You are on page 1of 8

Microbiological Research 207 (2018) 108–115

Contents lists available at ScienceDirect

Microbiological Research
journal homepage: www.elsevier.com/locate/micres

Leptospirosis in human: Biomarkers in host immune responses T


a,⁎ b,c b,d a,e a a b
Chin VK , Lee TY , Lim WF , Wan Shahriman YWY , Syafinaz AN , Zamberi S , Maha A
a
Department of Medical Microbiology & Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia
b
Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
c
School of Foundation Studies, Perdana University, 43400, Serdang, Malaysia
d
Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA Selangor, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia
e
Department of Medical Laboratory Technology, Faculty of Health Sciences, Universiti Teknologi MARA, Cawangan Selangor Kampus Puncak Alam, 42300 Bandar
Puncak Alam, Selangor, Malaysia

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

Keywords: Leptospirosis remains one of the most widespread zoonotic diseases caused by spirochetes of the genus
Leptospirosis Leptospira, which accounts for high morbidity and mortality globally. Leptospiral infections are often found in
Host immune responses tropical and subtropical regions, with people exposed to contaminated environments or animal reservoirs are at
Biomarkers high risk of getting the infection. Leptospirosis has a wide range of clinical manifestations with non-specific signs
Cytokines
and symptoms and often misdiagnosed with other acute febrile illnesses at early stage of infection. Despite being
Immune mediators
Proteomic approach
one of the leading causes of zoonotic morbidity worldwide, there is still a gap between pathogenesis and human
immune responses during leptospiral infection. It still remains obscure whether the severity of the infection is
caused by the pathogenic properties of the Leptospira itself, or it is a consequence of imbalance host immune
factors. Hence, in this review, we seek to summarize the past and present milestone findings on the biomarkers of
host immune response aspects during human leptospiral infection, including cytokine and other immune
mediators. A profound understanding of the interlink between virulence factors and host immune responses
during human leptospirosis is imperative to identify potential biomarkers for diagnostic and prognostic appli-
cations as well as designing novel immunotherapeutic strategies in future.

1. Introduction renal insufficiency and jaundice. Symptoms may be biphasic and some
can resolve spontaneously. However, these symptoms and signs are
Leptospirosis is a globally widespread zoonotic disease caused by non-specific and could be frequently misdiagnosed with other causes of
spirochetes Leptospira which has high impact on health consequences in acute febrile illness such as dengue fever, influenza or malaria (Bharti
human and domestic animals (Levett, 2001; Bharti et al., 2003). Human et al., 2003; Wuthiekanun et al., 2007).
leptospirosis is transmitted through direct contact of wounds with urine The development of severe leptospirosis could be accompanied by
and tissues of the infected animals or indirectly through the mucous symptoms which include jaundice, pulmonary haemorrhage, hepatic
membranes of the nose, eyes and mouth with contaminated water or and kidney failure (Chirathaworn et al., 2016). Of the complications
soil (Bharti et al., 2003; Palaniappan et al., 2007). Pathogenic Leptospira that arise from leptospiral infection, severe pulmonary haemorrhage
species have their own affinity and special adaptations towards specific syndrome (SPHS) and Weil’s syndrome (combination of acute renal
mammals and hence causes clinical manifestations in varying degrees insufficiency, haemorrhage and jaundice), are among the well-known
(Levett, 2001). For example, rodent reservoirs are carriers for Leptospira forms of severe leptospirosis. Notably, mortality recorded in Weil’s
species and do not show any diseases. Meanwhile, infected companion disease and severe pulmonary haemorrhage syndrome (SPHS) are >
animals or livestock may suffer from abortion and multiple organs in- 10% and 50%, respectively (McBride et al., 2005; Gouveia et al., 2008).
juries and infected humans may present with a variety of clinical High incidences of leptospiral infection are often seen in tropical
manifestations from asymptomatic infection to life threatening disease and subtropical regions. There are approximately 1.03 million cases
(Ashford et al., 2000; Ganoza et al., 2010). With mild infections, pa- and 59,000 deaths reported globally (Costa et al., 2015; Torgerson
tients typically present with symptoms viz fever, headache, and et al., 2015). Hence, the high frequency of leptospiral infection con-
myalgia, and less frequently by meningitis, conjunctival suffusion, rash, stitutes a serious clinical problem worldwide that needs to be addressed


Corresponding author.
E-mail addresses: cvk717@gmail.com (C. VK), tzeyan.lee@gmail.com (L. TY), limwaifeng85@gmail.com (L. WF), shahrimanuitm@gmail.com (W.S. YWY),
syafinaz@upm.edu.my (S. AN), zamberi@upm.edu.my (Z. S), maha@upm.edu.my (M. A).

https://doi.org/10.1016/j.micres.2017.11.015
Received 8 June 2017; Received in revised form 23 November 2017; Accepted 25 November 2017
Available online 28 November 2017
0944-5013/ © 2017 Elsevier GmbH. All rights reserved.
C. VK et al. Microbiological Research 207 (2018) 108–115

properly. Increased incidence of leptospiral infection indicates the dif- pathogenesis to adapt to host’s environment. To date, there are a lot of
ficulty of diagnosing and treating the disease and thus, a better un- studies documented the Leptospira virulence factors (Ko et al., 2009;
derstanding of leptospiral infection followed by the development of Adler et al., 2011; Narayanavari et al., 2012; Wang et al., 2012) in-
sensitive and specific diagnostic methods and appropriate strategies for cluding lipopolysaccharides (LPS) (outer membrane), outer membrane
leptospirosis treatment are beneficial. proteins (OMPs) like lipoprotein, hemolysins, OmpA-like Loa22, lep-
Hitherto, the underlying pathogenesis of leptospirosis remains ob- tospiral immunoglobulin-like (Lig) proteins, sphingomyelinases and
scure. It is believed that both pathogen and host factors are responsible adhesion molecules.
for the development and progression of severe infection. However, it is Leptospiral lipopolysaccharides (LPS) is the major outer membrane
still poorly understood whether the final outcome of severe illness is a of Leptospira, with varying LPS structure in different serovars and an-
result from direct tissue damage caused by the Leptospira, or it is a tigenically diverse (Bulach et al., 2000). Leptospiral LPS is a well-
consequence of an imbalance of host immune response to infection. known virulence factor contributing towards the pathogenesis in
Numerous studies have reported the involvement of immune mediators human, due to its ability to adhere with host’s extracellular matrix,
such as cytokines and chemokines and their potentials as promising including laminin, collagen and fibronectin (Hoke et al., 2008). Lep-
biomarkers for disease severity prediction or therapeutic efficacy. tospiral LPS exhibits similarity to gram-negative-bacterial LPS, both
However, there are still lots of unsolved questions pertinent to this structurally and immunologically. In human cells, leptospiral LPS ac-
disease that need to be addressed by further research. Hence, in this tivates macrophage through a Toll-like receptor 2 (TLR2) pathway, in
review, we focus on the leptospiral pathogenesis and host immune re- the presence of CD14 (Werts et al., 2001). In contrast to mouse cells,
sponse during leptospirosis to gain insights and for better strategized TLR2 and TLR4 pathways were involved, suggesting host-specific ac-
planning in the future to combat this disease. tivation (Nahori et al., 2005). Similarly, both pathogenic and inter-
mediately pathogenic leptospiral LPS confers different carbohydrate
2. Leptospiral pathogenesis and lipid compositions that undergo different leptospiral pathogenesis
(Patra et al., 2015). Other leptospiral outer membrane proteins (OMPs),
Based on 16SrRNA phylogeny, Leptospira genus can be categorized such as lipoproteins LipL32 is only conserved and expressed in patho-
in three large subgroups; Group I (pathogenic), Group II (intermediate genic species during infection (Haake et al., 2000).
pathogenic) and saprophytes (Lehmann et al., 2014). Hitherto, there Hemolysins is a toxin made from protein and lipid that can lyse cell
are nine pathogenic Leptospira species in Group I including L. kirschneri, membrane of erythrocytes and other cells (Thompson and Manktelow,
L. interrogans, L. noguchii, L. borgpetersenii, L. alexendari, L. weilii, L. 1986). Pathogenic Leptospira interrogans can express leptospiral SphH
santarosai, L. kmetyi and L. alstoni. Group I pathogens can be further that destroys mammalian cell membranes by pore formation, without
classified into more than 250 distinct serotypes which produce a wide the involvements of sphingomyelinase or phospholipase activities (Lee
variety of clinical manifestations, ranging from subclinical infection to et al., 2002). However, leptospiral SphA demonstrated sphingomyeli-
severe and fatal disease, with most of the severe disease arise from nase activity on cell membrane by increased membrane permeability,
serovars belonging to evolutionarily-related species, L. interrogans, L. aggregation and fusion (Lee et al., 2002; Goni and Alonso, 2002).
noguchii and L. kirschneri (Brenner et al., 1999; Slack et al., 2009). During Leptospira interrogans infection, hemolysin Sph1, Sph2, Sph3,
Meanwhile, Group II consists of five intermediately pathogenic Leptos- HlpA and TlyA are secreted to stimulate proinflammatory cytokines via
pira species including L. licerasiae, L. wolffii, L. broomii, L. inadai and L. TLR2, TLR4, JNK and NFkB pathways (Wang et al., 2012). Meanwhile,
fainei, where infection with these species will result in mild and self- Leptospiral immunoglobulin-like (Lig) proteins, such as LigA, LigB and
resolving and without fatal complications (Schmid et al., 1986; Brenner LigC are expressed only in pathogenic Leptospira species, which they are
et al., 1999; Petersen et al., 2001; Levett et al., 2006; Matthias et al., anchored to the outer membrane to facilitate mammalian host cell in-
2008; Slack et al., 2008). There are six saprophytic Leptospira species vasion or attachment (Matsunaga et al., 2003). Exposure to physiolo-
including L. meyeri, L. terpstrae, L. biflexa, L. vanthielii, L. yanagawae and gical osmolarity leads to high expression of LigA and LigB that interacts
L. wobachii, in which these species are free-living microorganisms in with extracellular matrix proteins, such as collagen I and IV, laminin,
environment and do not cause any complications in human (Brenner fibronectin and fibrinogen that may be involved in the colonization and
et al., 1999). In addition, a new non-pathogenic saphroyte, L. idonii has dissemination of leptospirosis (Choy et al., 2007).
been discovered and described recently (Saito et al., 2013). A recent study on Leptospira genome revealed that approximately
Before leptospiral genome era, the underlying mechanisms on the 900 genes in pathogenic Leptospira strains could be involved in the
pathogenesis of leptospira remains largely unknown (Ko et al., 2009). pathogenicity of leptospirosis. Moreover, the roles of most of the genes
With the advancement of next generation sequencing (NGS), the first remain obscure and current known proteins failed to explain the viru-
genome sequence of Leptospira interrogans was completed on 2003 (Ren lence mechanisms of leptospira as a whole. Furthermore, mutation
et al., 2003). In-depth analysis on a series of genes responsible for analysis has revealed that some of these genes have pronounced func-
chemotaxis and lipopolysaccharide (LPS) synthesis has revealed the tions in leptospira pathogenesis where mutations in these genes reduce
unique physiological and pathogenic features of this species con- virulence of leptospira. These genes include Loa22, OmpA-family pro-
tributing towards pathogenesis in human. Furthermore, Leptospira tein and several other proteins (Adler et al., 2011).
genome project which was initiated on 2011 has enhanced our under- The latest breakthrough on pathogenesis of Leptospira was demon-
standing on the pathogenesis mechanisms of Leptospira and aided us in strated by Fouts et al. (2016) where the authors performed comparative
distinguishing Leptospira species in terms of gene and pathway in- cross-species genomic analysis of the Leptospira genus to identify the
volvements (Ricaldi et al., 2012). virulence factors associated with the pathogenesis and mammalian host
Establishment of human leptospirosis lies on the ability of patho- adaptation during leptospirosis. In the study, the authors have identi-
genic Leptospira to invade, colonise, disseminate and extract host nu- fied few metabolic mechanisms related to host adaptation by Leptospira,
trients for survival. The persistency in host invasion, colonisation and including pathogen-specific porphyrin metabolism, sialic acid bio-
dissemination is assisted by the expression of various virulence factors synthesis and cobalamin (B12) autotrophy as a Leptospira virulence
by pathogenic Leptospira. The severity of leptospirosis can be de- factor. Besides that, the authors also identified some novel Leptospira
termined by different virulence factors related to pathogenic Leptospira virulence proteins including pathogen-specific adhesins and Virulence
(Thaipadungpanit et al., 2007), inoculum size that modifies infecting Modifying (VM) proteins. The authors also suggested that CRISPR/Cas
pathogen burden (Ganoza et al., 2006) or differences in host immune system, which is only present in pathogenic Leptospira, could be the
response (Wagenaar et al., 2009a). Different Leptospira serotypes confer underlying causes for Leptospira genus refractoriness to gene targeting
different virulence factors that play important roles in leptospiral (Fouts et al., 2016).

109
C. VK et al. Microbiological Research 207 (2018) 108–115

3. Biomarkers during severe leptospirosis (Lowanitchapat et al., 2010). However, another study by Volz et al.
(2015) reported that sufficient amount of IL-10 can confer protection to
Over the past few decades, studies on host immune response and the host by limiting tissue damage induced by inflammatory cytokines and
search for potential biomarkers during human leptospirosis in both prevent severe symptomatic infection (Volz et al., 2015). This warrants
animals and human have expanded tremendously. Identifying suitable further investigations in future to elucidate the role of IL-10 in the
biomarker during human leptospirosis is indeed important in aiding us pathological events during leptospirosis.
to differentiate the severity of the infection for diagnostic and prog- On the other hand, conflicting data on the IL-10/TNF-α ratio as a
nostic applications. Hence, in this review, we summarized the past and marker for leptospirosis severity have been published. High ratio of IL-
present studies on host immune response during human leptospirosis 10 to TNF-α was linked with lower disease severity (Tajiki et al., 1997)
and the relevancy of these biomarkers in severe leptospirosis context. while studies from Reis et al. (2013) and Kyriakidis et al. (2011) sug-
gested that high ratio of IL-10 to TNF-α was associated with severe or
3.1. Cytokines and chemokines fatal outcome. The latest study by Chirathaworn et al. (2016) reported
that there was no significant difference in the IL-10/TNF-α ratio in
Tumor necrosis factor-alpha (TNF-α) is considered as one of the leptospirosis patients with or without organ involvement
widely studied cytokine and is a prominent mediator for severe lep- (Chirathaworn et al., 2016).
tospirosis. TNF-α is a pro-inflammatory cytokine produced by mono- Another cytokine involved in disease progression is interleukin 6
cytes, macrophages and resident renal cells that plays crucial roles in (IL-6). IL-6 is an independent predictor of death in leptospirosis as
various biological events including vasodilatation, tissue development suggested by Reis et al. (2013). IL-6 possesses both pro-inflammatory
and death, cell differentiation and immune-mediated diseases and anti-inflammatory properties. It stimulates hypothalamus and in-
(Balamayooran et al., 2010). Earliest findings from Estavoyer et al. hibits IL-1 and TNF-α (Papa and Kotrotsiou, 2015). High level of IL-6
(1991) revealed that TNF-α was involved with renal impartment and was often seen in patients with severe leptospirosis and fatal cases
high TNF-α levels were prominent in severe non-fatal leptospirosis (Wagenaar et al., 2009a; Reis et al., 2013). Moreover, it is often asso-
(Estavoyer et al., 1991). Following study from Tajiki and Salomao ciated with severe pulmonary haemorrhage syndrome (SPHS), one of
(1996) also revealed that TNF-α expression was associated with liver, the fatal outcomes in severe leptospirosis (Reis et al., 2013). In addi-
kidney and lungs involvement as well as bleeding. In addition, high tion, Papa and Kotrotsiou (2015) reported that IL-6 levels were elevated
level of TNF-α was associated with higher mortality seen in patients at the early stages during severe cases, returning to normal levels at late
suffering from severe leptospirosis (Tajiki and Salomao, 1996). Similar stages. Therefore, these findings suggest that overproduction of IL-6
findings from Kyriakidis et al. (2011) also reported high level of TNF-α could lead to exacerbation of disease and become potentially fatal. This
was associated with pulmonary haemorrhage (Kyriakidis et al., 2011). indicates that IL-6 might have direct pathophysiological effect on the
This haemorrhaging condition could be due to secretion of exotoxin- severe pulmonary haemorrhage syndrome. In a study by Parsons et al.
haemolysins by Leptospira (Wang et al., 2012) which induces the pro- (2005), the authors observed increased levels of interleukin 8 (IL-8) and
duction of TNF-α that acts on both endothelium and macrophages, IL-6 were associated with increased risk of death in patients with acute
causing inflammation of blood vessels, cell and fluid leakage and ulti- respiratory distress syndrome (Parsons et al., 2005). Ahuja et al. (2012)
mately leads to severe haemorrhages seen in patients with Weil’s syn- demonstrated that in a mouse model of acute lung injury, IL-6 may have
drome (Diament et al., 2002). Recent studies also showed TNF-α was direct role in increasing lung inflammation and resulting in poor out-
significantly higher in severe leptospirosis as compared to the mild come (Ahuja et al., 2012).
leptospirosis (Chirathaworn et al., 2016; Reis et al., 2013). The study by Reis et al. (2013) has provided us with new insights on
However, in contrast to the above findings, Rizvi et al. (2011) re- host immunity during severe leptospirosis. In the study, the authors
ported that TNF-α level was greatly suppressed in most of the leptos- suggested ‘cytokine storm’ which comprises of both pro- and anti-in-
pira-positive patients with acute liver hepatitis (Rizvi et al., 2011, flammatory cytokines (IL-2, IL-1b, IL-4, IL-6, IL-17A, IL-8, TNF-α and
2014) and there were no significant differences in TNF-α level between IL-10) were significantly higher in severe cases compared to mild cases.
the mild and severe group of patients with leptospirosis (Chirathaworn In addition, the authors reported high levels of IL-6, IL-8 and IL-10 that
et al., 2016; Mikulski et al., 2015). Possible explanations on the dis- were observed in fatal cases compared to non-fatal cases during severe
crepancies between these findings could be due to the measurement of leptospirosis. Moreover, through the study, the authors surmised that
TNF-α level at different stages of infection, the involvement of multiple specific cytokine responses were varied with clinical outcomes, in-
organs dysfunction and the number of sample size studied. cluding the need of hospitalization, development of severe pulmonary
Interleukin 10 (IL-10) is another notable cytokine involved in the haemorrhage syndrome and death. However, no robust conclusion can
leptospirosis severity. IL-10 is an anti-inflammatory cytokine and has be drawn from this study regarding the role of ‘cytokine storm’ as
multiple roles in mediating immune response including regulation of mediators of pathogenesis during leptospirosis or they are just bio-
inflammation in sepsis (Standiford, 2000), down-regulating the ex- markers of disease severity or progression. The authors also suggested
pression of monocyte-derived interleukin 1 (IL-1) and TNF-α (Cassatella that investigation on temporal evolution of the cytokine response
et al., 1994; Bonfield et al., 1995), inhibits nuclear factor kappa B (NF- should be carried out to further elucidate the role of cytokines in the
kB), regulating nitric oxide synthesis and down-regulation of TNF-α pathogenesis of severe leptospirosis.
receptors after lipopolysaccharide (LPS) stimulation (Wang et al., By using suspension array system, Papa and Kotrotsiou (2015) re-
1995). IL-10 level is one of the independent determinant in fatal lep- ported clusters of cytokines, chemokines and colony-stimulating factors
tospirosis (Reis et al., 2013). Moreover, high fatality was observed in (CSF) were elicited during acute phase of leptospirosis. In addition, the
severe leptospirosis patients with high levels of IL-10 and elevated IL-10 authors speculate some of these cytokines have specific association with
levels were observed in severe leptospirosis patients than in those with sampling time, the presence of haemorrhagic manifestations and the
mild disease (Kyriakidis et al., 2011; Reis et al., 2013). Overall, these pulmonary involvement during leptospirosis. These cytokines include
findings indicated that IL-10 is a severe mediator during leptospirosis, IL-8, IL-6, granulocyte-macrophage CSF (GM-CSF), vascular endothelial
however, the immunopathogenic role of IL-10 during severe leptos- growth factor (VEGF), monocyte chemoattractant protein-1 (MCP-1)
pirosis remain unclear. It is postulated that overproduction of IL-10 and interferon-inducible protein 10 (IP-10) where MCP-1, IP-10 and IL-
may inhibit Th1 response which causes ineffective inflammatory re- 6 levels were increased in most of the cases. On the other hand, the
sponse which subsequently leads to immune pathology (Haake and authors reported that although elevated MCP-1 levels were observed in
Levett, 2015). This postulation is supported by animal study where IL- both mild and severe patients, however, only the severe cases showed
10 increased the mortality of animals infected with leptospires significantly higher levels of MCP-1 as compared to the control group.

110
C. VK et al. Microbiological Research 207 (2018) 108–115

Findings from animal study also showed that increased in the MCP-1 3.2. Serum mediators as potential biomarkers during severe leptospirosis
expression was well associated with the severity of the disease (da Silva
et al., 2009). Meanwhile, VEGF levels were significantly higher in se- A majority of the cytokines and chemokines studied during leptos-
vere as compared to non-severe cases. The similar findings also ob- pirosis are also implicated in other bacterial infections, making it dif-
served in hamsters infected with Leptospira interrogans (Coutinho et al., ficult to project them as biomarkers during leptospirosis. Hence, in this
2014) and patients with Hantavirus infection, suggesting the role of review, we also focus on the potential of serum mediators as biomarkers
VEGF in vascular damage repair (Tsergouli and Papa, 2013). during severe leptospirosis, including sST2, long pentraxin, nitric oxide
Based on the transcriptomic analysis of severe leptospirosis cases, and copeptin as these serum mediators are not implicated much in
Lindow et al. (2016) concluded that the failure of host immune system other bacterial infections and could serve as better biomarkers during
to mount effective innate and adaptive immune response against lep- leptospirosis.
tospires will result in the mortality seen in patients with severe lep-
tospirosis. In addition, through the study, a few unique biomarkers 3.2.1. Nitric oxide
including RANTES and CH3L1, could serve as potential diagnostic and Endothelial dysfunction is one of the possible mechanisms partici-
therapeutic candidates for leptospirosis patients at high risk of dying. pating in the pathogenesis of severe leptospirosis. Endothelial dys-
The function of RANTES as a chemokine is to recruit immune cells to function leads to vascular damage in affected organs and could result in
the infection site. This suggests that aberrant cell trafficking will lead to organ damage (De Brito et al., 1979; Nicodemo et al., 1997). Nitric
poor and slower adaptive immune adaptive immune responses parti- oxide (NO) is a crucial mediator of endothelial dysfunction in sepsis and
cularly in the fatal cases (Lindow et al., 2016). Pro-inflammatory cy- there are similarities in the clinical spectrum of severe sepsis and severe
tokines will induce CHI3L1 expression which is normally associated leptospirosis. Pro-inflammatory cytokines including TNF-α, IL-6 and IL-
with increased patient mortality in sepsis and other infectious or in- 1 can stimulate the expression of inducible endothelial nitric oxide
flammatory diseases (Prakash et al., 2013). Lindow and her colleagues synthase (iNOS) and in turn increased levels of NO in sepsis (Cohen,
has even suggested that CHI3L1 could well represent a new potential 2002). In leptospirosis, production of pro-inflammatory cytokines can
prognostic and therapeutic strategy for leptospirosis (Lindow et al., increase NO level as well (Yang and Hsu, 2005). The clear picture of NO
2016). Hence, both CHI3L1 and RANTES serum levels are potential in the pathogenesis of leptospirosis remains elusive.
candidates for diagnostic markers, which could help identify patients at A study by Prêtre et al. (2011) has shown that expression of in-
risk for developing severe leptospirosis (Lindow et al., 2016). ducible endothelial iNOS is important in eliminating the leptospires
In contrast to most of the findings published, Mikulski et al. (2015) where faster functional deterioration was observed in animals given
reported there was no significance in most of the cytokines studied with 4-aminopyridine, which is an iNOS inhibitor than the untreated
during severe and mild leptospirosis (Mikulski et al., 2015). In another animals (Prêtre et al., 2011). Meanwhile, several studies have docu-
study by Chirathaworn et al. (2016), where the authors compared the mented the involvement of NO or serum nitrate during severe leptos-
cytokine levels between two groups, with or without organ involvement pirosis (Maciel et al., 2006; Avdeeva et al., 2008; Gunaratna et al.,
in two stages, acute and convalescent. IL-6, IL-10 and IL-8 levels in 2012; Kalugalage et al., 2013). These studies indicate high levels of NO
acute stage were significantly higher in organ involvement group than and serum nitrite that were observed in patients with severe leptos-
in without organ involvement group, however, only IL-8 level was pirosis as well as leading to severe pathological condition. Another
significant higher with organ involvement when comparing the cyto- possible explanation is NO can drive oxidative stress, which is likely to
kine levels in convalescent stage (Chirathaworn et al., 2016). Taken be involved in tissue and organ damage in leptospirosis, although
together, these findings raise particular concerns. Firstly, the im- current evidence is still lacking.
portance of sampling time when considering cytokine levels as bio-
markers for leptospirosis severity. Second, the possibilities of an un- 3.2.2. Soluble serum stimulation-2
known dilution of serum due to patients who were treated with Soluble serum stimulation-2 (sST2) is a receptor presented in so-
antimicrobial therapy or massive fluid infusion may interfere with the luble secreted form (sST2) (Tominaga, 1989) or in a membrane-an-
cytokine levels measured. Therefore, in future, investigation of cytokine chored form (ST2L) (Yanagisawa et al., 1993). They are members of the
levels should be carried out in larger groups of patients and with dif- IL-1 receptor family. sST2 is imperative mediators in stimulating host
ferent sampling intervals as this will enable us to capture the most inflammatory response. Increased secretion of sST2 often involved pa-
suitable timing for specimen collection and to determine the most ap- tients with inflammatory disorders linked with abnormal Th2 mediated
propriate cytokine as biomarker in disease monitoring. responses, including, autoimmune diseases (Kuroiwa et al., 2001),
Taken together, the severity of leptospiral infection could be due to asthma (Oshikawa et al., 2001), idiopathic pulmonary fibrosis (Tajima
deranged immune response, especially innate immunity to mount spe- et al., 2003), and in patients with sepsis (Brunner et al., 2004).
cific innate immune responses against leptospires, which leads to in- An important discovery from Wagenaar et al. (2009a) is the asso-
effective early control of infection and followed by diminished activa- ciation of sST2 with bleeding during severe leptospirosis. In the study,
tion of adaptive immune responses. As a result, leptospires keep the authors demonstrated that sST2, IL-6, IL-10 and IL-8 levels were
proliferating and grow in host tissues which subsequently promotes increased upon patients’ admission, with sST2 levels well correlated
host systemic inflammation, followed by tissue damage and mortality with IL-6, IL-8, and IL-10. Furthermore, the authors showed that soluble
seen in leptospirosis patients. Numerous immune mediators including ST2 levels were significantly associated with mortality, severity and
cytokines are involved in either exacerbation or resolving leptospiral overall bleeding in patients. The association of sST2 with bleeding seen
infection. Certain studies have postulated that cytokine patterns elicited in severe leptospirosis could be due to disruption of the endothelial cell
may be differing depends on different clinical outcomes, disease se- barrier, which consequently leads to haemorrhages and exposure of
verity and sampling time which could serve as potential predictors in underlying fibroblasts and secretion of sST2 (Wagenaar et. al., 2009a).
disease monitoring or immunotherapeutic targets. However, more in-
vestigations are warranted to investigate the actual role of cytokines in 3.2.3. Long pentraxin
the pathogenesis of leptospirosis, either as predictors of severity of Pentraxins are a superfamily of multifunctional conserved proteins
disease or it is just a reflective of immune activation in general. The that are characterized by a pentameric structure. They are usually in-
relationship between excessive inflammatory response and the severity volved in acute immunological reactions and often known as acute
of leptospirosis is therefore of utmost importance and ought to be fur- phase proteins (Wagenaar et. al., 2009b). Long pentraxin (PTX3) is
ther investigated. secreted by a variety of cells such as dendritic cells, mononuclear
phagocytes, dendritic cells, epithelial and endothelial cells (Jumat and

111
C. VK et al. Microbiological Research 207 (2018) 108–115

John, 2017). Wagenaar et al. (2009b) demonstrated that high levels of recent study by Chagan-Yasutan et al. (2016) has unravelled the po-
long pentraxin, IL-6 and IL-8 in patients with severe leptospirosis were tential of defensins as potential biomarker during kidney injury in
associated with mortality. In addition, PTX3 was well correlated with leptospirosis patients where the authors suggest that urine defensin α1
IL-8 and to a lesser extent with C-reactive protein (CRP) and IL-6 levels. (uDA1) level may reflect kidney injury in leptospirosis patients due to
The authors further revealed that high PTX3 levels were significantly their correlation with the serum creatinine level (Chagan-Yasutan et al.,
linked with disease severity, suggesting that PTX3 could be used as an 2016).
indicator for monitoring the severity of leptospirosis and to predict the
outcome of the infection (Wagenaar et. al., 2009b). However, larger 5. Proteomic approaches in identifying potential biomarkers
cohort studies inclusive of mild, moderate and severely ill patients during human leptospirosis
should be adopted to evaluate the efficacy of PTX3 as biomarker in
leptospirosis. Host transcriptomic and proteomic studies are deemed to be im-
portant in both the septicemic and immune phases of leptospirosis in
3.2.4. Copeptin patients. However, proteomic study on the host-leptospira interaction is
Hyponatraemia is one of the common clinical manifestations seen in still in its infancy. Srivastava et al. (2012) has first reported on the
severe leptospirosis with renal impairment (Gancheva, 2012). Hypo- alterations of human proteomic profiles induced by Leptospira. The
natraemia arises from dysregulated vasopressin release (Rai et al., authors performed comparative proteomic analysis of leptospirosis
2006). Copeptin is a glycopeptide of the C-terminal part of the arginine patients with the aid of two dimensional gel electrophoresis (2DE) and
vasopressin (AVP) precursor which can be utilised to measure arginine 2D-fluorescence difference gel electrophoresis (2D-DIGE) as well as
vasopressin (AVP) release (Struck et al., 2005; Morgenthaler et al., MALDI-TOF/TOF mass spectrometer. From the findings, the authors
2006). Limper et al. (2010) surmised that copeptin could be a great documented that 22 serum proteins were differentially expressed in
marker in predicting the survivals of patients suffer from sepsis or se- leptospirosis patients as compared to healthy controls, with 8 candidate
vere sepsis arises from leptospirosis. The author reported that copeptin proteins showing different patterns when compared with febrile con-
levels were reduced during the course of the infection in survivors, and trols. From the study, the authors deduced that these differential ex-
its value was returned to baseline within 7 days. Meanwhile, higher pressed proteins are important mediators in physiological pathways,
copeptin levels were associated with disease severity. Furthermore, the such as complement, acute phase response, hemostasis and coagulation
author concluded that, unlike other biomarkers like C-reactive protein cascades. In addition, among these differential expressed proteins, the
(CRP) or procalcitonin (PCT), copeptin level seems to be stable and authors have identified several putative proteins viz α-1-antitrypsin,
does not vary much throughout the progression of leptospirosis. Hence, ceruloplasmin, vitronectin, apolipoprotein A-IV and G-protein signaling
it is a suitable candidate to discriminate between survival and patients regulator which could serve as potential diagnostic or prognostic can-
at high risk of dying. Besides that, copeptin was used as a marker to didates for leptospirosis.
monitor patients with leptospirosis during treatment. It is rather useful On the other hand, Ting et al. (2017) performed proteomic analysis
especially in endemic areas with limited resources as copeptin may during mild and severe leptospirosis through 2DE analysis in combi-
serve as additional tool for health practitioners to decide on the treat- nation with LC–MS/MS. The authors discovered five putative proteins
ment in patients admitted with leptospirosis (Limper et al., 2010). viz apolipoprotein A-I (APOA-I), transferrin (TF), serum amyloid A
(SAA), transthyretin (TTR) and haptoglobin (HP) were differentially
4. Cathelicidin and defensins as potential therapeutic agents expressed between mild and severe leptospirosis. With the aided of
during human leptospirosis Ingenuity® Pathway Analysis software, the authors further speculated
that acute-phase signaling pathway was involved in modulating the
Cathelicidin and defensins belong to large group of cationic anti- expression of these proteins as well as functional network of lipid me-
microbial peptides with amphipathic properties. They are conserved tabolism, small molecule biochemistry and molecular transport. The
component in the innate immune system in all organisms, including authors further suggested that these serum proteins could be potential
animals, plants together with human and exhibit antimicrobial activity biomarkers in differentiating between mild and severe leptospirosis
against many bacteria, fungi, and enveloped viruses (Sambri et al., (Ting et al., 2017).
2002; Isogai et al., 2004; Linde et al., 2013). Cathelicidin plays crucial Further comprehensive investigation into the proteome dynamics of
roles in stimulating phagocytosis, activating neutrophils, reducing LPS- leptospirosis infected patients should take into account of proteome
driven TLR-dependent proinflammatory responses and diminishing patterns that may be affected by the outcome and complications of the
apoptosis (Linde et al., 2013). In leptospirosis context, Lindow et al. patients, duration of the infection and also serogroup type of lepto-
(2016) reported that low serum levels of cathelicidin (LL-37) observed pirosis infected patients.
in fatal cases is responsible for high leptospires loads and therefore
suggests that cathelicidin is a potential therapeutic for leptospirosis. 6. Variation of host genetic
This is due to the involvement of cathelicidin in killing of leptospires in
vitro (Sambri et al., 2002) and decreased circulating cathelicidin will The study on host immune responses and in search of potential
contribute to higher bacterial loads and greater tissue damage, which biomarkers during leptospirosis remains challenging. Variation in host
ultimately result in poor outcome of the infection. The authors further genetic makeup may lead to differences in susceptibility towards lep-
tested the efficacy of LL-37 (the only human cathelicidin) in a hamster tospirosis. Fialho et al. (2009) investigated the association of poly-
model of lethal leptospirosis and reported that administration of LL-37 morphisms in human leukocyte antigen (HLA), cytokine genes and
can significantly protect hamsters from lethal infection by controlling killer-cell immunoglobulin-like receptors (KIR) between patients with
the bacteria loads. Hence, it is postulated that cathelicidin could serve past history of leptospirosis and healthy controls. The authors reported
as potential therapeutic molecule during fatal leptospirosis. that alleles in the HLA-A and B loci plus several HLA haplotypes were
On the other hand, defensins, which shows broad spectrum anti- significantly associated with susceptibility towards leptospiral infec-
microbial activity, is involved in the oxygen-independent microbicidal tion. Polymorphisms in IL-4 and IL-4Rα genes were also significantly
mechanisms of neutrophils. An earliest study from Wu et al. (1992) has higher in patients with past history of leptospirosis. Meanwhile, there
demonstrated the capability of the mixture of human defensins to kill was no significant association between KIR gene profile and leptos-
leptospires in vitro by neutrophils (Wu et al., 1992). However, there is pirosis. On the other hand, Esteves et al. (2014) conducted genetic
no further testing or subsequent study apart from to test the therapeutic susceptibility study in the population of São Miguel Island and reported
efficacy of human defensins in animal nor human studies. Meanwhile, a that genetic polymorphism in the IL12RB1, IL1β and CISH genes were

112
C. VK et al. Microbiological Research 207 (2018) 108–115

susceptible to leptospirosis infection. Recent studies also documented Brenner, D.J., Kaufmann, A.F., Sulzer, K.R., Steigerwalt, A.G., Rogers, F.C., Weyant, R.S.,
the gene polymorphisms in TLR1 Ile602Ser and TLR-2 Arg753Gln 1999. Further determination of DNA relatedness between serogroups and serovars in
the family Leptospiraceae with a proposal for Leptospira alexanderi sp. nov. and four
which have considerable effects on developing severe leptospirosis with new Leptospira genomospecies. Int. J. Syst. Evol. Microbiol. 49 (April 1 (2)),
jaundice and hepatic insufficiency (Cédola et al., 2015). Taken to- 839–858.
gether, these findings highlighted the importance of genetic poly- Brunner, M., Krenn, C., Roth, G., Moser, B., Dworschak, M., Jensen-Jarolim, E., Spittler,
A., Sautner, T., Bonaros, N., Wolner, E., Boltz-Nitulescu, G., Ankersmit, H.J., 2004.
morphisms in the host immune response towards leptospiral infection. Increased levels of soluble ST2 protein and IgG1 production in patients with sepsis
It is still remains elusive to what extent genetic variations contribute and trauma. Intensive Care Med. 30, 1468–1473.
towards different susceptibility of hosts towards leptospiral infection. In Bulach, D.M., Kalambaheti, T., de la Pena-Moctezuma, A., Adler, B., 2000.
Lipopolysaccharide biosynthesis in leptospira. J. Mol. Microbiol. Biotechnol. 2 (4),
addition, there is lack of current evidence to explain why certain in- 375–380.
dividuals are susceptible to severe leptospirosis while others only de- Cassatella, M.A., Meda, L., Gasperini, S., Calzetti, F., Bonora, S., 1994. Interleukin 10 (IL-
velop mild infections. 10) upregulates IL-1 receptor antagonist production from lipopolysaccharide- sti-
mulated human polymorphonuclear leukocytes by delaying mRNA degradation. J.
Exp. Med. 179, 1695–1699.
7. Conclusion Cédola, M., Chiani, Y., Pretre, G., Alberdi, L., Vanasco, B., Gómez, R.M., 2015. Association
of toll-like receptor 2 Arg753Gln and toll-like receptor 1 Ile602Ser single-nucleotide
The host-pathogen interaction remains a very complicated process; polymorphisms with leptospirosis in an Argentine population. Acta Trop. 30 (June
(146)), 73–80.
concerted effort is much needed to fully dissect the different aspects of Chagan-Yasutan, H., Chen, Y., Lacuesta, T.L., Leano, P.S., Iwasaki, H., Hanan, F.,
Leptospira virulence and to unravel its interactions with host immune Taurustiati, D., Ohmoto, Y., Ashino, Y., Saitoh, H., Kiyomoto, H., 2016. Urine levels
system. Understanding the role of immune mediators involved during of defensin α1 reflect kidney injury in leptospirosis patients. Int. J. Mol. Sci. 27
(September 17 (10)), 1637.
leptospirosis is crucial and needs to be highlighted as this will enable us Choy, H.A., Kelley, M.M., Chen, T.L., Moller, A.K., Matsunaga, J., Haake, D.A., 2007.
to elucidate the pathological events that occurs during leptospirosis and Physiological osmotic induction of Leptospira interrogans adhesion: LigA and LigB
also serve as a basis for diagnostic and prognostic applications as well bind extracellular matrix proteins and fibrinogen. Infect. Immun. 75 (5), 2441–2450.
Chirathaworn, C., Supputtamongkol, Y., Lertmaharit, S., Poovorawan, Y., 2016. Cytokine
as vaccine development. Extensive research on leptospirosis should be levels as biomarkers for leptospirosis patients. Cytokine 85, 80–82.
carried out to aid us in attaining more holistic understanding on the Cohen, J., 2002. The immunopathogenesis of sepsis. Nature 420, 885–891.
complexity between Leptospira and humans and for a better design of Costa, F., Hagan, J.E., Calcagno, J., Kane, M., Torgerson, P., Martinez-Silveira, M.S.,
Stein, C., Abela-Ridder, B., Ko, A.I., 2015. Global morbidity and mortality of lep-
improvised therapeutic regimens in future. tospirosis: a systematic review. PLoS Negl. Trop. Dis. 9, e0003898.
Coutinho, M.L., Matsunaga, J., Wang, L.C., de la Peña Moctezuma, A., Lewis, M.S.,
Author contributions Babbitt, J.T., Aleixo, J.A., Haake, D.A., 2014. Kinetics of Leptospira interrogans in-
fection in hamsters after intradermal and subcutaneous challenge. PLoS Negl. Trop. 8,
e3307.
Chin Voon Kin, Lee Tze Yan, Lim Wai Feng and Wan Shahriman, da Silva, J.B., Ramos, T.M., de Franco, M., Palva, D., Martins, E.A., Pereira, M.M., 2009.
YWY wrote the manuscript. Chin Voon Kin, Maha Abdullah, Syafinaz Chemokines expression during Leptospira interrogans serovar Copenhageni infection
Amin Nordin and Zamberi Sekawi conceptualized the idea and revised in resistant BALB/c and susceptible C3H/He. J. Mice. Microb. Pathog. 47, 87–93.
De Brito, T., Bohm, G.M., Yasuda, P.H., 1979. Vascular damage in acute experimental
the manuscript. All authors approved the final manuscript. leptospirosis of the guinea-pig. J. Pathol. 128, 177–182.
Diament, D., Brunialti, M.K., Romero, E.C., Kallas, E.G., Salomao, R., 2002. Peripheral
Conflict of interests blood mononuclear cell activation induced by Leptospira interrogan glycolipopro-
tein. Infect. Immun. 70 (4), 1677–1683.
Estavoyer, J.M., Racadot, E., Couetdic, G., Leroy, J., Grosperrin, L., 1991. Tumor necrosis
The authors declare that there is no conflict of interest that could be factor in patients with leptospirosis. Rev. Infect. Dis. 13 (November 1 (6)).
perceived as prejudicing the impartiality of the review reported. Esteves, L.M., Bulhões, S.M., Branco, C.C., Mota, F.M., Paiva, C., Cabral, R., Vieira, M.L.,
Mota-Vieira, L., 2014. Human leptospirosis: seroreactivity and genetic susceptibility
in the population of São Miguel Island (Azores, Portugal). PLoS One 9, e108534..
Acknowledgement Fialho, R.N., Martins, L., Pinheiro, J.P., Bettencourt, B.F., Couto, A.R., Santos, M.R.,
Peixoto, M.J., Garrett, F., Leal, J., Tomás, A.M., Bruges-Armas, J., 2009. Role of
human leukocyte antigen, killer-cell immunoglobulin-like receptors, and cytokine
We would like to express our appreciation to the management and
gene polymorphisms in leptospirosis. Hum. Immunol. 70, 915–920.
support staffs of the Faculty of Medicine and Health Sciences, Fouts, D.E., Matthias, M.A., Adhikarla, H., Adler, B., Amorim-Santos, L., Berg, D.E.,
University Putra Malaysia and Long Term Research Grant Scheme Bulach, D., Buschiazzo, A., Chang, Y.F., Galloway, R.L., Haake, D.A., 2016. What
makes a bacterial species pathogenic?: Comparative genomic analysis of the genus
(LRGS) by Ministry of Higher Education Malaysia (UPM/700-2/1/
Leptospira. PLoS Negl.Trop. Dis. 10 (February 18 (2)), e0004403.
LRGS/5526400) for providing financial and infrastructure support for Gancheva, G.I., 2012. Renal involvement in leptospirosis. J. IMAB–Ann. Proc. Sci. Papers
literature search. 18 (July 11 (3)), 265–269.
Ganoza, C.A., Matthias, M.A., Collins-Richards, D., Brouwer, K.C., Cunningham, C.B.,
Segura, E.R., Gilman, R.H., Gotuzzo, E., Vinetz, J.M., 2006. Determining risk for
References severe leptospirosis by molecular analysis of environmental surface waters for pa-
thogenic Leptospira. PLoS Med. 3 (August 22 (8)), e308.
Adler, B., Lo, M., Seemann, T., Murray, G.L., 2011. Pathogenesis of leptospirosis: the Ganoza, C.A., Matthias, M.A., Saito, M., Cespedes, M., Gotuzzo, E., Vinetz, J.M., 2010.
influence of genomics. Vet. Microbiol. 153, 73–81. Asymptomatic renal colonization of humans in the peruvian Amazon by Leptospira.
Ahuja, N., Andres-Hernando, A., Altmann, C., Bhargava, R., Bacalja, J., Webb, R.G., He, PLoS Negl. Trop. Dis. 4, e612.
Z., Edelstein, C.L., Faubel, S., 2012. Circulating IL-6 mediates lung injury via CXCL1 Goni, F.M., Alonso, A., 2002. Sphingomyelinases: enzymology and membrane activity.
production after acute kidney injury in mice. Am. J. Physiol. Renal Physiol. 303, FEBS Lett. 531 (1), 38–46.
F864–F872. Gouveia, E.L., Metcalfe, J., de Carvalho, A.L., Aires, T.S., Villasboas-Bisneto, J.C.,
Ashford, D.A., Kaiser, R.M., Spiegel, R.A., Perkins, B.A., Weyant, R.S., Bragg, S.L., Queirroz, A., et al., 2008. Leptospirosis-associated severe pulmonary hemorrhagic
Plikaytis, B., Jarquin, C., De Lose Reyes, J.O., Amador, J.J., 2000. Asymptomatic syndrome, Salvador, Brazil. Emerg. Infect. Dis. 14 (3), 505–508.
infection and risk factors for leptospirosis in Nicaragua. Am. J. Trop. Med. Hyg. 63, Gunaratna, R.I., Handunnetti, S.M., Bulathsinghalage, M.R., Somaratne, P., Jayanaga, A.,
249–254. de Silva, H.J., Rajapakse, S., 2012. Serum nitrite levels in Sri Lankan patients with
Avdeeva, M.G., Bondarenko, I.N., Perediri, I.R., 2008. Clinical significance of the activity leptospirosis. Asian Pac. J. Trop. Med. 5, 75–78.
of nitric oxide synthase in patients with leptospirosis. Klin. Lab Diagn. 1, 40–43. Haake, D.A., Levett, P.N., 2015. Leptospirosis in humans. Leptospira and Leptospirosis.
Balamayooran, G., Batra, S., Fessler, M.B., Happel, K.I., Jeyaseelan, S., 2010. Mechanisms Springer, Berlin Heidelberg, pp. 65–97.
of neutrophil accumulation in the lungs against bacteria. Am. J. Respir. Cell Mol. Haake, D.A., Chao, G., Zuerner, R.L., Barnett, J.K., Barnett, D., Mazel, M., Matsunaga, J.,
Biol. 43, 5–16. Levett, P.N., Bolin, C.A., 2000. The leptospiral major outer membrane protein LipL32
Bharti, A.R., Nally, J.E., Ricaldi, J.N., Matthias, M.A., Diaz, M.M., Lovett, M.A., Levett, is a lipoprotein expressed during mammalian infection. Infect. Immun. 68 (4),
P.N., Gilman, R.H., Willig, M.R., Gotuzzo, E., Vinetz, J.M., 2003. Leptospirosis: a 2276–2285.
zoonotic disease of global importance. Lancet Infect. Dis. 3, 757–771. Hoke, D.E., Egan, S., Cullen, P.A., Adler, B., 2008. LipL32 is an extracellular matrix-
Bonfield, T.L., Konstan, M.W., Burfeind, P., Panuska, J.R., Hilliard, J.B., Berger, M., 1995. interacting protein of Leptospira spp: and pseudoalteromonas tunicata. Infect.
Normal bronchial epithelial cells constitutively produce the anti-inflammatory cy- Immun. 76 (5), 2063–2069.
tokine interleukin-10, which is downregulated in cystic fibrosis. Am. J. Respir. Cell Isogai, E., Hirata, M., Isogai, H., Matsuo, K., Watarai, S., Miura, H., Oguma, K., 2004.
Mol. Biol. 13, 257–261. Antimicrobial and lipopolysaccharide-binding activities of C-terminal domain of

113
C. VK et al. Microbiological Research 207 (2018) 108–115

human CAP18 peptides to genus Leptospira. J. Appl. Res. 4, 180–185. in’signaling. Cell. Signal. 25 (Jul 31 (7)), 1567–1573.
Jumat, M., John, D., 2017. Review on severity markers in leptospirosis. Trop. Biomed. 34 Prêtre, G., Olivera, N., Cédola, M., Haase, S., Alberdi, L., Brihuega, B., Gómez, R.M., 2011.
(September 1 (3)), 494–506. Role of inducible nitric oxide synthase in the pathogenesis of experimental leptos-
Kalugalage, T., Rodrigo, C., Vithanage, T., Somaratne, P., De Silva, H.J., Handunnetti, S., pirosis. Microb. Pathog. 51, 203–208.
Rajapakse, S., 2013. Low serum total nitrite and nitrate levels in severe leptospirosis. Rai, A., Whaley-Connell, A., McFarlane, S., Sowers, J.R., 2006. Hyponatremia arginine
BMC Infect. Dis. 13, 206. vasopressin dysregulation, and vasopressin receptor antagonism. Am. J. Nephrol. 26
Ko, A.I., Goarant, C., Picardeau, M., 2009. Leptospira: the dawn of the molecular genetics (6), 579–589.
era for an emerging zoonotic pathogen. Nat. Rev. Microbiol. 7 (October 1 (10)), Reis, E.A., Hagan, J.E., Ribeiro, G.S., Teixeira-Carvalho, A., Martins-Filho, O.A.,
736–747. Montgomery, R.R., Shaw, A.C., Ko, A.I., Reis, M.G., 2013. Cytokine response sig-
Kuroiwa, K., Arai, T., Okazaki, H., Minota, S., Tominaga, S., 2001. Identification of natures in disease progression and development of severe clinical outcomes for lep-
human ST2 protein in the sera of patients with autoimmune diseases. Biochem. tospirosis. PLoS Negl. Trop. Dis. 7, e2457.
Biophys. Res Commun. 284, 1104–1108. Ren, S.X., Fu, G., Jiang, X.G., Zeng, R., Miao, Y.G., Xu, H., Zhang, Y.X., Xiong, H., Lu, G.,
Kyriakidis, I., Samara, P., Papa, A., 2011. Serum TNF-alpha, sTNFR1,IL-6, IL-8 and IL-10 Lu, L.F., Jiang, H.Q., Jia, J., Tu, Y.F., Jiang, J.X., Gu, W.Y., Zhang, Y.Q., Cai, Z.,
levels in weil’s syndrome. Cytokine 54, 117–120. Sheng, H.H., Yin, H.F., Zhang, Y., Zhu, G.F., Wan, M., Huang, H.L., Qian, Z., Wang,
Lee, S.H., Kim, S., Park, S.C., Kim, M.J., 2002. Cytotoxic activities of Leptospira inter- S.Y., Ma, W., Yao, Z.J., Shen, Y., Qiang, B.Q., Xia, Q.C., Guo, X.K., Danchin, A., Saint
rogans hemolysin SphH as a pore-forming protein on mammalian cells. Infect. girons, I., somerville, R.L., wen, Y.M., shi, M.H., chen, Z., xu, J.G., zhao, G.P., 2003.
Immun. 70 (1), 315–322. Unique physiological and pathogenic features of leptospira interrogans revealed by
Lehmann, J.S., Matthias, M.A., Vinetz, J.M., Fouts, D.E., 2014. Leptospiral pathoge- whole-genome sequencing. Nature 422 (6934), 888–893.
nomics. Pathogens 3, 280–308. Ricaldi, J.N., Fouts, D.E., Selengut, J.D., Harkins, D.M., Patra, K.P., Moreno, A., Lehmann,
Levett, P.N., 2001. Leptospirosis. Clin. Microbiol. Rev. 14, 296–326. J.S., Purushe, J., Sanka, R., Torres, M., Webster, N.J., Vinetz, J.M., Matthias, M.A.,
Levett, P.N., Morey, R.E., Galloway, R.L., Steigerwalt, A.G., 2006. Leptospira broomii sp. 2012. Whole genome analysis of Leptospira licerasiae provides insight into leptos-
nov., isolated from humans with leptospirosis. Int. J. Syst. Evol. Microbiol. 56 (March piral evolution and pathogenicity. PLoS Negl. Trop. Dis. 6 (10), e1853.
1 (3)), 671–673. Rizvi, M., Azam, M., Ajmal, M.R., Shukla, I., Malik, A., 2011. Prevalence ofleptospira in
Limper, M., Goeijenbier, M., Wagenaar, J.F., Gasem, M.H., Isbandrio, B., Kunde, J., acute hepatitis syndrome and assessment ofIL-8 and TNF-alpha level in leptospiral
Hartmann, O., Duits, A.J., Van Gorp, E.C., 2010. Copeptin as a predictor of disease hepatitis. Ann. Trop. Med. Parasitol. 105, 499–506.
severity and survival in leptospirosis. J. Infect. 61, 92–94. Rizvi, M., Azam, M., Sultan, A., Khan, F., Shukla, I., Malik, A., Ajmal, M.R., 2014. Role of
Linde, A., Lushington, G.H., Abello, J., Melgarejo, T., 2013. Clinical relevance of cathe- IL-8, IL-10 and TNF-alpha level in pathogenesis of leptospiral acute hepatitis syn-
licidin in infectious disease. J. Clin. Cell Immuno. S13 (003). drome. Ann. Pathol. Lab. Med. 1, A10–A17.
Lindow, J.C., Wunder E.A.Jr. Popper, S.J., Min, J.N., Mannam, P., Srivastava, A., Yao, Y., Saito, M., Villanueva, S.Y., Kawamura, Y., Iida, K.I., Tomida, J., Kanemaru, T., Kohno, E.,
Hacker, K.P., Raddassi, K., Lee, P.J., Montgomery, R.R., Shaw, A.C., Hagan, J.E., Miyahara, S., Umeda, A., Amako, K., Gloriani, N.G., 2013. Leptospiraidonii sp. nov.,
Araújo, G.C., Nery N.Jr. Relman, D.A., Kim, C.C., Reis, M.G., Ko, A.I., 2016. isolated from environmental water. Int. J. Syst. Evol. Microbiol. 63 (July 1 (7)),
Cathelicidin insufficiency in patients with fatal leptospirosis. PLoS Pathog. 12, 2457–2462.
e1005943. Sambri, V., Marangoni, A., Giacani, L., Gennaro, R., Murgia, R., Cevenini, R., Cinco, M.,
Lowanitchapat, A., Payungporn, S., Sereemaspun, A., Ekpo, P., Phulsuksombati, D., 2002. Comparative in vitro activity of five cathelicidin-derived synthetic peptides
Poovorawan, Y., Chirathaworn, C., 2010 Sep. Expression of TNF-α, TGF-β, IP-10 and against Leptospira, Borrelia and Treponema pallidum. J. Antimicrob. Chemother. 50,
IL-10 mRNA in kidneys of hamsters infected with pathogenic Leptospira. Comp. 895–902.
Immunol. Microbiol. Infect. Dis. 33 (September 30(5)), 423–434. Schmid, G.P., Steere, A.C., Kornblatt, A.N., Kaufmann, A.F., Moss, C.W., Johnson, R.C.,
Maciel, E.A., Athanazio, D.A., Reis, E.A., Cunha, F.Q., Queiroz, A., Almeida, D., McBride, Hovind-Hougen, K., Brenner, D.J., 1986. Newly recognized Leptospira species
A.J., Ko, A.I., Reis, M.G., 2006. High serum nitric oxide levels in patients with severe (Leptospira inadai serovar lyme) isolated from human skin. J. Clin. Microbiol. 24
leptospirosis. Acta Trop. 100, 256–260. (September 1 (3)), 484–486.
Matsunaga, J., Barocchi, M.A., Croda, J., Young, T.A., Sanchez, Y., Siqueira, I., Bolin, Slack, A.T., Kalambaheti, T., Symonds, M.L., Dohnt, M.F., Galloway, R.L., Steigerwalt,
C.A., Reis, M.G., Riley, L.W., Haake, D.A., Ko, A.I., 2003. Pathogenic Leptospira A.G., Chaicumpa, W., Bunyaraksyotin, G., Craig, S., Harrower, B.J., Smythe, L.D.,
species express surface-exposed proteins belonging to the bacterial immunoglobulin 2008. Leptospira wolffii sp. nov., isolated from a human with suspected leptospirosis
superfamily. Mol. Microbiol. 49 (4), 929–945. in Thailand. Int. J. Syst. Evol. Microbiol. 58 (October 1 (10)), 2305–2308.
Matthias, M.A., Ricaldi, J.N., Cespedes, M., Diaz, M.M., Galloway, R.L., Saito, M., Slack, A.T., Khairani-Bejo, S., Symonds, M.L., Dohnt, M.F., Galloway, R.L., Steigerwalt,
Steigerwalt, A.G., Patra, K.P., Ore, C.V., Gotuzzo, E., Gilman, R.H., 2008. Human A.G., Bahaman, A.R., Craig, S., Harrower, B.J., Smythe, L.D., 2009. Leptospira kmetyi
leptospirosis caused by a new, antigenically unique Leptospira associated with a sp. nov., isolated from an environmental source in Malaysia. Int. J. Syst. Evol.
Rattus species reservoir in the Peruvian Amazon. PLoS Negl. Trop. Dis. 2 (April 2(4)), Microbiol. 59 (April 1 (4)), 705–708.
e213. Srivastava, R., Ray, S., Vaibhav, V., Gollapalli, K., Jhaveri, T., Taur, S., Dhali, S., Gogtay,
McBride, A.J., Athanazio, D.A., Reis, M.G., Ko, A.I., 2005. Leptospirosis. Curr. Opin. N., Thatte, U., Srikanth, R., Srivastava, S., 2012. Serum profiling of leptospirosis
Infect. Dis. 18 (5), 376–386. patients to investigate proteomic alterations. J. Proteomics 76, 56–68.
Mikulski, M., Boisier, P., Lacassin, F., Soupé-Gilbert, M.E., Mauron, C., Bruyere-Ostells, L., Standiford, T.J., 2000. Anti-inflammatory cytokines and cytokine antagonists. Curr.
Bonte, D., Barguil, Y., Gourinat, A.C., Matsui, M., Vernel-Pauillac, F., Goarant, C., Pharm. Des. 6, 633–649.
2015. Severity markers in severe leptospirosis: a cohort study. Eur. J. Clin. Microbiol. Struck, J., Morgenthaler, N.G., Bergmann, A., 2005. Copeptin a stable peptide derived
Infect. Dis. 34, 687–695. from the vasopressin precursor, is elevated in serum of sepsis patients. Peptides 12,
Morgenthaler, N.G., Struck, J., Alonso, C., Bergmann, A., 2006. Assay for the measure- 2500–2504.
ment of copeptin, a stable peptide derived from the precursor of vasopressin. Clin. Tajiki, H., Salomao, R., 1996. Association of plasma levels of tumor necrosis factor alpha
Chem. 52 (January 1 (1)), 112–119. with severity of disease and mortality among patients with leptospirosis. Clin. Infect
Nahori, M.A., Fournie-Amazouz, E., Que-Gewirth, N.S., Balloy, V., Chignard, M., Raetz, Dis. 23, 1177–1178.
C.R., Saint Girons, I., Werts, C., 2005. Differential TLR recognition of leptospiral lipid Tajiki, M.H., Satie, N.A., Salomão, R., 1997. The ratio of plasma levels of IL-10/TNF-alpha
A and lipopolysaccharide in murine and human cells. J. Immunol. 175 (9), and its relationship to disease severity and survival in patients with leptospirosis.
6022–6031. Braz. J. Infect. Dis. 1 (June (3)), 138–141 (an official publication of the Brazilian
Narayanavari, S.A., Sritharan, M., Haake, D.A., Matsunaga, J., 2012. Multiple leptospiral Society of Infectious Diseases).
sphingomyelinases (or are there?). Microbiology 158 (May 1 (5)), 1137–1146. Tajima, S., Oshikawa, K., Tominaga, S., Sugiyama, Y., 2003. The increase in serum soluble
Nicodemo, A.C., Duarte, M.I., Alves, V.A., Takakura, C.F., Santos, R.T., Nicodemo, E.L., ST2 protein upon acute exacerbation of idiopathic pulmonary fibrosis. Chest 124,
1997. Lung lesions in human leptospirosis: microscopic, immunohistochemical, and 1206–1214.
ultrastructural features related to thrombocytopenia. Am. J. Trop. Med. Hyg. 56, Thaipadungpanit, J., Wuthiekanun, V., Chierakul, W., Smythe, L.D., Petkanchanapong,
181–187. W., Limpaiboon, R., Apiwatanaporn, A., Slack, A.T., Suputtamongkol, Y., White, N.J.,
Oshikawa, K., Kuroiwa, K., Tago, K., Iwahana, H., Yanagisawa, K., Ohno, S., Tominaga, Feil, E.J., 2007. A dominant clone of Leptospira interrogans associated with an
S.I., Sugiyama, Y., 2001. Elevated soluble ST2 protein levels in sera of patients with outbreak of human leptospirosis in Thailand. PLoS Negl. Trop. Dis. 1 (October 31(1)),
asthma with an acute exacerbation. Am. J. Respir. Crit. Care Med. 164, 277–281. e56.
Palaniappan, R.U., Ramanujam, S., Chang, Y.F., 2007. Leptospirosis: pathogenesis, im- Thompson, J.C., Manktelow, B.W., 1986. Pathogenesis and red blood cell destruction in
munity, and diagnosis. Curr. Opin. Infect. Dis. 20 (June 1 (3)), 284–292. haemoglobinaemic leptospirosis. J. Comp. Pathol. 96 (5), 529–540.
Papa, A., Kotrotsiou, T., 2015. Cytokines in human leptospirosis. Trans. R. Soc. Trop. Ting, T.X., Amran, F.b., Thayan, R., Ahmad, N., Jaafar, R., Haron, R., Abdullah, R.,
Med. Hyg. 109, 749–754. Shamsuddin, S.R.b., Riffin, N.S.b.Md., Abdul-Rahman, P.S., 2017. Potential serum
Parsons, P.E., Eisner, M.D., Thompson, B.T., Matthay, M.A., Ancukiewicz, M., Bernard, biomarkers associated with mild and severe leptospirosis infection: a cohort study in
G.R., Wheeler, A.P., 2005. Lower tidal volume ventilation and plasma cytokine the Malaysian population. Electrophoresis. http://dx.doi.org/10.1002/elps.
markers of inflammation in patients with acute lung injury. Crit. Care Med. 33, 1–6. 201600471.
Patra, K.P., Choudhury, B., Matthias, M.M., Baga, S., Bandyopadhya, K., Vinetz, J.M., Tominaga, S., 1989. A putative protein of a growth specific cDNA from BALB/c- 3T3 cells
2015. Comparative analysis of lipopolysaccharides of pathogenic and intermediately is highly similar to the extracellular portion of mouse interleukin 1 receptor. FEBS
pathogenic Leptospira species. BMC Microbiol. 15, 244. Lett. 258, 301–304.
Petersen, A.M., Boye, K., Blom, J., Schlichting, P., Krogfelt, K.A., 2001. First isolation of Torgerson, P.R., Hagan, J.E., Costa, F., Calcagno, J., Kane, M., Martinez-Silveira, M.S.,
leptospira fainei serovar hurstbridge from two human patients with Weil's syndrome. Goris, M.G.A., Stein, C., Ko, A.I., Abela-Ridder, B., 2015. Global burden of leptos-
J. Med. Microbiol. 50 (January 1 (1)), 96–100. pirosis: estimated in terms of disability adjusted life years. PLoS Negl. Trop. Dis. 9,
Prakash, M., Bodas, M., Prakash, D., Nawani, N., Khetmalas, M., Mandal, A., Eriksson, C., e0004122.
2013. Diverse pathological implications of YKL-40: answers may lie in ‘outside- Tsergouli, K., Papa, A., 2013. Vascular endothelial growth factor levels in dobrava/

114
C. VK et al. Microbiological Research 207 (2018) 108–115

belgrade virus infections. Viruses 5, 3109–3118. Werts, C., Tapping, R.I., Mathison, J.C., Chuang, T.H., Kravchenko, V., Saint Girons, I.,
Volz, M.S., Moos, V., Allers, K., Luge, E., Mayer-Scholl, A., Nöckler, K., Loddenkemper, C., Haake, D.A., Godowski, P.J., Hayashi, F., Ozinsky, A., Underhill, D.M., Kirschning,
Jansen, A., Schneider, T., 2015. Specific CD4+ T-cell reactivity and cytokine release C.J., Wagner, H., Aderem, A., Tobias, P.S., Ulevitch, R.J., 2001. Leptospiral lipopo-
in different clinical presentations of leptospirosis. Clin. Vaccine Immunol. 22 lysaccharide activates cells through a TLR2-dependent mechanism. Nat. Immunol. 2
(December 1 (12)), 1276–1284. (4), 346–352.
Wagenaar, J.F., Gasem, M.H., Goris, M.G., Leeflang, M., Hartskeerl, R.A., van der Poll, T., Wuthiekanun, V., Sirisukkarn, N., Daengsupa, P., Sakaraserane, P., Sangkakam, A.,
van ‘t Veer, C., van Gorp, E.C., 2009a. Soluble ST2 levels are associated with bleeding Chierakul, W., Smythe, L.D., Symonds, M.L., Dohnt, M.F., Slack, A.T., Day, N.P.,
in patients with severe leptospirosis. PLoS Negl. Trop. Dis. 3, e453. 2007. Clinical diagnosis and geographic distribution of leptospirosis, Thailand.
Wagenaar, J.F., Goris, M.G., Gasem, M.H., Isbandrio, B., Moalli, F., Mantovani, A., Boer, Emerg. Infect. Dis. 13 (January (1)), 124.
K.R., Hartskeerl, R.A., Garlanda, C., van Gorp, E.C., 2009b. Long pentraxin PTX3 is Wu, Q., Xu, L., Wang, X., Li, S., Wang, B., 1992. Investigation of microbicidal activity of
associated with mortality and disease severity in severe leptospirosis. J. Infect. 58, neutrophil defensins against leptospires. Hua xi yi ke da xue xue bao. J. West China
425–432. Univ. Med. Sci. 23 (June (2)), 126–129 (Huaxi yike daxue xuebao/[bian ji zhe, Hua xi
Wang, P., Wu, P., Siegel, M.I., Egan, R.W., Billah, M.M., 1995. Interleukin (IL)-10 inhibits yi ke da bao bian wei hui]).
nuclear factor kappa B (NF kappa B) activation in human monocytes.IL-10 and IL-4 Yanagisawa, K., Takagi, T., Tsukamoto, T., Tetsuka, T., Tominaga, S., 1993. Presence of a
suppress cytokine synthesis by different mechanisms. J. Biol. Chem. 270, 9558–9563. novel primary response gene ST2L, encoding a product highly similar to the inter-
Wang, H., Wu, Y., Ojcius, D.M., Yang, X.F., Zhang, C., Ding, S., Lin, X., Yan, J., 2012. leukin 1 receptor type 1. FEBS Lett. 318, 83–87.
Leptospiral hemolysins induce proinflammatory cytokines through Toll-like receptor Yang, G.G., Hsu, Y.H., 2005. Nitric oxide production and immunoglobulin deposition in
2-and 4-mediated JNK and NF-kappaB signaling pathways. PLoS One 7 (8), e42266. leptospiral hemorrhagic respiratory failure. J. Formos. Med. Assoc. 104 (7), 59–763.

115

You might also like