer MG, Garza NL, Norris S, Gamble S, Sullivan LJ, Lind CM, Raymond JL, 2007. Severe encephalitis in cynomolgus macaques exposed to aerosolized Eastern equine encephalitis virus. J. Infect.Dis 196, 441–450. 10.1086/519391. [DOI] [PubMed] [Google Scholar]
- Reed DS, Larsen T, Sullivan LJ, Lind CM, Lackemeyer MG, Pratt WD, Parker MD, 2005. Aerosol exposure to western equine encephalitis virus causes fever and encephalitis in cynomolgus macaques. J. Infect. Dis 192, 1173–1182. 10.1086/444397. [DOI] [PubMed] [Google Scholar]
- Reilly JM, Xing W, Levicky V, Souccar S, Rogers C, Sathyavagiswaran L, 2020. Postmortem chikungunya diagnosis: a case report and literature review. Am. J. Forensic Med. Pathol 41, 48–51. 10.1097/PAF.0000000000000519. [DOI] [PubMed] [Google Scholar]
- Rezza G, Chen R, Weaver SC, 2017. O’nyong-nyong fever: a neglected mosquito-bome viral disease. Pathog. Glob. Health 111, 271–275. 10.1080/20477724.2017.1355431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rivas F, Diaz LA, Cardenas VM, Daza E, Bruzon L, Alcala A, Hoz l., De O, Caceres FM, Aristizabal G, Martinez JW, Revelo D, Hoz l., De F, Boshell J, Camacho T, Calderon L, Olano VA, Villarreal LI, Roselli D, Alvarez G, Ludwig G, Tsai T, 1997. Epidemie Venezuelan equine encephalitis in La Guajira, Colombia, 1995. J. Infect. Dis 175, 828–832. [DOI] [PubMed] [Google Scholar]
- Riyaz N, Riyaz A, Rahima A, Latheef EN, Anitha PM, Aravindan KP, Nair AS, Shameera P, 2010. Cutaneous manifestations of chikungunya during a recent epidemic in Calicut, north Kerala, south India. Indian J. Dermatol. Venereol. Leprol 76, 671–676. 10.4103/0378-6323.72466. [DOI] [PubMed] [Google Scholar]
- Roberts ED, Sanmartin C, Payán J, Mackenzie RB, 1970. Neuropathologie changes in 15 horses with naturally occurring Venezuelan equine encephalomyelitis. Am. J. Vet. Res 31, 1223–1229. [PubMed] [Google Scholar]
- Robinson MC, 1955. An epidemic of virus disease in Southern Province, Tanganyika territory, in 1952–1953. Trans. R. Soc. Trop. Med. Hyg 49, 28–32. [DOI] [PubMed] [Google Scholar]
- Rocha VFD, de Oliveira AHP, Bandeira AC, Sardi SI, Garcia RF, Magalhães S. d.A., Sampaio CA, Campos Soares G, 2018. Chikungunya virus infection associated with encephalitis and anterior uveitis. Ocul. Immunol. Inflamm 26, 677–679. 10.1080/09273948.2017.1358378. [DOI] [PubMed] [Google Scholar]
- Rose N, Anoop TM, John AP, Jabbar PK, George KC, 2011. Acute optic neuritis following infection with chikungunya virus in southern rural India. Int. J. Infect. Dis 15, e147–e150. 10.1016/j.ijid.2010.10.004. [DOI] [PubMed] [Google Scholar]
- Ross RW, 1956. The Newala epidemic. III. The virus: isolation, pathogenic properties and relationship to the epidemic. J. Hyg. (Lond) 54, 177–191. 10.1017/s0022172400044442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rowell JF, Griffin DE, 1999. The inflammatory response to nonfatal Sindbis virus infection of the nervous system is more severe in SJL than in BALB/c mice and is associated with low levels of IL-4 mRNA and high levels of IL-10-producing CD4+ T cells. J. Immunol 162, 1624–1632. [PubMed] [Google Scholar]
- Rowell JF, Griffin DE, 2002. Contribution of T cells to mortality in neurovirulent Sindbis virus encephalomyelitis. J. Neuroimmunol 127, 106–114. [DOI] [PubMed] [Google Scholar]
- Roy CJ, Adams AP, Wang E, Leal CL, Seymour RL, Sivasubramani SK, Mega W, Frolov I, Didier PJ, Weaver SC, 2013. A chimeric Sindbis-based vaccine protects cynomolgus macaques against a lethal aerosol challenge of eastern equine encephalitis virus. Vaccine 31, 1464–1470. 10.1016/j.vaccine.2013.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roy CJ, Reed DS, Wilhelmsen CL, Hartings J, Norris S, Steele KE, 2009. Pathogenesis of aerosolized Eastern Equine Encephalitis virus infection in guinea pigs. Virol. J 6, 170. 10.1186/1743-422X-6-170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rozdilsky B, Robertson HE, Chomey J, 1968. Western encephalitis: report of eight fatal cases. Saskatchewan epidemic, 1965. Can. Med. Assoc. J 98, 79–86. [PMC free article] [PubMed] [Google Scholar]
- Rudd PA, Wilson J, Gardner J, Larcher T, Babarit C, Le TT, Anraku I, Kumagai Y, Loo Y-M, Gale M, Akira S, Khromykh AA, Suhrbier A, 2012. Interferon response factors 3 and 7 protect against Chikungunya virus hemorrhagic fever and shock. J. Virol 86, 9888–9898. 10.1128/JVI.00956-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rulli NE, Guglielmotti A, Mangano G, Rolph MS, Apicella C, Zaid A, Suhrbier A, Mahalingam S, 2009. Amelioration of alphavirus-induced arthritis and myositis in a mouse model by treatment with bindarit, an inhibitor of monocyte chemotactic proteins. Arthritis Rheum. 60, 2513–2523. 10.1002/art.24682. [DOI] [PubMed] [Google Scholar]
- Ryman KD, Klimstra WB, Nguyen KB, Biron CA, 2000. Alpha/beta interferon protects adult mice from fatal Sindbis virus infection and is an important determinant of cell and tissue tropism. J. Virol 74, 3366–3378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ryzhikov AB, Tkacheva NV, Sergeev AN, Ryabchikova EI, 1991. Venezuelan equine encephalitis virus propagation in the olfactory tract of normal and immunized mice. Biomed. Sci 2, 607–614. [PubMed] [Google Scholar]
- Safavi F, Feliberti JP, Raine CS, Mokhtarian F, 2011. Role of γδ T cells in antibody production and recovery from SFV demyelinating disease. J. Neuroimmunol 235, 18–26. 10.1016/j.jneuroim.2011.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sahu R, Verma R, Jain A, Garg RK, Singh MK, Malhotra HS, Sharma PK, Parihar A, 2014. Neurologic complications in dengue virus infection: a prospective cohort study. Neurology 83, 1601–1609. 10.1212/WNL.0000000000000935. [DOI] [PubMed] [Google Scholar]
- Sahu SP, Pedersen DD, Jenny AL, Schmitt BJ, Alstad AD, 2003. Pathogenicity of a Venezuelan equine encephalomyelitis serotype IE virus isolate for ponies. Am. J. Trop. Med. Hyg 68, 485–494. 10.4269/ajtmh.2003.68.485. [DOI] [PubMed] [Google Scholar]
- Salceanu SO, Raman V, 2018. Recurrent chikungunya retinitis. BMJ Case Rep. 2018, 1–4. 10.1136/bcr-2017-222864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sane J, Kurkela S, Lokki M-L, Miettinen A, Helve T, Vaheri A, Vapalahti O, 2012. Clinical Sindbis alphavirus infection is associated with HLA-DRB1*01 allele and production of autoantibodies. Clin. Infect. Dis 55, 358–363. 10.1093/cid/cis405. [DOI] [PubMed] [Google Scholar]
- Santiago FW, Halsey ES, Siles C, Vilcarromero S, Guevara C, Silvas JA, Ramal C, Ampuero JS, Aguilar PV, 2015. Long-term arthralgia after mayaro virus infection correlates with sustained pro-inflammatory cytokine response. PLoS Negl. Trop. Dis 9, e0004104. 10.1371/joumal.pntd.0004104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sarid R, Ben-Moshe T, Kazimirsky G, Weisberg S, Appel E, Kobiler D, Lustig S, Brodie C, 2001. vFLIP protects PC-12 cells from apoptosis induced by Sindbis virus: implications for the role of TNF-α. Cell Death Differ. 8, 1224–1231. 10.1038/sj.cdd.4400926. [DOI] [PubMed] [Google Scholar]
- Sattler R, Tymianski M, 2001. Molecular mechanisms of glutamate receptor-mediated excitotoxic neuronal cell death. Mol. Neurobiol 24, 107–129. 10.1385/MN:24:1-3:107. [DOI] [PubMed] [Google Scholar]
- Sánchez JS, Cañón AM, Lombo JC, 2019. Subacute and chronic symptoms of chikungunya fever in a group of adults in Colombia. Biomedica 39, 587–594. 10.7705/biomedica.4350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schilte C, Couderc T, Chrétien F, Sourisseau M, Gangneux N, Guivel-Benhassine F, Kraxner A, Tschopp J, Higgs S, Michault A, Arenzana-Seisdedos F, Colonna M, Peduto L, Schwartz O, Lecuit M, Albert ML, 2010. Type I IFN controls chikungunya virus via its action on nonhematopoietic cells. J. Exp. Med 207, 429–442. 10.1084/jem.20090851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schoneboom BA, Catlin KM, Marty AM, Grieder FB, 2000a. Inflammation is a component of neurodegeneration in response to Venezuelan equine encephalitis vims infection in mice. J. Neuroimmunol 109, 132–146. 10.1016/s0165-5728(00)00290-3. [DOI] [PubMed] [Google Scholar]
- Schoneboom BA, Fultz MJ, Miller TH, McKinney LC, Grieder FB, 1999. Astrocytes as targets for Venezuelan equine encephalitis virus infection. J. Neurovirol 5, 342–354. 10.3109/13550289909029475. [DOI] [PubMed] [Google Scholar]
- Schoneboom BA, Lee JS, Grieder FB, 2000b. Early expression of IFN-alpha/beta and iNOS in the brains of Venezuelan equine encephalitis vims-infected mice. J. Interferon Cytokine Res 20, 205–215. 10.1089/107999000312621. [DOI] [PubMed] [Google Scholar]
- Schultz KLW, Troisi EM, Baxter VK, Glowinski R, Griffin DE, 2019. Interferon regulatory factors 3 and 7 have distinct roles in the pathogenesis of alphavims enceph-alomyelitis. J. Gen. Virol 100, 46–62. 10.1099/jgv.0.001174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scripsema NK, Sharifi E, Samson CM, Kedhar S, Rosen RB, 2015. Chikungunya-associated uveitis and exudative retinal detachment: A case report. Retin Cases Brief Rep. 9, 352–356. 10.1097/ICB.0000000000000232. [DOI] [PubMed] [Google Scholar]
- Sepúlveda-Delgado J, Vera-Lastra OL, Tmjillo-Murillo K, Canseco-Ávila LM, Sánchez-González RA, Gómez-Cmz O, Lugo-Trampe A, Fernández-Salas I, Danis-Lozano R, Contreras-Contreras A, Mendoza-Torres A, Domínguez-Arrevillaga S, Mena-Vela BA, Ocaña-Sibilla M, Ramirez-Valdespino JC, Jara LJ, 2017. Inflammatory biomarkers, disease activity index, and self-reported disability may be predictors of chronic arthritis after chikungunya infection: brief report. Clin. Rheumatol 36, 695–699. 10.1007/s10067-016-3419-2. [DOI] [PubMed] [Google Scholar]
- Seth P, Husain MM, Gupta P, Schoneboom A, Grieder BFB, Mani H, Maheshwari RK, 2003. Early onset of virus infection and up-regulation of cytokines in mice treated with cadmium and manganese. Biometals 16, 359–368. 10.1023/a:1020682716212. [DOI] [PubMed] [Google Scholar]
- Seymour RL, Adams AP, Leal G, Alcorn MDH, Weaver SC, 2015. A rodent model of chikungunya vims infection in RAG1 −/− mice, with features of persistence, for vaccine safety evaluation. PLoS Negl. Trop. Dis 9, e0003800. 10.1371/journal.pntd.0003800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharma A, Bhattacharya B, Puri RK, Maheshwari RK, 2008. Venezuelan equine encephalitis virus infection causes modulation of inflammatory and immune response genes in mouse brain. BMC Genomics 9, 289. 10.1186/1471-2164-9-289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharma A, Maheshwari RK, 2009. Oligonucleotide array analysis of Toll-like receptors and associated signaling genes in Venezuelan equine encephalitis virus-infected mouse brain. J. Gen. Virol 90, 1836–1847. 10.1099/vir.0.010280-0. [DOI] [PubMed] [Google Scholar]
- Sheahan BJ, Gates MC, Caffrey JF, Atkins GJ, 1983. Oligodendrocyte infection and demyelination produced in mice by the M9 mutant of Semliki Forest virus. Acta Neuropathol. 60, 257–265. 10.1007/bf00691874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Silverman MA, Misasi J, Smole S, Feldman HA, Cohen AB, Santagata S, McManus M, Ahmed AA, 2013. Eastern equine encephalitis in children, Massachusetts and New Hampshire, USA, 1970–2010. Emerg. Infect. Dis 19, 194–201. 10.3201/eid1902.120039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sissoko D, Malvy D, Ezzedine K, Renault P, Moscetti F, Ledrans M, Pierre V, 2009. Post-epidemic Chikungunya disease on reunion Island: course of rheumatic manifestations and associated factors over a 15-month period. PLoS Negl. Trop. Dis 3, e389. 10.1371/journal.pntd.0000389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soilu-Hänninen M, Erälinna JP, Hukkanen V, Röyttä M, Salmi AA, Salonen R, 1994. Semliki Forest virus infects mouse brain endothelial cells and causes blood-brain barrier damage. J. Virol 68, 6291–6298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sosa-Martínez M.d.J., Orea-Flores M, Vázquez-Cruz I, Palacios-Castillo V, Juanico-Morales G, Pérez-Mijangos L, 2018. Characterization of chronic clinical manifestations in patients with chikungunya fever. Rev. Med. Inst. Mex. Seguro Soc 56, 239–245. [PubMed] [Google Scholar]
- Steele KE, Davis KJ, Stephan K, Kell W, Vogel P, Hart MK, 1998. Comparative neurovirulence and tissue tropism of wild-type and attenuated strains of Venezuelan equine encephalitis virus administered by aerosol in C3H/HeN and BALB/c mice. Vet. Pathol 35, 386–397. 10.1177/030098589803500508. [DOI] [PubMed] [Google Scholar]
- Steele KE, Reed DS, Glass PJ, Hart MK, Ludwig GV, Pratt WD, Parker MD, Smith JF, 2007. Alphavirus encephalitides. In: Dembek ZF (Ed.), Medical Aspects of Biological Warfare. Government Printing Office, Washington, DC, pp. 241–270. [Google Scholar]
- Steele KE, Seth P, Catlin-Lebaron KMK, Schoneboom BA, Husain MM, Grieder F, Maheshwari RK, 2006. Tunicamycin enhances neuroinvasion and encephalitis in mice infected with Venezuelan equine encephalitis virus. Vet. Pathol 43, 904–913. 10.1354/vp.43-6-904. [DOI] [PubMed] [Google Scholar]
- Steele KE, Twenhafel NA, 2010. Pathology of animal models of alphavirus encephalitis. Vet. Pathol 47, 790–805. 10.1177/0300985810372508. [DOI] [PubMed] [Google Scholar]
- Stegmann-Planchard S, Gallian P, Tressières B, Leparc-Goffart I, Lannuzel A, Signate A, Laouénan C, Cabie A, Hoen B, 2019. Chikungunya, a risk factor for Guillain-Barré syndrome. Clin. Infect. Dis 70, 1233–1235. 10.1093/cid/ciz625. ciz625 [DOI] [PubMed] [Google Scholar]
- Stohlman SA, Bergmann CC, Lin MT, Cua DJ, Hinton DR, 1998. CTL effector function within the central nervous system requires CD4+ T cells. J. Immunol 160, 2896–2904. [PubMed] [Google Scholar]
- Storm N, Weyer J, Markotter W, Kemp A, Leman PA, Dermaux-Msimang V, Nel LH, Paweska JT, 2014. Human cases of Sindbis fever in South Africa, 2006–2010. Epidemiol. Infect 142, 234–238. 10.1017/S0950268813000964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Styczynski AR, Malta JMAS, Krow-Lucal ER, Percio J, Nóbrega ME, Vargas A, Lanzieri TM, Leite PL, Staples JE, Fischer MX, Powers AM, Chang G-JJ, Bums PL, Borland EM, Ledermann JP, Mossel EC, Schonberger LB, Belay EB, Salinas JL, Badaro RD, Sejvar JJ, Coelho GE, 2017. Increased rates of Guillain-Barré syndrome associated with Zika virus outbreak in the Salvador metropolitan area, Brazil. PLoS Negl. Trop. Dis 11, e0005869. 10.1371/journal.pntd.0005869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Subak-Sharpe I, Dyson H, Fazakerley J, 1993. In vivo depletion of CD8+ T cells prevents lesions of demyelination in Semliki Forest virus infection. J. Virol 67, 7629–7633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suhrbier A, Jaffar-Bandjee MC, Gasque P, 2012. Arthritogenic alphaviruses—an overview. Nat. Rev. Rheumatol 8, 420–429. 10.1038/nrrheum.2012.64. [DOI] [PubMed] [Google Scholar]
- Tandale BV, Sathe PS, Arankalle VA, Wadia RS, Kulkami R, Shah SV, Shah SK, Sheth JK, Sudeep AB, Tripathy AS, Mishra AC, 2009. Systemic involvements and fatalities during Chikungunya epidemic in India, 2006. J. Clin. Virol 46, 145–149. 10.1016/j.jcv.2009.06.027. [DOI] [PubMed] [Google Scholar]
- Tappe D, Pérez-Girón JV, Just-Nübling G, Schuster G, Gómez-Medina S, Günther S, Muñoz-Fontela C, Schmidt-Chanasit J, 2016. Sustained elevated cytokine levels during recovery phase of Mayaro virus infection. Emerg. Infect. Dis 22, 750–752. 10.3201/eid2204.151502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taraphdar D, Roy BK, Chatteqee S, 2015. Chikungunya virus infection amongst the acute encephalitis syndrome cases in West Bengal, India. Indian J. Med. Microbiol 33, 153–156. 10.4103/0255-0857.150946. Suppl. [DOI] [PubMed] [Google Scholar]
- Taylor RM, Hurlbut HS, Work TH, Kingston JR, Frothingham TE, 1955. Sindbis virus: a newly recognized arthropod transmitted virus. Am. J. Trop. Med. Hyg 4, 844–862. [DOI] [PubMed] [Google Scholar]
- Teixeira AAR, Ferreira LF, Pires Neto RDJ, 2019. Acute disseminated encephalomyelitis after Chikungunya infection. JAMA Neurol. 76, 619. 10.1001/jamaneurol.2019.0113. [DOI] [PubMed] [Google Scholar]
- Teng T-S, Kam Y-W, Lee B, Hapuarachchi HC, Wimal A, Ng L-C, Ng LFP, 2015. A systematic meta-analysis of immune signatures in patients with acute Chikungunya Virus infection. J. Infect. Dis 211, 1925–1935. 10.1093/infdis/jiv049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teo T-H, Lum F-M, Glaser C, Lulla V, Lulla A, Merits A, Renia L, Ng LFP, 2013. A pathogenic role for CD4+ T cells during Chikungunya virus infection in mice. J. Immunol 190, 259–269. 10.4049/jimmunol.1202177. [DOI] [PubMed] [Google Scholar]
- Thach DC, Kimura T, Griffin DE, 2000. Differences between C57BL/6 and BALB/cBy mice in mortality and virus replication after intranasal infection with neuroadapted Sindbis virus. J. Virol 74, 6156–6161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thanapati S, Ganu M, Giri P, Kulkarni S, Sharma M, Babar P, Ganu A, Tripathy AS, 2017. Impaired NK cell functionality and increased TNF-α production as biomarkers of chronic chikungunya arthritis and rheumatoid arthritis. Hum. Immunol 78, 370–374. 10.1016/j.humimm.2017.02.006. [DOI] [PubMed] [Google Scholar]
- Theilacker C, Held J, Allering L, Emmerich P, Schmidt-Chanasit J, Kern WV, Panning M, 2013. Prolonged polyarthralgia in a German traveller with Mayaro virus infection without inflammatory correlates. BMC Infect. Dis 13, 369. 10.1186/1471-2334-13-369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Touret Y, Randrianaivo H, Michault A, Schuffenecker I, Kauffinann E, Lenglet Y, Barau G, Fourmaintraux A, 2006. Early maternal-fetal transmission of the Chikungunya virus. Presse Med. 35, 1656–1658. 10.1016/S0755-4982(06)74874-6. [DOI] [PubMed] [Google Scholar]
- Toumebize P, Charlin C, Lagrange M, 2009. Neurological manifestations in Chikungunya: about 23 cases collected in Reunion Island. Rev. Neurol. (Paris) 165, 48–51. 10.1016/j.neurol.2008.06.009. [DOI] [PubMed] [Google Scholar]
- Tritsch SR, Encinales L, Pacheco N, Cadena A, Cure C, McMahon E, Watson H, Porras-Ramirez A, Mendoza AR, Li G, Khurana K, Jaller-Raad JJ, Mejia Castillo S, Barrios Taborda C, Jaller-Char JJ, Avendaño Echavez L, Jiménez D, Gonzalez Coba A, Alarcon Gomez M, Ariza Orozco D, Bravo E, Martinez V, Guerra B, Simon G, Firestein GS, Chang AY, 2019. Chronic joint pain 3 years after Chikungunya virus infection largely characterized by relapsing-remitting symptohms. J. Rheumatol 46, 1–8. 10.3899/jrheum.190162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsetsarkin KA, Vanlandingham DL, McGee CE, Higgs S, 2007. A single mutation in Chikungunya virus affects vector specificity and epidemic potential. PLoS Pathog. 3, e201–e212. 10.1371/journal.ppat.0030201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ulloa-Padilla JP, Dávila PJ, Izquierdo NJ, García-Rodríguez O, Jiménez IZ, 2018. Ocular symptoms and signs of Chikungunya fever in Puerto Rico. P. R. Health Sci. J 37, 83–87. [PubMed] [Google Scholar]
- Venigalla SSK, Premakumar S, Janakiraman V, 2020. A possible role for autoimmunity through molecular mimicry in alphavirus mediated arthritis. Sci. Rep 10, 938. 10.1038/s41598-019-55730-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Venugopalan A, Ghorpade RP, Chopra A, 2014. Cytokines in acute chikungunya. PLoS One 9, e111305. 10.1371/journal.pone.0111305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vieira MADCES, Costa CHN, Linhares ADC, Borba A.d.S., Henriques DF, Silva EVPD, Tavares FN, Batista F.M.d.A., Guimarães HCL, Martins LC, Monteiro TAF, Cruz ACR, Azevedo RDSDS, Vasconcelos PFDC, 2018. Potential role of dengue virus, chikungunya virus and Zika virus in neurological diseases. Mem. Inst. Oswaldo Cruz 113, e170538. 10.1590/0074-02760170538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vijayakumar KP, Nair Anish TS, George B, Lawrence T, Muthukkutty SC, Ramachandran R, 2011. Clinical profile of Chikungunya patients during the epidemic of 2007 in Kerala, India. J. Glob. Infect 3, 221–226. 10.4103/0974-777X.83526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Villamil-Gómez W, Silvera LA, Páez-Castellanos J, Rodriguez-Morales AJ, 2016. Guillain-Barré syndrome after Chikungunya infection: a case in Colombia. Enferm. Infecc. Microbiol. Clin 34, 140–141. 10.1016/j.eimc.2015.05.012. [DOI] [PubMed] [Google Scholar]
- Villari P, Spielman A, Komar N, McDowell M, Timperi RJ, 1995. The economic burden imposed by a residual case of eastern encephalitis. Am. J. Trop. Med. Hyg 52, 8–13. [DOI] [PubMed] [Google Scholar]
- Vogel P, Abplanalp D, Kell W, Ibrahim MS, Downs MB, Pratt WD, Davis KJ, 1996. Venezuelan equine encephalitis in BALB/c mice: kinetic analysis of central nervous system infection following aerosol or subcutaneous inoculation. Arch. Pathol. Lab. Med 120, 164–172. [PubMed] [Google Scholar]
- Vogel P, Kell WM, Fritz DL, Parker MD, Schoepp RJ, 2005. Early events in the pathogenesis of eastern equine encephalitis virus in mice. Am. J. Pathol 166, 159–171. 10.1016/S0002-9440(10)62241-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wachira VK, Peixoto HM, de Oliveira MRF, 2019. Systematic review offactors associated with the development of Guillain-Barré syndrome 2007–2017: what has changed? Trop. Med. Int. Health 24, 132–142. 10.1111/tmi.13181. [DOI] [PubMed] [Google Scholar]
- Walker DH, Harrison A, Murphy K, Flemister M, Murphy FA, 1976. Lymphoreticular and myeloid pathogenesis of Venezuelan equine encephalitis in hamsters. Am. J. Pathol 84, 351–370. [PMC free article] [PubMed] [Google Scholar]
- Walton TE, Alvarez O Jr., Buckwalter RM, Johnson KM, 1973. Experimental infection of horses with enzootic and epizootic strains of Venezuelan equine encephalomyelitis virus. J. Infect. Dis 128, 271–282. [DOI] [PubMed] [Google Scholar]
- Wang R, Dillon CP, Shi LZ, Milasta S, Carter R, Finkeistein D, McCormick LL, Fitzgerald P, Chi H, Munger J, Green DR, 2011. The transcription factor myc controls metabolic reprogramming upon T Lymphocyte activation. Immunity 35, 871–882. 10.1016/j.immuni.2011.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watson H, Tritsch SR, Encinales L, Cadena A, Cure C, Ramirez AP, Mendoza AR, Chang AY, 2020. Stiffness, pain, and joint counts in chronic chikungunya disease: relevance to disability and quality of life. Clin. Rheumatol 372, 1231–1238. 10.1007/s10067-019-04919-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wauquier N, Becquart P, Nkoghe D, Padilla C, Ndjoyi-Mbiguino A, Leroy EM, 2011. The acute phase of Chikungunya virus infection in humans is associated with strong innate immunity and T CD8 cell activation. J. Infect. Dis 204, 115–123. 10.1093/infdis/jiq006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weaver SC, Ferro C, Barrera R, Boshell J, Navarro J-C, 2004. Venezuelan equine encephalitis. Annu. Rev. Entomol 49, 141–174. 10.1146/annurev.ento.49.061802.123422. [DOI] [PubMed] [Google Scholar]
- Weaver SC, Lecuit M, 2015. Chikungunya virus and the global spread ofa mosquito-bome disease. N. Engl. J. Med 372, 1231–1239. 10.1056/NEJMra1406035. [DOI] [PubMed] [Google Scholar]
- Weaver SC, Salas R, Rico-Hesse R, Ludwig GV, Oberste MS, Boshell J, Tesh RB, 1996. Re-emergence of epidemic Venezuelan equine encephalomyelitis in South America. VEE Study Group. Lancet: 348, 436–440. [DOI] [PubMed] [Google Scholar]
- Webb HE, Mehta S, Gregson NA, Leibowitz S, 1984. Immunological reaction of the demyelinating Semliki Forest virus with immune serum to glycolipids and its possible importance to central nervous system viral auto-immune disease. Neuropathol. Appi. Neurobiol 10, 77–84. [PubMed] [Google Scholar]
- Wesselingh SL, Levine B, Fox RJ, Choi S, Griffin DE, 1994. Intracerebral cytokine mRNA expression during fatal and nonfatal alphavirus encephalitis suggests a predominant type 2 T cell response. J. Immunol 152, 1289–1297. [PubMed] [Google Scholar]
- Weyand CM, Hicok KC, Conn DL, Goronzy JJ, 1992. The influence of HLA-DRB1 genes on disease severity in rheumatoid arthritis. Ann. Intern. Med 117, 801–806. 10.7326/0003-4819-117-10-801. [DOI] [PubMed] [Google Scholar]
- Wielanek AC, Monredon JD, Amrani ME, Roger JC, Serveaux JP, 2007. Guillain-Barré syndrome complicating a Chikungunya virus infection. Neurology 69, 2105–2107. 10.1212/01.wnl.0000277267.07220.88. [DOI] [PubMed] [Google Scholar]
- Willems WR, Kaluza G, Boschek CB, Bauer H, Hager H, Schütz HJ, Feistner H, 1979. Semliki forest virus: cause of a fatal case of human encephalitis. Science 203, 1127–1129. 10.1126/science.424742. [DOI] [PubMed] [Google Scholar]
- Willison HJ, Jacobs BC, van Doom PA, 2016. Guillain-Barré syndrome. Lancet 388, 717–727. 10.1016/S0140-6736(16)00339-1. [DOI] [PubMed] [Google Scholar]
- Wollish AC, Ferris MT, Blevins LK, Loo Y-M, Gale M, Heise MT, 2013. An attenuating mutation in a neurovirulent Sindbis virus strain interacts with the IPS-1 signaling pathway in vivo. Virology 435, 269–280. 10.1016/j.virol.2012.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woodman DR, McMANUS AT, Eddy GA, 1975. Extension of the mean time to death of mice with a lethal infection of Venezuelan equine encephalomyelitis virus by antithymocyte serum treatment. Infect. Immun 12, 1006–1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu LX, Mäkelä MJ, Röyttä M, Salmi A, 1988. Effect of viral infection on experimental allergic encephalomyelitis in mice. J. Neuroimmunol 18, 139–153. 10.1016/0165-5728(88)90062-8. [DOI] [PubMed] [Google Scholar]
- Yactayo S, Staples JE, Millot V, Cibrelus L, Ramon-Pardo P, 2016. Epidemiology of Chikungunya in the Americas. J. Infect. Dis 214, S441–S445. 10.1093/infdis/jiw390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zaid A, Gérardin P, Taylor A, Mostafavi H, Malvy D, Mahalingam S, 2018. Chikungunya arthritis: implications of acute and chronic inflammation mechanisms on disease management. Arthritis Rheumatol. 70, 484–495. 10.1002/art.40403. [DOI] [PubMed] [Google Scholar]
- Zambrano H, Waggoner JJ, Almeida C, Rivera L, Benjamin JQ, Pinsky BA, 2016. Zika virus and Chikungunya virus coinfections: a series of three cases from a single center in Ecuador. Am. J. Trop. Med. Hyg 95, 894–896. 10.4269/ajtmh.16-0323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zeller H, Van Bortel W, Sudre B, 2016. Chikungunya: its history in Africa and asia and its spread to new regions in 2013–2014. J. Infect. Dis 214, S436–S440. 10.1093/infdis/jiw391. [DOI] [PubMed] [Google Scholar]