arthritogenic alphaviruses trends in microbiology 2014 usar

9
Arthritogenic alphaviruses: new insights into arthritis and bone pathology Weiqiang Chen 1* , Suan-Sin Foo 1* , Natalie A. Sims 2 , Lara J. Herrero 1 , Nicole C. Walsh 2 , and Suresh Mahalingam 1 1 Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia 2 St Vincent’s Institute of Medical Research and Department of Medicine, St. Vincent’s Hospital, The University of Melbourne, Melbourne, VIC 3065, Australia Arthritogenic alphaviral infection begins as a febrile illness and often progresses to joint pain and rheumatic symptoms that are described as polyarthritis. Alphaviral arthritis and classical arthritides share many similar cellular and immune mediators involved in their patho- genesis. Recent in vitro and in vivo evidence suggests that bone loss resulting from increased expression of bone resorption mediators may accompany alphaviral infection. In addition, several longitudinal studies have reported more severe and delayed recovery of alphaviral disease in patients with pre-existing arthritic conditions. This review aims to provide insights into alphavirus- induced bone loss and focuses on aspects of disease exacerbation in patients with underlying arthritis and on possible therapeutic targets. Epidemiological and clinical burden of arthritogenic alphavirus infection Arthropod-borne alphaviruses are globally distributed and cause considerable human morbidity and mortality. Alpha- viruses cause a range of human diseases, with manifesta- tions varying from fever or rash to significant inflammatory pathologies including encephalitis and severe arthritis. En- cephalitic alphaviruses such as Venezuelan equine enceph- alitis virus (VEEV), Western equine encephalitis virus (WEEV) and Eastern equine encephalitis virus (EEEV) are neuroinvasive and frequently cause encephalitic man- ifestations [1]. By contrast, arthritogenic alphaviruses such as Ross River virus (RRV), Barmah Forest virus (BFV), o’nyong-nyong virus (ONNV), Mayaro virus (MAYV), chi- kungunya virus (CHIKV), Sindbis virus (SINV), and Sem- liki Forest virus (SFV) cause acute febrile illnesses, malaise, maculopapular rashes, myalgia, and severe arthralgia [2–8]. The potential of these viruses to cause chronic joint symptoms combined with their ability to cause large-scale epidemics makes alphavirus-associated disease an illness of major socioeconomic concern [9]. Typical acute clinical diag- nosis of arthritogenic alphaviruses involves virus detection through reverse transcription-polymerase chain reaction (RT-PCR) [10]. Serological testing for alphavirus-specific antibodies may also be performed using ELISA. Specific- IgM and IgG against alphavirus may be detected 3–7 days after disease onset, while specific-IgG could be detected up to 4 months post-onset [11]. In this review we focus on arthritogenic CHIKV and RRV, which have caused sporadic outbreaks worldwide. CHIKV was first isolated during an outbreak of dengue- like disease in southern Tanzania in 1952–1953 [12]. The recent reemergence of CHIKV in La Reunion in 2005 led to 300 000 infections with an estimated 250 deaths [13]. CHIKV then spread to India, where it affected over 1.3 million people [14] and continued to spread across the Asia-Pacific. Recently, CHIKV entered several Caribbean islands in the Americas, and to date the Pan American Health Organization (PAHO) has reported an estimated 715 000 suspected and confirmed cases as of September 12, 2014 (http://www.paho.org/hq/index.php?option=com_ content&view=article&id=9053&Itemid=39843). In con- trast to the widespread distribution of CHIKV, RRV is endemic to Australia, Papua New Guinea, and Western Pacific Islands and causes approximately 6000 cases annually [15]. Recent alphavirus outbreaks have provided increasing evidence of painful and persistent arthritic manifestations following infection. For example, patients with acute CHIKV infection during the 2005–2006 La Re ´union out- break presented with chronic polyarthritis in the ankles, wrists, knees, and small joints of the extremities [16]. Fif- teen months after CHIKV disease onset, only 43% of patients reported full remission, with more than half of the patients still experiencing arthritic rheumatic joint symptoms [17]. Other prospective longitudinal studies involving SINV-related alphaviruses in Northern Europe [18,19] and RRV in Australia [15,20] have reported similar arthritic symptoms to CHIKV infection, with over 95% of these patients developing polyarthritis during the acute phase of infection. Joint symptoms in these patients can last from months to years. Thus, arthritogenic alpha- viruses can cause arthralgia that is likely to evolve into Review 0966-842X/ ß 2014 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tim.2014.09.005 Corresponding author: Mahalingam, S. ([email protected]). Keywords: alphavirus; bone loss; arthritis; inflammation; Ross River virus; chikungunya virus. * These authors contributed equally to this manuscript. TIMI-1130; No. of Pages 9 Trends in Microbiology xx (2014) 1–9 1

Upload: inma-vazgut

Post on 08-Sep-2015

222 views

Category:

Documents


1 download

DESCRIPTION

virus

TRANSCRIPT

  • Arthritogenic alphavi

    A1

    222Me

    Review

    TIMI-1130; No. of Pages 9pathologies including encephalitis and severe arthritis. En-cephalitic alphaviruses such as Venezuelan equine enceph-alitis virus (VEEV), Western equine encephalitis virus(WEEV) and Eastern equine encephalitis virus (EEEV)are neuroinvasive and frequently cause encephalitic man-ifestations [1]. By contrast, arthritogenic alphaviruses suchas Ross River virus (RRV), Barmah Forest virus (BFV),onyong-nyong virus (ONNV), Mayaro virus (MAYV), chi-kungunya virus (CHIKV), Sindbis virus (SINV), and Sem-liki Forest virus (SFV) cause acute febrile illnesses, malaise,maculopapular rashes, myalgia, and severe arthralgia[28]. The potential of these viruses to cause chronic jointsymptoms combined with their ability to cause large-scaleepidemics makes alphavirus-associated disease an illness of

    trast to the widespread distribution of CHIKV, RRV isendemic to Australia, Papua New Guinea, and WesternPacific Islands and causes approximately 6000 casesannually [15].

    Recent alphavirus outbreaks have provided increasingevidence of painful and persistent arthritic manifestationsfollowing infection. For example, patients with acuteCHIKV infection during the 20052006 La Reunion out-break presented with chronic polyarthritis in the ankles,wrists, knees, and small joints of the extremities [16]. Fif-teen months after CHIKV disease onset, only 43% ofpatients reported full remission, with more than half ofthe patients still experiencing arthritic rheumatic jointsymptoms [17]. Other prospective longitudinal studiesinvolving SINV-related alphaviruses in Northern Europe[18,19] and RRV in Australia [15,20] have reported similararthritic symptoms to CHIKV infection, with over 95% ofthese patients developing polyarthritis during the acutephase of infection. Joint symptoms in these patients canlast from months to years. Thus, arthritogenic alpha-viruses can cause arthralgia that is likely to evolve into

    0966-842X/

    2014 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tim.2014.09.005

    Corresponding author: Mahalingam, S. ([email protected]).Keywords: alphavirus; bone loss; arthritis; inflammation; Ross River virus;chikungunya virus.*These authors contributed equally to this manuscript.insights into arthritpathologyWeiqiang Chen1*, Suan-Sin Foo1*, NatalieNicole C. Walsh2, and Suresh Mahalingam1 Institute for Glycomics, Griffith University, Gold Coast, QLD 42St Vincents Institute of Medical Research and Department of Melbourne, VIC 3065, Australia

    Arthritogenic alphaviral infection begins as a febrileillness and often progresses to joint pain and rheumaticsymptoms that are described as polyarthritis. Alphaviralarthritis and classical arthritides share many similarcellular and immune mediators involved in their patho-genesis. Recent in vitro and in vivo evidence suggeststhat bone loss resulting from increased expression ofbone resorption mediators may accompany alphaviralinfection. In addition, several longitudinal studies havereported more severe and delayed recovery of alphaviraldisease in patients with pre-existing arthritic conditions.This review aims to provide insights into alphavirus-induced bone loss and focuses on aspects of diseaseexacerbation in patients with underlying arthritis and onpossible therapeutic targets.

    Epidemiological and clinical burden of arthritogenicalphavirus infectionArthropod-borne alphaviruses are globally distributed andcause considerable human morbidity and mortality. Alpha-viruses cause a range of human diseases, with manifesta-tions varying from fever or rash to significant inflammatoryiruses: news and bone

    . Sims2, Lara J. Herrero1,

    , Australiadicine, St. Vincents Hospital, The University of Melbourne,

    major socioeconomic concern [9]. Typical acute clinical diag-nosis of arthritogenic alphaviruses involves virus detectionthrough reverse transcription-polymerase chain reaction(RT-PCR) [10]. Serological testing for alphavirus-specificantibodies may also be performed using ELISA. Specific-IgM and IgG against alphavirus may be detected 37 daysafter disease onset, while specific-IgG could be detected upto 4 months post-onset [11].

    In this review we focus on arthritogenic CHIKV andRRV, which have caused sporadic outbreaks worldwide.CHIKV was first isolated during an outbreak of dengue-like disease in southern Tanzania in 19521953 [12]. Therecent reemergence of CHIKV in La Reunion in 2005 ledto 300 000 infections with an estimated 250 deaths[13]. CHIKV then spread to India, where it affected over1.3 million people [14] and continued to spread across theAsia-Pacific. Recently, CHIKV entered several Caribbeanislands in the Americas, and to date the Pan AmericanHealth Organization (PAHO) has reported an estimated715 000 suspected and confirmed cases as of September 12,2014 (http://www.paho.org/hq/index.php?option=com_content&view=article&id=9053&Itemid=39843). In con-Trends in Microbiology xx (2014) 19 1

  • persistent arthritis. Despite a high percentage of patientsdeveloping persistent arthralgia, there is a lack of routineclinical surveillance of bone pathology. Hence, the impacton bone during alphavirus-induced arthritis warrants fur-ther investigation.

    Inflammation and bone lossThe maintenance of normal healthy bone structure is atightly regulated process requiring the coordinated actionsof bone-resorbing osteoclasts and bone-forming osteo-blasts. Signals elicited from cells within the local bonemicroenvironment, including signals arising from cellswithin the bone matrix [21] and the bone matrix itself[22], are crucial for regulating the activity of osteoblastsand osteoclasts. It is increasingly recognized that immunecells and their molecular mediators may contribute tonormal bone homeostasis and are key drivers of bone lossin chronic inflammatory conditions including the prototyp-ical inflammatory arthritis, rheumatoid arthritis (RA)[23]. In RA, periarticular (or juxta-articular) bone loss isevident in the inner network of trabecular bone in themetaphyseal region of the bone adjacent to the inflamedjoint; this often precedes focal bone erosion that occurs oncortical bone surfaces at the sites of inflammation(Figure 1). Systemic bone loss remote from the affectedjoints within both the appendicular and axial skeleton isalso commonplace in RA patients and is associated withincreased fracture risk (reviewed in [24,25]). A study in

    diagnosis and progressive bone loss in patients with activedisease (defined by serum C-reactive protein (CRP) levels>20 mg/dL) over 12 months [26]. Inflammation in RA leadsto bone loss by both increasing osteoclast-mediated boneresorption [27] and impairing osteoblast-mediated boneformation ([28], reviewed in [24]).

    The differentiation of osteoclasts from myeloid precur-sor cells requires the local presence of a pro-osteoclasto-genic cytokine termed RANKL [receptor activator ofnuclear factor (NF)-kB ligand] [29,30] and its interactionwith its receptor, RANK [receptor activator of (NF)-kB][31], which is expressed on the cell surface of osteoclastprecursors. The actions of RANKL can be inhibited byosteoprotegerin (OPG), a natural decoy receptor for RANKLthat prevents its interaction with RANK [32]. The relativeexpression of RANKL to OPG (RANKL:OPG ratio) withinthe local bone microenvironment controls osteoclast differ-entiation [33,34], with a high ratio leading to increasedosteoclastogenesis.

    In bone homeostasis, RANKL is supplied by cells of theosteoblast lineage, including early to midstage osteoblasts[35,36] and osteocytes [37]. In inflammatory arthritis suchas RA, other cell types including synovial fibroblasts[38,39] and T cells [38,40] provide additional sources ofRANKL, leading to increased RANKL:OPG in the inflamedsynovial tissue [41] and at the pannusbone interface [42],thus contributing to increased osteoclastogenesis withininflamed joints [27,43]. A study in treatment nave RA

    artht

    rus-i

    tion

    obse

    Review Trends in Microbiology xxx xxxx, Vol. xxx, No. x

    TIMI-1130; No. of Pages 9treatment-nave RA patients showed reduced bone miner-al density in patients with disease duration >6 months at

    Normaljoint

    Alphaviral join

    Bone

    Inamedmuscle

    Ligament

    Synovialmembrane

    Synovialuid

    Arcularcarlage

    Alphavirus

    Figure 1. Comparative joint pathology in rheumatoid arthritis (RA) and alphavi

    macrophages, synoviocytes, and bone-forming osteoblasts leading to the produc

    mouse model of alphavirus infection, both periarticular and systemic bone loss are local inflammation, with proliferation of the synovial lining cells and infiltration of this tis

    inflamed synovial tissue promote destruction of the articular cartilage and bone. Bone lo

    bone erosion at the margins of cartilage and bone; systemic bone loss away from the

    2patients also showed elevated systemic levels of serumRANKL:OPG at diagnosis [44].

    ris Rheumatoid arthrisjoint

    Inamedsynovial membrane

    Focal boneerosion

    Thinningarcular carlage

    Periarcular bone loss

    Auto-anbody

    TRENDS in Microbiology

    nduced arthritis. In alphavirus infection the virus infects skeletal muscle cells,

    of proinflammatory cytokines and chemokines which promote bone loss. In the

    rved. In RA, the joint lining (synovium) of diarthrodial joints is the primary target ofsue by inflammatory cells. Cytokines and chemokines produced by cells within this

    ss is evident in RA joints as periarticular bone loss adjacent to the joint, and as focal

    affected joint is also evident.

  • TIMI-1130; No. of Pages 9Emerging evidence for alphavirus-induced arthritisaffecting boneAlphaviral infection shares some features with inflamma-tory arthritic diseases such as RA. Whether these infec-tions affect bone in a similar manner to RA remains to beclearly determined. In the few studies that have includedanalysis of bone structure, the majority of CHIKV-infectedpatients do not show radiographic signs of focal boneerosion [45,46]. However, several case reports suggest thatfocal bone erosion does occur in a small number of CHIKV-infected patients. CHIKV-infected patients with no priorhistory of inflammatory arthritis showed magnetic reso-nance imaging (MRI) evidence of joint inflammation [47]and bone erosion [48] in the hands, and abnormal uptake ofradioactive tracers in the wrists and ankles. After the20052006 CHIKV outbreaks in La Reunion, Bouquillardand colleagues [49] reported 21 cases of CHIKV-infectedpatients diagnosed with RA according to the 1987 Ameri-can College of Rheumatology (ACR) criteria. Of these21 cases, five exhibited focal bone erosion at RA diagnosis(mean time of 10 months post-viral infection) and thisincreased to 17 patients 2 years post RA diagnosis [49]. Ra-diographic focal bone erosion was also reported by Mani-munda et al. [50] in three CHIKV-infected patients withoutpre-existing joint pain from a cohort of 20 patients report-ing persistent joint pain 10 months post-infection. Further-more, in this cohort MRI demonstrated that inflammationof the tendon, joint effusion, and bone marrow edema werecommon in the arthritis-affected joints [50]. Urinary hy-droxyproline in CHIKV-infected patients during activeviral infection has been reported to be higher than inhealthy controls, suggesting that bone resorption may beincreased in these patients [51]. In support of increasedosteoclast activity in alphaviral patients, a higher level ofcirculating serum TRAP5b activity was recently reported,together with greater synovial expression of RANKL:OPGratio in RRV-infected patients [52].

    The capacity of alphaviral infection to have a directeffect on skeletal health was highlighted by a recentpreclinical study in a mouse model of RRV infection[52]. Although focal bone erosion was not observed withinthe joints of RRV-infected mice, there was clear evidencefor periarticular bone loss within the tibial epiphysisand systemic bone loss within the vertebrae of these mice[52]. This bone loss was associated with increasedRANKL:OPG ratio and increased osteoclastogenesis[52,53]. Combined with the case reports in human alpha-viral infections, these findings suggest that effects ofalphaviral infection on skeletal health should be consid-ered in these patients, including study not only of thearthritis-affected joint but also of systemic bone health.It is worth noting that persistent low-grade inflammation,diagnosed by serum high-sensitivity CRP levels, is anindependent risk predictor for non-traumatic fractures[54]. This suggests that persistent low-grade inflamma-tion in patients with persistent alphavirus-induced ar-thritis might also place these patients at risk of systemicbone loss and therefore at greater risk of fracture. Future

    Reviewstudies investigating the effects of alphaviral infection onpatient bone mineral density over time are needed toconfirm this hypothesis.Recent evidence also suggests that alphaviruses canreplicate in joint tissues and stimulate an immune re-sponse in the bone microenvironment. Synovial biopsiesfrom RRV- [55] and CHIKV-infected [56] patients clearlyindicate the presence of viral antigens within cells of theinfected joints. Phuklia et al. demonstrated direct infectionof primary synoviocytes by CHIKV, and reported thatthese infected cells have the ability to drive monocyte/macrophage migration and osteoclast formation [57]. Re-cent studies using primary human osteoblasts have dem-onstrated that these cells also have high susceptibility toinfection with RRV and CHIKV, and that the infectedosteoblasts can induce the formation of bone-resorbingosteoclasts through RANKL expression [52,53], providinga mechanism by which alphaviruses could mediate boneloss.

    Recent preclinical models have provided clear evidencethat alphavirus infections result in inflammatory arthrop-athy affecting the joint, muscle, tendon, and bone. Existingmouse models of CHIKV arthritis using ventral foot injec-tion of the virus recapitulated the rheumatic symptoms ofhuman CHIKV infection, including localized foot swelling,tenosynovitis, and myositis [58,59]. By contrast, subcuta-neous and intradermal injections of CHIKV in this mousemodel produced viremia without foot swelling [58]. Withthis in mind, it is possible that the virus can still bedisseminated into joints and induces bone activity in theabsence of foot swelling. Earlier work with SINV-relatedalphavirus in adult mice demonstrated that the virusreplicated within the epiphyses of long bones adjacent toarticular joints when inoculated through intravenous,intraperitoneal, or subcutaneous routes [60]. This is sup-ported by recent work with RRV, where high levels ofinfectious virus were recovered from the femur, tibia,patella, and foot of infected mice [52]. In addition, thereis clear evidence of systemic bone loss in the tibial epiphy-sis and vertebrae in subcutaneously RRV-infected mice,suggesting that alphaviral infection of osteoblasts in ani-mals may directly lead to inflammation within the jointthat may share features with other human inflammatoryarthritides.

    Impact of the alphavirus-induced immune response onboneThe innate immune system acts as the frontline defenseagainst alphaviral invasion. Upon activation by alpha-virus, a sophisticated network of immune responses iselicited. Growing evidence has pinpointed the cell-mediat-ed immune response as the primary contributor to theimmunopathology and persistence of joint manifestationsduring alphaviral infection.

    There is a strong body of evidence that infiltratingmacrophages determine the severity and persistence ofalphaviral infection. Persistent infection of perivascularsynovial macrophages by CHIKV and constitutive infiltra-tion of CD14+ monocytic cells into the synovial cavity werereported in a CHIKV-infected patient 18 months postillness onset [56]. Extensive mononuclear cell infiltration

    Trends in Microbiology xxx xxxx, Vol. xxx, No. xand persistent CHIKV infection of macrophages are alsoevident in animal models [58,61], and depletion of macro-phages protects RRV-infected mice from disease.

    3

  • TIMI-1130; No. of Pages 9A vast array of cytokines, chemokines, and growth factorswere detected in the plasma of CHIKV-infected patients,including type I interferons (IFNs), C-X-C motif chemokineligand 10 (CXCL10/IP-10), interleukins (IL)-1b, IL-5, IL-6,IL-7, IL-10, IL-15, IL-17, chemokine (C-C motif) ligand(CCL)-3 (also known as macrophage inflammatory pro-tein-1a MIP-1a), CCL-4 (MIP-1b), CCL-2 (monocyte che-moattractant protein-1, MCP-1), granulocyte macrophagecolony-stimulating factor (GM-CSF), CCL-11 (eotaxin), andCCL-5 (also known as RANTES regulated on activation,normal T cell expressed and secreted) [62,63]. In the syno-vial fluid and tissue of CHIKV-infected patients, elevatedlevels of CCL-2, IL-6, and IL-8 have been reported [56]. Asimilar elevation of these immune mediators was also ob-served in RRV infection. Synovial fluid samples obtainedfrom RRV-infected patients exhibited significantly higherlevels of tumor necrosis factor (TNF)-a, IFN-g, IL-6, andreactive nitrogen intermediates (RNI) when compared toosteoarthritis (OA) patients [64]. Consistent with this ob-servation, RRV- and CHIKV-infected primary osteoblastsalso showed a greater increase in RANKL:OPG ratio andhigher expression of IL-6 and CCL-2; all are associated withenhanced osteoclastogenesis in response to alphaviral in-fection [52,53].

    A striking similarity in inflammatory responses be-tween the animal models of alphaviral infection and hu-man disease has been observed. Elevated expression ofimmune mediators such as TNF-a, CCL-2, IFN-g, and IL-6was evident in RRV-infected mice [64] and in CHIKV-infected mice [58] and non-human primates [61]. Inaddition, there was an IL-6-dependent increase in theRANKL:OPG ratio in serum from RRV-infected mice dur-ing peak disease [52], indicating that the osteoclastogenicenvironment induced by RRV could be therapeuticallytargeted by IL-6 inhibition. Furthermore, reduced produc-tion of TNF-a, CCL-2, IFN-g, and RNI was evident inRRV-induced arthritis in mice upon macrophage depletion,indicating that these cells may play a significant role inalphaviral joint pathology [64]. In a recent study, Poo et al.[65] reported that mice deficient in CCR2, a receptor forseven ligands including CCL2, CCL8, and CCL7, displayedexacerbated disease following CHIKV infection. This wascharacterized by neutrophil-dominant cellular infiltration,suggesting a role of neutrophils in mediating cartilagedamage during CHIKV infection [65]. However, treatmentof RRV-infected mice at therapeutic dose with bindarit, aninhibitor of CCL-2, CCL-8, and CCL-7, leads to diseaseamelioration [66]. Together, these studies suggest thatsuppressing the overexpression of alphavirus-inducedproinflammatory factors using a specific anti-inflammato-ry drug at therapeutic dose may be a viable option forpreventing joint inflammation.

    Pro-osteoclastic cytokines TNF-a, IL-6, and IL-1, whichare also prominent alphavirus-induced proinflammatorycytokines, have been implicated in alphavirus-inducedosteoclastogenesis and bone loss [67,68]. Moreover, chemo-tactic cytokines such as CXCL1, CCL-2, CCL-3, CCL-4,CCL-5, and CCL-11, which have each been implicated in

    Reviewthe egress of osteoclast precursors [69] and osteoclastdifferentiation [53,7072] in pathological bone conditions,are also elevated during alphaviral infection.

    4Recent studies have identified an indispensable role foradaptive immunity in modulating alphaviral disease patho-genesis. In particular, increased T cell activation was ob-served during acute CHIKV infection in humans[56,73]. Consistent with these clinical studies, egress of Tcells to the inflamed joints and muscle tissues was alsoobserved in acute models of RRV and CHIKV infection[53,59,74,75]. Further characterization identified a patho-genic role for CD4+T helper (Th) cells in disease pathogenesisthrough mediating inflammatory processes [76]. Similarly,T cell infiltration has been identified as a hallmark of RA, inwhich Th17 cells have been characterized as a T cell subsetthat can support osteoclastogenesis by virtue of theirRANKL expression [77]. Hence, the presence of Th17-asso-ciated cytokines IL-17, RANKL, TNF, and IL-6 during alpha-viral infection strongly suggests that Th17 cells may also beinvolved in alphaviral disease, including osteoclastogenesis.

    Taken together, the elevated profiles of inflammatorycytokines, pro-osteoclastic factors, chemokines, and Th17-associated cytokines play a crucial role in alphaviral ar-thritis. Most importantly, the increased RANKL:OPG ratioand osteoclastogenesis in alphavirus infection as a result ofinflammation demonstrate a strong association betweenthe immune response following alphaviral infection andbone health (Figure 2).

    Underlying inflammatory arthritis: potential risk factorfor exacerbated alphavirus-induced arthritisThere has been considerable interest in the persistent butperiodically flaring arthralgia that lasts for months afteralphavirus infection. Potential risk factors, including age45 years, initial severe joint pain, and pre-existing jointdisorders, were independently associated with persistenceof CHIKV arthralgia [17].

    Patients with underlying degenerative joint disordersmay be predisposed to delayed recovery from alphavirus-induced arthralgia [17,50,78]. For example, Borgheriniet al. reported that 44% of CHIKV-infected patients witha prior history of joint symptoms self-reported an extendedperiod of CHIKV arthralgia [79]. In addition, RRV-infectedpatients with pre-dating joint disease such as OA, gout, orpsoriatic arthritis complained of worse arthralgia monthsafter disease onset [15,20]. Similarly, the SINV-relatedalphavirus that causes Pogosta disease has been reportedto cause exacerbation of joint arthralgia in OA patients[19,80]. These observations suggest that underlying ar-thritic conditions may be associated with disease exacer-bation after alphavirus infection.

    The delayed recovery from arthralgia in alphavirus-infected patients with pre-existing arthritis may be areflection of ongoing inflammation caused by the underly-ing disease. However, it is very challenging to distinguishbetween the symptoms associated with pre-dating arthriticconditions and those associated with alphavirus-inducedarthritis. To date, alphavirus-infected patients with un-derlying arthritis have not been studied in detail. Thestriking similarities between the immunological responsessuch as the cellular and proinflammatory mediators eli-

    Trends in Microbiology xxx xxxx, Vol. xxx, No. xcited during alphaviral infection and arthritis such as RAfurther suggests the likelihood of interplay between theseinflammatory arthritides (Figure 3). It will be of interest to

  • avir

    n, v

    eocl

    to b

    TIMI-1130; No. of Pages 9identify and dissect the mechanisms by which arthrito-genic alphaviruses can exacerbate underlying arthritis-

    OPGRANKLRANK

    Osteoblasts

    Alphavirus

    Boneformaon

    Figure 2. Schematic representation of immune and skeletal responses during alph

    forming osteoblasts and bone-resorbing osteoclasts. Following alphaviral infectio

    infection of osteoblasts gives rise to the secretion of proinflammatory and pro-ost

    RANKL:OPG ratio is increased, favoring osteoclast differentiation and contributing

    Reviewinduced bone pathology.

    Potential therapeutic strategies to prevent alphavirus-induced arthritisBone loss is a newly identified characteristic of alphavirus-induced disease, which has yet to be addressed in currenttherapeutic approaches. The similarities in the immuneand osteoimmunological responses during alphavirus in-fection and inflammatory arthritis such as RA suggest thatcurrent therapeutics for RA may be useful in preventingalphavirus-induced bone loss. Many of the biologic treat-ments targeting proinflammatory cytokines in RA haveproved effective at halting the progression of focal erosion.These therapies may show similar efficacy in alphavirusinfection.

    Anti-IL-6 therapyIL-6 is a potent stimulus for RANKL expression in osteo-blast-lineage cells, which then promotes osteoclastogenesisand bone resorption [81]. IL-6 is produced by immune cells,osteoblasts, and synoviocytes [81,82]. In addition to its rolein alphavirus arthritis, overt expression of IL-6 has alsobeen implicated in bone loss in other bone disorders such asRA [83] and bone metastases [84]. Tocilizumab (TCZ), ahumanized anti-human IL-6R monoclonal antibody, iscurrently used to treat systemic juvenile idiopathic arthri-tis and RA [85,86], in which it successfully controls syno-vial inflammation and protects against progressive jointdestruction. In our mouse model of RRV infection, treat-ment with a neutralizing anti-IL-6 antibody effectivelyprevented the increase in the RANKL:OPG ratio and boneloss triggered by RRV [52]. Inhibition of IL-6 may alsoalleviate chronic joint pain associated with alphavirus

    Bone loss

    Osteoclasts

    Recruit

    Monocyc precursors

    OsteoclastogenesisRANKL:OPG

    Proinammatorycytokines

    (IL-6, IL-1, CCL-2)

    TRENDS in Microbiology

    al infection. In the bone microenvironment the two major cell subsets are bone-

    iruses are disseminated to inflamed joints, where osteoblasts are infected. The

    astic factors including IL-6, IL-1b, and CCL-2, as well as RANKL. Furthermore, the

    one loss.

    Trends in Microbiology xxx xxxx, Vol. xxx, No. xinfection because inflammatory mediators such as IL-6have been associated with hyperalgesia [87,88].

    TNF antagonistsTNF is a multifunctional cytokine that modulates inflam-matory and bone remodeling processes. It is produced atelevated levels by activated macrophages, synoviocytes,and T cells in RA synovial tissue [89,90]. Several TNFantagonists such as adalimumab, infliximab, and etaner-cept have been established as potent drugs for treatment ofinflammatory joint diseases including RA [91], psoriaticarthritis [92], and juvenile idiopathic arthritis [93]. Despitethe reported efficacy of etanercept in treating inflammato-ry diseases, detrimental effects of etanercept administra-tion have been shown in an acute RRV disease mousemodel: etanercept treatment at disease onset led to pro-nounced myositis and increased viral titers as a result ofdiminished alphavirus-triggered type I IFN responses[94,95]. These undesirable effects on acute infection indi-cate that caution should be taken during any trials ofetanercept for viral arthritis. These side-effects might beavoided if the drug is administered after the acute phase ofdisease, when the persistent joint manifestations develop.The potential of adalimumab, infliximab, and etanercept totreat persistent alphavirus-induced bone loss warrantsfurther investigation.

    Direct targeting of Th17 cells and related cytokinesActivation of T cells plays a pivotal role in the interplaybetween the immune and skeletal systems. Th17 cells arecapable of supporting osteoclast differentiation and there

    5

  • am

    R

    ma

    TIMI-1130; No. of Pages 9Proinammatorycytokines

    (TNF, IL-6, IL-1)

    B cell

    Macrophage

    In

    Carlage immune complex

    Inam

    Dierenate

    Dierenate

    Reviewis a growing interest in therapeutic targeting of Th17-related cytokines for pathological bone conditions. In par-ticular, IL-17 has been the primary target for therapeuticintervention because it is capable of inducing RANKLexpression on synovial fibroblasts [91]. Anti-IL-17A anti-body treatment is efficacious in alleviating joint pathologyand bone erosions and in lowering RANKL expression inseveral arthritic mouse models including adjuvant-in-duced arthritis (AIA) [96], collagen-induced arthritis(CIA) [97], and antigen-induced experimental arthritis[98]. A Phase II clinical trial is currently underway usingan anti-IL-17 antibody (AIN457) in RA patients [99].

    Anti-RANKL therapyRANKL is a pivotal cytokine that stimulates bone resorp-tion. RANKL has been targeted during pathological boneconditions using a neutralizing anti-RANKL antibody,denosumab. Denosumab is efficacious in treating osteopo-rosis [100102] and has been approved by several healthauthorities including Health Canada and the US FDA forthe treatment of postmenopausal women predisposed toosteoporotic fractures [101,103]. In RA patients, despitehaving no effect on disease activity, denosumab givenevery 6 months in combination with standard methotrex-ate treatment resulted in reduced radiographic scores forfocal bone erosion, suppressed serum markers of boneresorption [serum C-telopeptide of type I collagen (CTX-I) and serum N-propeptide of type I collagen (PINP)] and

    Figure 3. Comparative diagram of immune responses regulated during alphavirus-ind

    alphaviral infection and in RA can affect several immune parameters. During inflamma

    which contribute to the clinical outcomes. Following activation, macrophages present an

    Nave CD8+ T cells can also be activated by antigen-presenting cells such as macrophag

    contribute to radiologic bone erosion. Nave CD4 T helper cells can differentiate into type

    express proinflammatory factors (IL-6, IL-1b, and TNF-a) which have the ability to

    osteoclastogenesis is pivotal in the induction of bone pathologies.

    6RRV-induced arthris

    Rheumatoid arthris

    Osteoclast

    IL-17

    Interplay?

    Bone loss

    maon

    ANKL

    on

    Bone erosion/loss

    TRENDS in Microbiology

    Trends in Microbiology xxx xxxx, Vol. xxx, No. ximproved bone mineral density compared to patients re-ceiving placebo [104]. These clinical observations clearlysuggest that denosumab is effective in amelioratingRANKL-induced bone erosion in RA and may be a potentialtherapeutic option for alphavirus-induced bone loss.

    Concluding remarksDebilitating persistent polyarthralgias induced by arthri-togenic alphavirus infection is a clinical enigma. Emergingclinical evidence of bone pathologies in alphavirus-infectedpatients has identified this as a possible risk factor for theaggravation of joint inflammation. In light of the ominousthreat of a global chikungunya epidemic, and the lack oftherapeutic alternatives, intensive research into the exactmechanisms underlying the development of persistentpolyarthralgias is warranted. Preclinical studies suggestthat there is a clear link between alphavirus-inducedinflammation and periarticular and systemic bone loss.The potential for alphavirus infection to precipitate boneloss and increase fracture risk in humans is an importantclinical question (Box 1). Furthermore, the impact of alpha-viral infection on bone health in patients with pre-existingarthritis also remains to be determined. In the prototypicalinflammatory arthritis, RA, local and systemic inflamma-tion leads to focal bone erosion within the cortical bone,bone loss in the periarticular trabecular bone adjacent tothe affected joint, and systemic bone loss. By contrast,inflammation in the mouse models of alphavirus infection

    uced and RA bone loss. The expression of host proinflammatory factors during

    tion, macrophages take central stage to facilitate several immunological pathways

    tigen to CD4 T helper cells, which in turn activate cytotoxic CD8 T cells and B cells.

    es. In RA, B cells are a source of rheumatoid factors (RF) and autoantibodies, which

    17 T helper (Th17) subsets which express IL-17 and RANKL. Macrophages can also

    enhance osteoclastogenesis. In both RA and alphaviral arthritis, the enhanced

  • TIMI-1130; No. of Pages 9leads primarily to periarticular and systemic trabecularbone loss with no evidence of focal bone erosion. Despitedistinct differences in bone pathologies resulting fromthese two forms of arthritides, some similarities have alsobeen observed, such as inflammatory mediators and cellu-lar responses leading to increased osteoclastogenesis. Thesimilarities between RA-induced inflammation and boneloss and that observed in alphavirus infection suggeststherapeutic approaches used in the treatment of RA, suchas cytokine and RANKL inhibition, may also be appropri-ate for the treatment of persistent alphavirus-inducedbone disease.

    AcknowledgmentsS.M. is the recipient of an Australian National Health and MedicalResearch Council (NHMRC) Senior Research Fellowship (APP1059167).

    References1 Zacks, M.A. and Paessler, S. (2010) Encephalitic alphaviruses. Vet.Microbiol. 140, 281286

    2 Rulli, N. et al. (2007) The molecular and cellular aspects of arthritisdue to alphavirus infections: lesson learned from Ross River virus.Ann. N. Y. Acad. Sci. 1102, 96108

    3 Suhrbier, A. and La Linn, M. (2004) Clinical and pathologic aspects ofarthritis due to Ross River virus and other alphaviruses. Curr. Opin.Rheumatol. 16, 374379

    4 Kiwanuka, N. et al. (1999) Onyong-nyong fever in south-centralUganda, 1996-1997: clinical features and validation of a clinicalcase definition for surveillance purposes. Clin. Infect. Dis. 29,12431250

    5 Laine, M. et al. (2004) Sindbis viruses and other alphaviruses as causeof human arthritic disease. J. Intern. Med. 256, 457471

    6 Munoz, M. and Navarro, J.C. (2012) Mayaro: a re-emergingarbovirus in Venezuela and Latin America. Biomedica 32, 286302(in Spanish)

    7 Phillips, D.A. et al. (1990) Clinical and subclinical Barmah Forestvirus infection in Queensland. Med. J. Aust. 152, 463466

    8 Paquet, C. et al. (2006) Chikungunya outbreak in Reunion:epidemiology and surveillance, 2005 to early January 2006. EuroSurveill. 11, E060202E060203

    9 Suhrbier, A. and Mahalingam, S. (2009) The immunobiology of viralarthritides. Pharmacol. Ther. 124, 301308

    10 Parida, M.M. (2008) Rapid and real-time detection technologies foremerging viruses of biomedical importance. J. Biosci. 33, 617628

    Box 1. Outstanding questions

    Do underlying arthritic conditions exacerbate alphavirus-inducedbone loss (or vice versa)?

    What is the effect of alphaviral infection on bone health? Are thesepatients at risk of fracture as a result of infection?

    Does alphavirus infection of bone persist during disease progres-sion? Is alphavirus infection of bone responsible for periodicflares of disease?

    Do immune cells contribute to alphavirus-induced bone loss?

    Review11 Aoyama, I. et al. (2010) A case of chikungunya fever imported fromIndia to Japan, follow-up of specific IgM and IgG antibodies over a 6-month period. Jpn. J. Infect. Dis. 63, 6566

    12 Robinson, M.C. (1955) An epidemic of virus disease in SouthernProvince, Tanganyika Territory, in 1952-53. I. Clinical features.Trans. R. Soc. Trop. Med. Hyg. 49, 2832

    13 Josseran, L. et al. (2006) Chikungunya disease outbreak, ReunionIsland. Emerg. Infect. Dis. 12, 19941995

    14 Krishnamoorthy, K. et al. (2009) Burden of chikungunya in India:estimates of disability adjusted life years (DALY) lost in2006 epidemic. J. Vector Borne Dis. 46, 2635

    15 Harley, D. et al. (2002) Ross River virus disease in tropicalQueensland: evolution of rheumatic manifestations in an inceptioncohort followed for six months. Med. J. Aust. 177, 35235516 Staikowsky, F. et al. (2009) Prospective study of chikungunya virusacute infection in the Island of La Reunion during the 20052006outbreak. PLoS ONE 4, e7603

    17 Sissoko, D. et al. (2009) Post-epidemic chikungunya disease onReunion Island: course of rheumatic manifestations and associatedfactors over a 15-month period. PLoS Negl. Trop. Dis. 3, e389

    18 Kurkela, S. et al. (2005) Clinical and laboratory manifestations ofSindbis virus infection: prospective study, Finland, 20022003. J.Infect. Dis. 191, 18201829

    19 Laine, M. et al. (2000) Prolonged arthritis associated with sindbis-related (Pogosta) virus infection. Rheumatology (Oxford) 39,12721274

    20 Mylonas, A.D. et al. (2002) Natural history of Ross River virus-inducedepidemic polyarthritis. Med. J. Aust. 177, 356360

    21 Sims, N.A. and Vrahnas, C. (2014) Regulation of cortical and trabecularbone mass by communication between osteoblasts, osteocytes andosteoclasts. Arch. Biochem. Biophys. http://dx.doi.org/10.1016/j.abb.2014.05.015

    22 McHugh, K.P. et al. (2010) The role of cell-substrate interaction inregulating osteoclast activation: potential implications in targetingbone loss in rheumatoid arthritis. Ann. Rheum. Dis. 69 (Suppl. 1),i83i85

    23 Danks, L. and Takayanagi, H. (2013) Immunology and bone. J.Biochem. 154, 2939

    24 Deal, C. et al. (2012) Bone loss in rheumatoid arthritis: systemic,periarticular, and focal. Curr. Rheumatol. Rep. 14, 231237

    25 Vis, M. et al. (2013) Can bone loss in rheumatoid arthritis beprevented? Osteoporos. Int. 24, 25412553

    26 Gough, A.K. et al. (1994) Generalised bone loss in patients with earlyrheumatoid arthritis. Lancet 344, 2327

    27 Gravallese, E.M. et al. (1998) Identification of cell types responsiblefor bone resorption in rheumatoid arthritis and juvenile rheumatoidarthritis. Am. J. Pathol. 152, 943951

    28 Walsh, N.C. et al. (2009) Osteoblast function is compromised at sites offocal bone erosion in inflammatory arthritis. J. Bone Miner. Res. 24,15721585

    29 Lacey, D.L. et al. (1998) Osteoprotegerin ligand is a cytokine thatregulates osteoclast differentiation and activation. Cell 93, 165176

    30 Kong, Y.Y. et al. (1999) OPGL is a key regulator of osteoclastogenesis,lymphocyte development and lymph-node organogenesis. Nature 397,315323

    31 Dougall, W.C. et al. (1999) RANK is essential for osteoclast and lymphnode development. Genes Dev. 13, 24122424

    32 Simonet, W.S. et al. (1997) Osteoprotegerin: a novel secreted proteininvolved in the regulation of bone density. Cell 89, 309319

    33 Horwood, N.J. et al. (1998) Osteotropic agents regulate the expressionof osteoclast differentiation factor and osteoprotegerin in osteoblasticstromal cells. Endocrinology 139, 47434746

    34 Fazzalari, N.L. et al. (2001) The ratio of messenger RNA levels ofreceptor activator of nuclear factor kappaB ligand to osteoprotegerincorrelates with bone remodeling indices in normal human cancellousbone but not in osteoarthritis. J. Bone Miner. Res. 16, 10151027

    35 Gori, F. et al. (2000) The expression of osteoprotegerin and RANKligand and the support of osteoclast formation by stromal-osteoblastlineage cells is developmentally regulated. Endocrinology 141,47684776

    36 Atkins, G.J. et al. (2003) RANKL expression is related to thedifferentiation state of human osteoblasts. J. Bone Miner. Res. 18,10881098

    37 Nakashima, T. et al. (2011) Evidence for osteocyte regulation of bonehomeostasis through RANKL expression. Nat. Med. 17, 12311234

    38 Gravallese, E.M. et al. (2000) Synovial tissue in rheumatoid arthritisis a source of osteoclast differentiation factor. Arthritis Rheum. 43,250258

    39 Takayanagi, H. et al. (2000) Involvement of receptor activator ofnuclear factor kappaB ligand/osteoclast differentiation factor inosteoclastogenesis from synoviocytes in rheumatoid arthritis.Arthritis Rheum. 43, 259269

    40 Kong, Y.Y. et al. (1999) Activated T cells regulate bone loss and jointdestruction in adjuvant arthritis through osteoprotegerin ligand.Nature 402, 304309

    Trends in Microbiology xxx xxxx, Vol. xxx, No. x41 Haynes, D. et al. (2008) Modulation of RANKL and osteoprotegerinexpression in synovial tissue from patients with rheumatoid arthritis

    7

  • TIMI-1130; No. of Pages 9in response to disease-modifying antirheumatic drug treatment andcorrelation with radiologic outcome. Arthritis Rheum. 59, 911920

    42 Pettit, A.R. et al. (2006) RANKL protein is expressed at the pannus-bone interface at sites of articular bone erosion in rheumatoidarthritis. Rheumatology (Oxford) 45, 10681076

    43 Romas, E. et al. (2000) Expression of osteoclast differentiation factorat sites of bone erosion in collagen-induced arthritis. Arthritis Rheum.43, 821826

    44 Xu, S. et al. (2012) Osteoprotegerin and RANKL in the pathogenesisof rheumatoid arthritis-induced osteoporosis. Rheumatol. Int. 32,33973403

    45 Chopra, A. et al. (2008) Chikungunya virus aches and pains: anemerging challenge. Arthritis Rheum. 58, 29212922

    46 Jaffar-Bandjee, M.C. et al. (2009) Chikungunya virus takes centrestage in virally induced arthritis: possible cellular and molecularmechanisms to pathogenesis. Microbes Infect. 11 (1415), 12061218

    47 Parola, P. et al. (2006) Novel chikungunya virus variant in travelersreturning from Indian Ocean islands. Emerg. Infect. Dis. 12,14931499

    48 Malvy, D. et al. (2009) Destructive arthritis in a patient withchikungunya virus infection with persistent specific IgM antibodies.BMC Infect. Dis. 9, 200

    49 Bouquillard, E. and Combe, B. (2009) A report of 21 cases ofrheumatoid arthritis following chikungunya fever. A mean follow-up of two years. Joint Bone Spine 76, 654657

    50 Manimunda, S.P. et al. (2010) Clinical progression of chikungunyafever during acute and chronic arthritic stages and the changes injoint morphology as revealed by imaging. Trans. R. Soc. Trop. Med.Hyg. 104, 392399

    51 Lokireddy, S. et al. (2008) Connective tissue metabolism inchikungunya patients. Virol. J. 5, 31

    52 Chen, W. et al. (2014) Arthritogenic alphaviral infection perturbsosteoblast function and triggers pathologic bone loss. Proc. Natl.Acad. Sci. U.S.A. 111, 60406045

    53 Noret, M. et al. (2012) Interleukin 6, RANKL, and osteoprotegerinexpression by chikungunya virus-infected human osteoblasts. J.Infect. Dis. 206, 455457

    54 Schett, G. et al. (2006) High-sensitivity C-reactive protein and risk ofnontraumatic fractures in the Bruneck study. Arch. Intern. Med. 166,24952501

    55 Soden, M. et al. (2000) Detection of viral ribonucleic acid and histologicanalysis of inflamed synovium in Ross River virus infection. ArthritisRheum. 43, 365369

    56 Hoarau, J.J. et al. (2010) Persistent chronic inflammation andinfection by chikungunya arthritogenic alphavirus in spite of arobust host immune response. J. Immunol. 184, 59145927

    57 Phuklia, W. et al. (2013) Osteoclastogenesis induced by CHIKV-infected fibroblast-like synoviocytes: a possible interplay betweensynoviocytes and monocytes/macrophages in CHIKV-inducedarthralgia/arthritis. Virus Res. 177, 179188

    58 Gardner, J. et al. (2010) Chikungunya virus arthritis in adult wild-type mice. J. Virol. 84, 80218032

    59 Morrison, T.E. et al. (2011) A mouse model of chikungunya virus-induced musculoskeletal inflammatory disease: evidence of arthritis,tenosynovitis, myositis, and persistence. Am. J. Pathol. 178, 3240

    60 Heise, M.T. et al. (2000) Sindbis-group alphavirus replication inperiosteum and endosteum of long bones in adult mice. J. Virol.74, 92949299

    61 Labadie, K. et al. (2010) Chikungunya disease in nonhuman primatesinvolves long-term viral persistence in macrophages. J. Clin. Invest.120, 894906

    62 Chow, A. et al. (2011) Persistent arthralgia induced by chikungunyavirus infection is associated with interleukin-6 and granulocytemacrophage colony-stimulating factor. J. Infect. Dis. 203, 149157

    63 Ng, L.F. et al. (2009) IL-1beta, IL-6, and RANTES as biomarkers ofchikungunya severity. PLoS ONE 4, e4261

    64 Lidbury, B.A. et al. (2008) Macrophage-derived proinflammatoryfactors contribute to the development of arthritis and myositisafter infection with an arthrogenic alphavirus. J. Infect. Dis. 197,15851593

    65 Poo, Y.S. et al. (2014) CCR2 deficiency promotes exacerbated chronic

    Reviewerosive neutrophil-dominated chikungunya virus arthritis. J. Virol.88, 68626872

    866 Rulli, N.E. et al. (2009) Amelioration of alphavirus-induced arthritisand myositis in a mouse model by treatment with bindarit, aninhibitor of monocyte chemotactic proteins. Arthritis Rheum. 60,25132523

    67 Boyle, W.J. et al. (2003) Osteoclast differentiation and activation.Nature 423, 337342

    68 Suda, T. et al. (1999) Modulation of osteoclast differentiation andfunction by the new members of the tumor necrosis factor receptor andligand families. Endocr. Rev. 20, 345357

    69 Onan, D. et al. (2009) The chemokine Cxcl1 is a novel target gene ofparathyroid hormone (PTH)/PTH-related protein in committedosteoblasts. Endocrinology 150, 22442253

    70 Abe, M. et al. (2009) Vicious cycle between myeloma cell binding to bonemarrow stromal cells via VLA-4VCAM-1 adhesion and macrophageinflammatory protein-1alpha and MIP-1beta production. J. BoneMiner. Metab. 27, 1623

    71 Choi, S.J. et al. (2000) Macrophage inflammatory protein 1-alpha is apotential osteoclast stimulatory factor in multiple myeloma. Blood 96,671675

    72 Hueber, W. et al. (2007) Proteomic analysis of secreted proteins inearly rheumatoid arthritis: anti-citrulline autoreactivity is associatedwith up regulation of proinflammatory cytokines. Ann. Rheum. Dis.66, 712719

    73 Wauquier, N. et al. (2011) The acute phase of chikungunya virusinfection in humans is associated with strong innate immunity and TCD8 cell activation. J. Infect. Dis. 204, 115123

    74 Gunn, B.M. et al. (2012) Mannose binding lectin is required foralphavirus-induced arthritis/myositis. PLoS Pathog. 8, e1002586

    75 Herrero, L.J. et al. (2013) Macrophage migration inhibitory factorreceptor CD74 mediates alphavirus-induced arthritis and myositisin murine models of alphavirus infection. Arthritis Rheum. 65,27242736

    76 Teo, T.H. et al. (2013) A pathogenic role for CD4+ T cells duringchikungunya virus infection in mice. J. Immunol. 190, 259269

    77 Sato, K. et al. (2006) Th17 functions as an osteoclastogenic helper Tcell subset that links T cell activation and bone destruction. J. Exp.Med. 203, 26732682

    78 Kularatne, S.A. et al. (2012) Epidemiology, clinical manifestations,and long-term outcomes of a major outbreak of chikungunya in ahamlet in Sri Lanka, in 2007: a longitudinal cohort study. J. Trop.Med. 2012, 639178

    79 Borgherini, G. et al. (2008) Persistent arthralgia associated withchikungunya virus: a study of 88 adult patients on Reunion Island.Clin. Infect. Dis. 47, 469475

    80 Sane, J. et al. (2011) Epidemic Sindbis virus infection in Finland: apopulation-based case-control study of risk factors. J. Infect. Dis. 204,459466

    81 Ishimi, Y. et al. (1990) IL-6 is produced by osteoblasts and inducesbone resorption. J. Immunol. 145, 32973303

    82 Guerne, P-A. et al. (1989) Synovium as a source of interleukin 6 invitro. Contribution to local and systemic manifestations of arthritis. J.Clin. Invest. 83, 585

    83 Lems, W.F. and Dijkmans, B.A. (1998) Should we look for osteoporosisin patients with rheumatoid arthritis? Ann. Rheum. Dis. 57, 325327

    84 Zheng, Y. et al. (2014) Direct cross-talk between cancer and osteoblastlineage cells fuels metastatic growth in bone via auto-amplificationof IL-6 and RANKL signaling pathways. J. Bone Miner. Res. 29,19381949

    85 Yokota, S. et al. (2005) Therapeutic efficacy of humanizedrecombinant anti-interleukin-6 receptor antibody in children withsystemic-onset juvenile idiopathic arthritis. Arthritis Rheum. 52,818825

    86 Smolen, J.S. et al. (2008) Effect of interleukin-6 receptor inhibitionwith tocilizumab in patients with rheumatoid arthritis (OPTIONstudy): a double-blind, placebo-controlled, randomised trial. Lancet371, 987997

    87 Svensson, C.I. (2010) Interleukin-6: a local pain trigger? Arthritis Res.Ther. 12, 145

    88 Schaible, H.G. et al. (2002) Mechanisms of pain in arthritis. Ann. N. Y.Acad. Sci. 966, 343354

    89 Danning, C.L. et al. (2000) Macrophage-derived cytokine and nuclear

    Trends in Microbiology xxx xxxx, Vol. xxx, No. xfactor kappaB p65 expression in synovial membrane and skin ofpatients with psoriatic arthritis. Arthritis Rheum. 43, 12441256

  • 90 MacNaul, K.L. et al. (1990) Analysis of IL-1 and TNF-alpha geneexpression in human rheumatoid synoviocytes and normal monocytesby in situ hybridization. J. Immunol. 145, 41544166

    91 Fossiez, F. et al. (1996) T cell interleukin-17 induces stromal cells toproduce proinflammatory and hematopoietic cytokines. J. Exp. Med.183, 25932603

    92 Schabert, V.F. et al. (2012) Disability outcomes and dose escalationwith etanercept, adalimumab, and infliximab in rheumatoid arthritispatients: a US-based retrospective comparative effectiveness study.Curr. Med. Res. Opin. 28, 569580

    93 Ungar, W.J. et al. (2011) Cost-effectiveness of biologics in polyarticular-course juvenile idiopathic arthritis patients unresponsive to disease-modifying antirheumatic drugs. Arthritis Care Res. (Hoboken) 63,111119

    94 Zaid, A. et al. (2011) Disease exacerbation by etanercept in a mousemodel of alphaviral arthritis and myositis. Arthritis Rheum. 63, 488491

    95 Zaid, A. et al. (2013) Exacerbation of alphaviral arthritis and myositisin a mouse model after etanercept treatment is due to diminishedlevels of interferon a/b. Virol. Mycol. 2, 122

    96 Chao,C.C.etal. (2011)Anti-IL-17Atherapy protectsagainstboneerosionin experimental models of rheumatoid arthritis. Autoimmunity 44,243252

    97 Lubberts, E. et al. (2004) Treatment with a neutralizing anti-murineinterleukin-17 antibody after the onset of collagen-induced arthritis

    reduces joint inflammation, cartilage destruction, and bone erosion.Arthritis Rheum. 50, 650659

    98 Koenders, M.I. et al. (2005) Blocking of interleukin-17 duringreactivation of experimental arthritis prevents joint inflammationand bone erosion by decreasing RANKL and interleukin-1. Am. J.Pathol. 167, 141149

    99 Miossec, P. et al. (2009) Interleukin-17 and type 17 helper T cells. N.Engl. J. Med. 361, 888898

    100 Cummings, S.R. et al. (2009) Denosumab for prevention of fracturesin postmenopausal women with osteoporosis. N. Engl. J. Med. 361,756765

    101 Josse, R. et al. (2013) Denosumab, a new pharmacotherapy option forpostmenopausal osteoporosis. Curr. Med. Res. Opin. 29, 205216

    102 Tyagi, A.M. et al. (2014) Enhanced immunoprotective effects by anti-IL17 antibody translates to improved skeletal parameters underestrogen deficiency compared to anti-RANKL and anti-TNFalphaantibodies. J. Bone Miner. Res. 29, 19811992

    103 Rizzoli, R. et al. (2010) Denosumab. Nature reviews. Drug Discov. 9,591592

    104 Cohen, S.B. et al. (2008) Denosumab treatment effects on structuraldamage, bone mineral density, and bone turnover in rheumatoidarthritis: a twelve-month, multicenter, randomized, double-blind,placebo-controlled, phase II clinical trial. Arthritis Rheum. 58,12991309

    Review Trends in Microbiology xxx xxxx, Vol. xxx, No. x

    TIMI-1130; No. of Pages 99

    Arthritogenic alphaviruses: new insights into arthritis and bone pathologyEpidemiological and clinical burden of arthritogenic alphavirus infectionInflammation and bone lossEmerging evidence for alphavirus-induced arthritis affecting boneImpact of the alphavirus-induced immune response on boneUnderlying inflammatory arthritis: potential risk factor for exacerbated alphavirus-induced arthritisPotential therapeutic strategies to prevent alphavirus-induced arthritisAnti-IL-6 therapyTNF antagonistsDirect targeting of Th17 cells and related cytokinesAnti-RANKL therapy

    Concluding remarksAcknowledgmentsReferences