Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

International Reviews of Immunology

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/iiri20

Immunology of leprosy

Luis Alberto Ribeiro Froes Jr , Maria Angela Bianconcini Trindade & Mirian
Nacagami Sotto

To cite this article: Luis Alberto Ribeiro Froes Jr , Maria Angela Bianconcini Trindade & Mirian
Nacagami Sotto (2020): Immunology of leprosy, International Reviews of Immunology, DOI:
10.1080/08830185.2020.1851370

To link to this article: https://doi.org/10.1080/08830185.2020.1851370

Published online: 26 Nov 2020.

Submit your article to this journal

Article views: 7

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iiri20
INTERNATIONAL REVIEWS OF IMMUNOLOGY
https://doi.org/10.1080/08830185.2020.1851370

REVIEW

Immunology of leprosy
Luis Alberto Ribeiro Froes Jra, Maria Angela Bianconcini Trindadea,b, and Mirian Nacagami Sottoa
a
Departamento de Patologia, University of Sao Paulo, S~ao Paulo, Brazil; bImunodermatologia, Universidade de S~ao Paulo Hospital das
Clınicas, S~ao Paulo, Brazil

ABSTRACT ARTICLE HISTORY


Leprosy is a disease caused by Mycobacterium leprae (ML) with diverse clinical manifesta- Received 10 July 2020
tions, which are strongly correlated with the host’s immune response. Skin lesions may be Accepted 9 November 2020
accompanied by peripheral neural damage, leading to sensory and motor losses, as well as
KEYWORDS
deformities of the hands and feet. Both innate and acquired immune responses are
Immunology; leprosy;
involved, but the disease has been classically described along a Th1/Th2 spectrum, where leprosy reactions;
the Th1 pole corresponds to the most limited presentations and the Th2 to the most disse- lymphocyte activation;
minated ones. We discuss this dichotomy in the light of current knowledge of cytokines, Th Mycobacterium leprae
subpopulations and regulatory T cells taking part in each leprosy presentation. Leprosy reac-
tions are associated with an increase in inflammatory activity both in limited and dissemi-
nated presentations, leading to a worsening of previous symptoms or the development of
new symptoms. Despite the efforts of many research groups around the world, there is still
no adequate serological test for diagnosis in endemic areas, hindering the eradication of
leprosy in these regions.

Introduction by hereditary traits, with variable expressiveness in sev-


eral genes demonstrated, according to the clinical pres-
Leprosy is an infectious disease caused by
entation. Of particular relevance, in lepromatous leprosy
Mycobacterium leprae, the main host of which is
and leprosy reactions, is the fact that genes involved in
humans. Notwithstanding the falling incidence, the
the humoral immune response are highly expressed, not-
disease is still an important cause of morbidity, with
ably genes for immunoglobulin receptors or proteins of
208,641 new cases recorded in the world in 2018 [1].
the classical complement pathway [7].
The stigma caused by the disease has changed in
Whole-genome sequencing of Mycobacterium leprae
recent years. The introduction of multidrug treat- collected from different parts of the world has
ment recommended by the WHO allowed a change revealed a low interregional genetic variability, with
in the natural history of the disease, with a signifi- 99.995% of the genetic material identical among differ-
cant increase in cure rates and a substantial drop in ent strains [8]. This is due, in large part, to the fact that
the number of new cases. Despite these advances, the the genome of M. leprae contains a low percentage of
disease still has a high disabling potential, being the functional genes (less than 50%), with a high percentage
main cause of infectious neuropathy in tropical and of pseudogenes [9]. This finding suggests that the vari-
subtropical countries [2]. ability in the clinical presentation of the disease among
The pathophysiology of leprosy is multifactorial, individuals is mainly due to idiosyncratic factors of the
with genetic, immunological and environmental host, rather than to variations specific to the micro-
aspects determining the individual’s susceptibility to organism, which has also been demonstrated by studies
the bacillus [3–5]. Individuals whose skin bacilloscopic matching certain immunogenic variations to specific
index reveals a high concentration of bacilli are called clinical presentations [4,10–12].
multibacillary. These individuals typically develop a It is estimated that more than 95% of infected indi-
weak cellular immune response, unable to contain the viduals are naturally resistant to M. leprae, never devel-
proliferation of M. leprae, with an intense humoral oping any symptoms of the disease [13]. Among
response and high titers of specific serum antibodies symptomatic individuals, the disease manifests itself along
against the bacillus [6]. Susceptibility is also influenced a clinical spectrum with two poles and is classically

CONTACT Luis Alberto Ribeiro Froes lulafroes@usp.br Departamento de Patologia, University of Sao Paulo, S~ao Paulo, Brazil.
ß 2020 Taylor & Francis Group, LLC
2 L. A. R. FROES ET AL.

divided into five different presentations in between these induced high levels of negative regulatory molecules,
poles. Tuberculoid leprosy lies in one of the poles, pre- such as the Monocyte Chemoattractant Protein-1
senting as well-defined annular erythematous plaques (MCP-1) and the Interleukin-1 Receptor Antagonist
and sensitivity loss. The anatomopathological examin- (IL-1RA). In addition, exposure to LPS subsequent to
ation of these lesions is characterized by well-defined exposure to PGL-1 suppressed the monocyte response
granulomas, formed by epithelioid cells, multinucleated to LPS, limiting the secretion of TNF-a, IL-1b and IL-
giant cells and macrophages, surrounded by a ring of 6 [18,19].
lymphocytes, with few or no bacilli inside. On the other The microbicidal activity of macrophages involves
end of the spectrum, individuals with lepromatous lep- the production of reactive oxygen and nitrogen spe-
rosy exhibit intense humoral immune response, abundant cies, through the NADPH-oxidase complex and nitric
production of specific anti-M. leprae antibodies and very oxide, respectively [20], and greater expression of the
weak cellular immune response. Clinically, these individ- enzyme nitric oxide synthase (iNOS) has been
uals present with a higher number of lesions, and no described in skin tuberculoid lesions, in comparison
granulomas on histological examination, but abundant with lepromatous lesions, which might be ascribed to
foamy macrophages full of bacilli – the so-called the higher Th1 response in localized leprosy [21].
Virchow cells [14]. Between the poles lie the borderline Macrophages are generally classified into two
presentations, namely: borderline-tuberculoid, borderline- groups, namely M1 and M2, according to the Th
borderline and borderline-lepromatous. As the clinical response (Th1/Th2). M2 macrophages are associated
presentation moves from the tuberculoid to the leproma- with Th2 responses and be highly present in leproma-
tous pole, a gradual transition occurs from a Th1 to a tous leprosy [22,23]. M2 macrophages from patients
Th2 immune response. with disseminated presentations have shown increased
Finally, the so-called “indeterminate” leprosy is expression of costimulatory molecules (CD68 and
clinically represented solely by a hypochromic macule CD163), Arginase 1 (an enzyme involved in mecha-
with reduced local sensitivity and, on histological nisms regeneration and repairing of tissue) and the
examination, the presence of a lymphohistiocytic infil-
anti-inflammatory cytokines IL-10 and TGF- b [24].
trate of perineural and periadnexal location, which
In addition, growing evidence suggests the existence
can evolve with spontaneous healing or toward any of
of a new subpopulation of macrophages known as
the five classic presentations [14].
M4, originating from M0 macrophages that alter their
behavior in the presence of CXCL4 to produce CD68,
The innate response to M. leprae IL-6, TNF- a, Myeloid-Related Protein-8 (MRP8) and
M. leprae is a gram-positive, acid-fast bacillus and Matrix Metalloproteinases (MMP) 7 and 12 [25–27].
obligate intracellular parasite, exhibiting a pronounced M4 phenotype is more strongly expressed in patients
tropism for macrophages and Schwann cells [15,16]. with lepromatous leprosy, suggesting that this subpo-
The leprosy bacillus is distinguished from other gram- pulation is less effective in eliminating the bacillus,
positive and gram-negative bacteria by its lipid-rich favoring disseminated presentations [28].
cell wall, in particular mycolic acids and the phenolic IL-37 expression in the skin of leprosy patients was
glycolipid-1 (PGL-1). PGL-1 mediates bacillus pene- measured in keratinocytes, endothelial cells, macro-
tration into macrophages by binding to the comple- phages and lymphocytes, revealing higher secretion
ment protein C3, via complement receptors CR1, CR3 than in the controls. In keratinocytes, endothelial cells
and CR4, inducing its phagocytosis [16,17]. It is a and lymphocytes, the level of IL-37 were even higher
component that is central to the pathogenesis of the in lepromatous than in tuberculoid leprosy. IL-37
disease and is involved in the mechanism of phagoly- induces inhibitory mechanisms that negatively regu-
sosome escape [16]. late the production of TNF-a, IL-6 and IL-1b in mac-
Inside the monocyte, PGL-1 plays an immunosup- rophages and T lymphocytes in the dermis [29,30].
pressive role that facilitates its survival in the cell. Hence, the predominance of IL-37 expression in lep-
Analyses of cytokines secreted by “naive” monocytes romatous leprosy might be related to the M2 macro-
exposed to PGL-1 and lipopolysaccharides (LPS) from phages phenotype, since IL-37 has already been
the outer membrane of gram-negative bacteria showed shown to induce differentiation into the M2 pheno-
that PGL-1 alone induced a very weak production of type through production of the CD206, CD86, TGF-b,
inflammatory cytokines, such as tumor necrosis fac- IL-10 and Arginase 1 in studies with M. tubercu-
tor-alpha (TNF-a), IL-1b and IL-10, although it also losis [31].
INTERNATIONAL REVIEWS OF IMMUNOLOGY 3

Curiously, a higher expression of genes related to cell to the pro-inflammatory cytokine TNF-a, via
the phagocytic activity was observed in macrophages induction of the expression of transmembrane TNF-a
in disseminated leprosy, whereas in limited leprosy, and the TNF-a receptor in these cells [38].
genes linked to microbicidal pathways related to vita- The role of the inflammasome in leprosy has also
min D metabolism were more highly expressed [32]. been studied. A component of the innate immune
Immunohistochemistry studies have also shown that response, the inflammasome is a complex of cytosolic
macrophages in disseminated leprosy express CD209 proteins that mediate the inflammatory response
– a scavenger receptor with an affinity for both LDL through molecular patterns associated with pathogens
cholesterol and M. leprae, thus reinforcing the belief (PAMPs) and molecular patterns associated with dam-
that there is indeed more intense phagocytic activity age (DAMPs), the latter being responsible for the mat-
in lepromatous leprosy. The explanation proposed for uration of caspase 1, secretion IL-1b and IL-18, as
that apparent contradiction is that the increase in the well as a type of cell death called pyroptosis [39–41].
phagocytic capacity of macrophages might somehow Greater expression of inflammasome markers in lep-
reduce their microbicidal activity, subsequently limit- romatous lesions has been reported, compared to
ing their ability to control the infection [32]. indeterminate and tuberculoid presentations, pointing
An important role in the immune response to M. to the inefficiency of this protein complex in control-
leprae has been attributed to the Toll-Like Receptors ling the infection [42].
(TLRs) – a type of molecular pattern recognition Certain genetic polymorphisms in the vitamin D
receptor (PRR) found in monocytes and dendritic receptor (VDR) have been associated with limited lep-
cells. TLR 2 and 4 recognize the leprosy bacillus, thus rosy, whereas others have been seen in disseminated
leading to secretion of IL-12 – an interleukin that cases [12]. The VDR receptor is constitutively
induces the production of other pro-inflammatory expressed in macrophages and dendritic cells and can
cytokines and elimination of the bacillus [10,11,33]. induce the secretion of IL-1 and cathelicidin (an anti-
One genetic variation, more strongly associated with microbial peptide), thus meaning that it plays a role
the lepromatous presentation, has been described on in the innate response against some bacteria [12].
the gene coding for TLR2 [34]. A functional genetic Thus, it is reasonable to assume that polymorphisms
study has shown that a loss-of-function mutation at associated with greater activation of this microbicidal
this receptor causes less production of IL-12 by mac- pathway may favor the development of more effective
rophages in response to M. leprae [11], suggesting immune responses against the bacillus, whereas poly-
that eventual genetic polymorphisms in this receptor morphisms that cause less activation of the VDR
might also affect the natural history of the disease. microbicidal pathway should favor the dissemi-
The presence of dendritic cells in tuberculoid and nated disease.
lepromatous lesions was assessed via immunohisto-
chemistry techniques using the CD1a and CD207
The T cell response to M. leprae
markers, revealing a higher number of dendritic cells
in the tuberculoid pole, therefore suggesting that the As a consequence of the Th1/Th2 leprosy paradigm,
presence of such cells favors a more efficient immune the cytokine profile found at each pole differs, with a
response against M. leprae [35]. It has also been greater expression of Th1 cytokines (such as IL-7 and
reported that stimulation of TLR2 in limited presenta- IL-15) in patients with tuberculoid leprosy, and
tions elicited both CD209þ macrophages and greater expression of Th2 cytokines (such as IL-4, IL-
CD1b þ dendritic cells, whereas, in disseminated presen- 5, IL-10 and the transforming growth factor [TGF-b])
tations, TLR2 activation elicited CD209þ macrophages, in patients with disseminated presentations [43,44].
but not CD1b þ dendritic cells. This finding suggests Blood measurements of leprosy patients have also
that a failure in activating CD1b þ dendritic cells might shown that, after stimulation with recombinant anti-
also be at the root of disease dissemination [33]. gens from M. leprae, there is a predominant induction
Also relevant to the response to M. leprae is the of Th1 cytokine secretion (IFN-c, IL-2 and IL-12) in
fact that Schwann cells can process and present anti- limited presentations, and Th2 cytokines (IL-4, IL-5
gens to CD4þ T cells, triggering an inflammatory and IL-6) in disseminated presentations [45].
process that is harmful to Schwann cells, leading to Accordingly, the expression of the transcription fac-
demyelination of peripheral nerves and neural lesions tors involved in Th1 cytokine signaling pathways,
[36,37]. M. leprae also stimulates the inflammatory such as STAT1, STAT4, JAK2 and NFjB, has been
response by increasing the sensitivity of the Schwann found at higher levels in tuberculoid leprosy [46].
4 L. A. R. FROES ET AL.

Beyond the classic Th1/Th2 paradigm, studies have comprise 10% of the CD4þ T cell population in the
shown differences along the spectrum also regarding human peripheral blood [68] and have been shown to
Th9, Th17, Th22 and Treg cells [47–51]. The role of suppress cellular immune responses, both through dir-
Th9 lymphocytes, for example, was assessed in a study ect contact with immune effector cells and through the
that demonstrated higher levels of IL-9 at the tubercu- production of regulatory cytokines, such as TGF-b and
loid pole, suggesting that this cytokine has an antag- IL-10, thus playing an important role in maintaining
onistic function to IL-4 and IL-10, switching the immune self-tolerance and homeostasis [69–75].
immune response from pole Th2 to pole Th1 [52]. In leprosy, Treg cells are more abundantly found in
The potential role of T helper 25 (Th25) cells in lep- patients with lepromatous leprosy, who also display
rosy has also recently been evaluated using monoclo- an increased expression of IL-10 and CTLA-4, thus
nal antibodies against cytokines typical of the Th25 suggesting that these cells may have a pathogenic role
response, such as IL-4, IL-13, IL-25 and IL-17E. These in disseminated presentations [69,76–81]. One single
cytokines, which stimulate a Th2 response, were study, however, has reported a higher frequency of
found in significantly higher concentrations in Tregs in tuberculoid leprosy than in lepromatous lep-
patients with disseminated presentations [53]. rosy [82], making the precise role of such cells still
T Helper 17 cells are similar to Th1 cells in that unclear. It has also been shown that Tregs in local-
they both produce pro-inflammatory cytokines, most ized leprosy produce more IL-17 (proinflammatory
prominently IL-17, and have also been associated with cytokine) and less IL-10 (regulatory cytokine), result-
the development of leprosy reverse reactions [54–56]. ing in a microenvironment that favors inflammation
An increase in the expression of IL-17 in tuberculoid over-regulation and enhances tissue damage [83].
leprosy was also reported, contributing to the recruit- Similarly, a higher concentration of IL-17 in limited
ment of inflammatory cells, activation of endothelial leprosy has also been described, in association with
cells and maintenance of the chronic inflammatory higher expression of TGF-b (regulatory cytokine) in
process [57]. It is believed that the Th17 response may disseminated leprosy [84].
play a critical role in modulation of macrophage activ- Still, with regard to Tregs, an increase in Interleukin
ity, as IL-17 can induce production of TNF-a, IL-6 and 35 (IL-35) has been reported in leprosy patients, dir-
iNOS, leading to the generation of reactive oxygen spe- ectly proportional to the skin bacilloscopy index. IL-
cies (ROS) that should destroy the bacillus [58,59]. 35 is a potent immunosuppressive cytokine secreted
T helper 22 cells (Th22) have been recognized as by Tregs, which functions as an important immuno-
an important subpopulation involved in the immune suppressive factor in immune-mediated diseases,
response to infections. These cells are part of a group inhibiting T cells’ proliferation and production of
of CD4þ T cells that produce isoforms of the IFN- c, TNF-a and IL-2. One of the suppressive
Fibroblast-Growth-Factor (FGF) family and cytokines mechanisms is based on the expression of the inhibi-
such as IL-22, TNF-a, IL-13 and IL-26 [60–64]. tory molecule Programmed cell Death-1 (PD-1) on
Studies have evaluated the existence of IL-22 in lep- the surface of Tregs, which is more evident in disse-
rosy lesions, revealing the presence of this cytokine in minated leprosy [85].
disseminated, rather than limited, presentations A difference has also been described between the lep-
[65,66], even though IL-22 is known to act on macro- rosy poles in terms of the expression of the adhesion mol-
phages stimulating phagolysosomal maturation and ecules E-selectin and P-selectin, as well as the integrins
fusion. The release of IL-22 in these cases might be an VCAM-1, ICAM-1 and VLA-4. Immunohistochemistry
alternative attempt to stimulate macrophage phago- studies have shown a greater expression of these molecules
cytic activity, which is compromised by M. leprae’s in endothelial cells and leukocytes in tuberculoid than in
immune escape mechanisms [65]. Furthermore, the lepromatous leprosy [86,87]. The expression of these mole-
increase in FGF-b in lepromatous leprosy reinforces cules is mediated by Th1 cytokines such as TNF-a, IFN-
the crucial role of the aforementioned growth factor c and IL-1, thus meaning that the higher production of
in the healing response, since this clinical presentation the aforementioned cytokines in tuberculoid leprosy
is associated with a higher number of lesions and should be accountable for the higher expression of selec-
more extensive tissue damage [65,67]. tins in these cases. As a consequence, the deficiency of
Regulatory T cells (Tregs), phenotypically charac- these cytokines observed in lepromatous leprosy should
terized by the expression of CD4, CD25 (interleukin-2 reduce the migration of T cells to the tissues [86,88].
receptor [IL-2R]) and the transcription factor FOXP3, Finally, B-cells are also found in infiltrates of
have been receiving particular attention. They inflammatory leprosy lesions, even though their role is
INTERNATIONAL REVIEWS OF IMMUNOLOGY 5

poorly understood [89]. A study comparing the gene neutrophilic infiltrate. It is not yet clear whether the
expression profile in tuberculoid and lepromatous IC mediates a type III hypersensitivity reaction or
lesions showed that disseminated disease was associ- whether their presence is only an epiphenomenon
ated with increased expression of multiple pathways [101]. Since this reaction occurs more commonly in
and functional groups related to B-cell genes, patients with borderline and lepromatous presenta-
increased frequency of plasma cells (CD138þ), and tions, it is possible that a large number of antigens
increased expression of IL-5 and IgM levels in the and the higher production of antibodies, typical of
lesions, consistent with the fact that lepromatous lep- Th2 responses, may contribute to the formation of
rosy is closer to the Th2 pole [90]. these immune complexes [51,102,103].
ENL is also characterized by the presence of high
levels of TNF-a, both in lesions and in the peripheral
Immunology of leprosy reactions
blood [104], in addition to the cytokines IL-2, IL-4,
Leprosy reactions are episodes of acute hypersensitiv- IL-5, IL-6 and IL-10, typical of Th2 responses [100].
ity presenting as aggravation of the previous lesions Successful use of anti-TNF therapy with infliximab
or the appearance of new ones, occurring before, dur- and etanercept has been reported in three patients
ing or after treatment [91,92]. Most commonly, lep- with ENL, providing additional evidence on the
rosy reactions happen in disseminated presentations inflammatory role of TNF-a in the process
during the first three months of treatment and cur- [96,105,106]. The use of thalidomide in the treatment
rently represent the main complication of the disease, of these patients is also believed to be based on TNF-
requiring immediate treatment to prevent permanent a suppression [58].
neural damage [93]. They can be of two types: reverse CD64 (FccRI), a high-affinity receptor for mono-
reaction (RR) or erythema nodosum leprosum (ENL). meric IgG1 and IgG3, present on the surface of neu-
RR occurs in approximately one-third of patients with trophils, was described in higher levels in ENL
the borderline presentation, whereas ENL occurs in patients than in patients with lepromatous or border-
around 50% of patients with lepromatous and 10% of line presentations, potentially serving as a biomarker
patients with borderline leprosy [51], especially in for ENL and severe disease [107]. It is believed that
those with a skin bacilloscopy index equal to, or the increase in CD64 expression during ENL might be
greater than, 4 þ [94]. induced by cytokines such as IFN-c and GM-CSF
RR is a type IV hypersensitivity reaction, character- [108], or by intracellular antigens from bacilli
ized by an exacerbation of the cellular immune destroyed with treatment [107], consistent with the
response against M. leprae. If, on the one hand, this fact that the incidence of ENL is maximal during
reaction leads to an enhanced elimination of bacilli, treatment when intense bacterial destruction is taking
on the other hand, it causes an exacerbation of the place [109].
inflammatory process and disease symptoms, includ- Finally, T cells also seem to play a role in ENL;
ing neural damage [51,95]. These lesions are charac- indeed, several studies have reported an increase in the
terized by the presence of cellular infiltrate composed CD4/CD8 ratio both in the skin and in the peripheral
predominately of CD4þ T lymphocytes, as well as blood, with a reduction in the ratio being noted when
CD163þ macrophages [96]. The CD4þ T cells found patients are treated, and an increase when disease
in the skin and neural lesions release TNF-a and IFN- relapses [110–114]. ENL is also characterized by a
c, leading to the pain and edema typically observed in Th17 pattern of an immune response, with high
RR [97,98]. A higher expression of cytokines such as expression of the pro-inflammatory cytokine IL-17 and
IL-12, IL-1b and IL-2 has also been described in RR, marked reduction of Tregs [115], thus suggesting that
in addition to the monocyte-1 chemotactic protein suppression of Tregs gives rise to pro-inflammatory
(MCP-1), all associated with Th1 responses [99,100]. responses mediated by Th-17. Indeed, treatment with
It has also been shown that, in RR, similar to tubercu- thalidomide has already been shown to inhibit Th17
loid leprosy, there is an increase in the expression of response [58].
genes related to the production of IFN-c, such as
genes involved in the vitamin D-mediated antimicro-
Complement system in leprosy
bial pathway [19].
In terms of ENL, it is characterized by a systemic The role of complement in leprosy has been assessed
inflammatory process related to an extravascular in the past [116–118]. Complement C3 fraction levels
deposition of immune complexes (IC) and a were found to be reduced in patients with ENL, with
6 L. A. R. FROES ET AL.

no changes in lepromatous control patients. This developed RR, thus suggesting a possible predictable
reduction in C3 provides support to the theory that role for that kind of reaction too [133]. These findings
ENL is an IC-related reaction, similar to diseases such were confirmed by another investigation with 440
as lupus or acute glomerulonephritis, which also pre- patients, which also revealed that a positive PCR for
sent with complement consumption [17,118–120]. M. leprae DNA at the time of diagnosis is a risk factor
Moreover, it has been suggested that there is a hyper- for reverse reactions. In addition, reactions were more
catabolism of complement in ENL since the C3d frequent among patients who, by the end of treat-
metabolite levels were found to increase by 70%, ment, remained positive for anti-PGL-1 serology or
whereas only an 18% increase was seen in uncompli- M. leprae PCR [134], with a 10.4-fold increase in the
cated lepromatous patients [121,122]. risk of reactions after treatment [135].
A Brazilian study with 109 patients analyzed the A large cohort of 753 patients, however, showed
association between leprosy, leprosy reactions and the divergent results. This study did report that reactions
complement proteins BF, C2, C4A and C4B, showing after diagnosis were more commonly seen in individu-
that all patients with C4B deficiency developed ENL als with a positive skin bacilloscopy index prior to
[119,121]. C4 deficiency has also been associated with beginning the treatment, although with no significant
lupus erythematosus, thus suggesting that this factor relation between anti-PGL-1 levels and leprosy reac-
might play a role in the removal of ICs from the cir- tions. Also, statistical analyses in this study did not
culation [121]. Interestingly, another study reported demonstrate significant sensitivity or specificity for
that M. leprae only induced activation of the comple- anti-PGL-1 serology in the diagnosis of leprosy reac-
ment system when its cellular integrity was lost, with tions [136].
intact bacillus unable to activate the complement Furthermore, two antigens called ML0405 and
[123], thus supporting the notion that the release of ML2331, originating from a fusion protein, were
antigens from destroyed bacilli during treatment developed and named “Leprosy IDRI diagnostic-1”
might trigger reactions. (LID-1), with increased sensitivity [125, 137,138].
LID-1 was fused to PGL-1, forming the NDO-LID
molecule, which also showed higher sensitivity
Serological testing
compared to its isolated components [139,140]. A
The main leprosy control strategies are still based on different study assessed anti-PGL-I, anti-LID-1 and
early diagnosis and treatment, to halt its spread and anti-NDO-LID antibodies as predictors of type I
prevent, or, at least minimize, permanent damage. reverse reaction, revealing low positive and negative
However, there is currently no serological analysis predictive values for the reaction. The same study,
that, in isolation, could be adopted as a reliable tool however, suggested the usefulness of anti-LID-1 ser-
for diagnosis of the disease [6,124–127]. In this ology as a predictor of the development of ENL in
regard, some specific antibodies against M. leprae multibacillary patients [102,141].
have been studied as potential markers [6,124–127],
with particular attention paid to the phenolic glycoli-
Conclusion
pid-1 (PGL-1) [128]. Multibacillary subjects produce
large amounts of IgM-specific anti-PGL-1, and this Leprosy’s clinical manifestations are characterized by
production is proportional to bacterial load, although its bipolar spectral presentation, with a strong correl-
with relatively low sensitivity. The average sensitivity ation between the immune system branch (Th1/Th2)
for multibacillary individuals is 78% and for the pau- most prominently activated and the clinical picture.
cibacillary only 23% [6,129,130]. The very low genetic variability found in the M. leprae
The correlation between serum levels of anti-PGL-1 genome strongly suggests that the immunological
IgM antibodies and reaction episodes has also been response depends mostly on the host, rather than on
studied. A comparative analysis between patients with variations in the pathogen. Indeed, some genetic stud-
RR, ENL, uncomplicated leprosy and healthy controls ies have already demonstrated a correlation between
showed a significantly higher amount of anti-PGL-1 certain haplotypes and specific clinical manifestations,
antibodies in ENL patients compared to all the others even though the immunogenetic determinants of the
[131]. Accordingly, another study found serum levels natural history of the disease are still far from being
of anti-PGL-1 IgM and IgG to be low or absent in RR fully known [11,12, 69,70].
patients [132]. Seropositivity for anti-PGL-1 was also The cornerstone of the elimination and control of
higher among borderline patients who subsequently leprosy has primarily consisted of improving access to
INTERNATIONAL REVIEWS OF IMMUNOLOGY 7

health care, combined with a better ability to screen with reversal reaction in leprosy. J Infect Dis. 2008;
the cases and diagnose them early. A study indicated 197(2):253–261. doi:10.1086/524688.
11. Kang TJ, Lee SB, Chae GT. A polymorphism in the
that Brazilian patients begin treatment, on average,
toll-like receptor 2 is associated with IL-12 production
1.5–2 years after the appearance of the first symptoms, from monocyte in lepromatous leprosy. Cytokine.
and the delay is credited, at least in part, to the lim- 2002;20(2):56–62. doi:10.1006/cyto.2002.1982.
ited clinical skills of the health workers [142]. In this 12. Roy S, Frodsham A, Saha B, Hazra SK, Mascie-
regard, comprehension of the immune response to the Taylor CG, Hill AV. Association of vitamin D recep-
disease could offer new opportunities for early diagno- tor genotype with leprosy type. J Infect Dis. 1999;
179(1):187–191. doi:10.1086/314536.
sis and treatment, control of reactions and permanent 13. Scollard DM, Adams LB, Gillis TP, Krahenbuhl JL,
damage, prevention of physical disabilities, and block- Truman RW, Williams DL. The continuing chal-
ing of the transmission of disease – aspects which lenges of leprosy. Clin Microbiol Rev. 2006;19(2):
have been neglected in many parts of the world for 338–381. doi:10.1128/CMR.19.2.338-381.2006.
so long. 14. Ridley DS, Jopling WH. Classification of leprosy
according to immunity. A five-group system. Int J
Lepr Other Mycobact Dis. 1966;34(3):255–273.
Funding 15. Britton WJ, Lockwood DN. Leprosy. Lancet. 2004;
363(9416):1209–1219. doi:10.1016/S0140-6736(04)15952-7.
Fundo de Apoio a Dermatologia de S~ao Paulo – Sebasti~ao 16. Schlesinger LS, Horwitz MA. Phenolic glycolipid-1
Sampaio (FUNADERSP) da Sociedade Brasileira de of Mycobacterium leprae binds complement compo-
Dermatologia. nent C3 in serum and mediates phagocytosis by
human monocytes. J Exp Med. 1991;174(5):
1031–1038. doi:10.1084/jem.174.5.1031.
References 17. Schifferli JA, Morris SM, Dash A, Peters DK.
1. World Health O. Global leprosy update, 2018. mov- Complement-mediated solubilization in patients with
systemic lupus erythematosus, nephritis or vasculitis.
ing towards a leprosyfree world. Weekly epidemio-
Clin Exp Immunol. 1981;46(3):557–564.
logical record (WER). 2019;Nos. 35/36(94):389–412.
18. Manca C, Peixoto B, Malaga W, Guilhot C, Kaplan
2. Cardoso FM, De Freitas MR, Escada TM, Nevares
G. Modulation of the cytokine response in human
MT, Nascimento OJ. Late onset neuropathy in lep-
monocytes by mycobacterium leprae phenolic glyco-
rosy patients released from treatment: not all due to
lipid-1. J Interferon Cytokine Res. 2012;32(1):27–33.
reactions? Lepr Rev. 2013;84(2):128–135.
doi:10.1089/jir.2011.0044.
3. Alcais A, Mira M, Casanova JL, Schurr E, Abel L.
19. Teles RM, Graeber TG, Krutzik SR, Montoya D,
Genetic dissection of immunity in leprosy. Curr
et al. Type I interferon suppresses type II interferon-
Opin Immunol. 2005;17(1):44–48. doi:10.1016/j.coi. triggered human anti-mycobacterial responses.
2004.11.006. Science. 2013;339(6126):1448–1453. doi:10.1126/sci-
4. Mira MT. Genetic host resistance and susceptibility ence.1233665.
to leprosy. Microbes Infect. 2006;8(4):1124–1131. doi: 20. Wynn TA, Chawla A, Pollard JW. Macrophage biol-
10.1016/j.micinf.2005.10.024. ogy in development, homeostasis and disease. Nature.
5. Sauer ME, Salomao H, Ramos GB, D’Espindula HR, 2013;496(7446):445–455. doi:10.1038/nature12034.
et al. Genetics of leprosy: expected-and unexpected- 21. Khanolkar-Young S, Snowdon D, Lockwood DN.
developments and perspectives. Clin Dermatol. 2016; Immunocytochemical localization of inducible nitric
34(1):96–104. doi:10.1016/j.clindermatol.2015.10.005. oxide synthase and transforming growth factor-beta
6. Moura RS, Penna GO, Fujiwara T, et al. Evaluation (TGF-beta) in leprosy lesions. Clin Exp Immunol.
of a rapid serological test for leprosy classification 1998;113(3):438–442. doi:10.1046/j.1365-2249.1998.
using human serum albumin as the antigen carrier. J 00669.x.
Immunol Methods. 2014;412:35–41. doi:10.1016/j.jim. 22. Bobosha K, Wilson L, van Meijgaarden KE, et al. T-
2014.06.014. cell regulation in lepromatous leprosy. PLoS Negl
7. Dupnik KM, Bair TB, Maia AO, et al. Trop Dis. 2014;8(4):e2773. doi:10.1371/journal.pntd.
Transcriptional changes that characterize the 0002773.
immune reactions of leprosy. J Infect Dis. 2015; 23. Moura DF, de Mattos KA, Amadeu TP, et al. CD163
211(10):1658–1676. doi:10.1093/infdis/jiu612. favors Mycobacterium leprae survival and persistence
8. Monot M, Honore N, Garnier T, et al. Comparative gen- by promoting anti-inflammatory pathways in lep-
omic and phylogeographic analysis of Mycobacterium romatous macrophages. Eur J Immunol. 2012;42(11):
leprae. Nat Genet. 2009;41(12):1282–1289. doi:10.1038/ng. 2925–2936. doi:10.1002/eji.201142198.
477. 24. Caldwell RB, Toque HA, Narayanan SP, Caldwell
9. Cole ST, Eiglmeier K, Parkhill J, et al. Massive gene RW. Arginase: an old enzyme with new tricks.
decay in the leprosy bacillus. Nature. 2001;409(6823): Trends Pharmacol Sci. 2015;36(6):395–405. doi:10.
1007–1011. doi:10.1038/35059006. 1016/j.tips.2015.03.006.
10. Bochud PY, Hawn TR, Siddiqui MR, et al. Toll-like 25. Chistiakov DA, Bobryshev YV, Orekhov AN.
receptor 2 (TLR2) polymorphisms are associated Changes in transcriptome of macrophages in
8 L. A. R. FROES ET AL.

atherosclerosis. J Cell Mol Med. 2015;19(6): 39. Amin J, Boche D, Rakic S. What do we know about
1163–1173. doi:10.1111/jcmm.12591. the inflammasome in humans? Brain Pathol. 2017;
26. Butcher MJ, Galkina EV. Phenotypic and functional 27(2):192–204. doi:10.1111/bpa.12479.
heterogeneity of macrophages and dendritic cell sub- 40. Zamboni DS, Lima-Junior DS. Inflammasomes in
sets in the healthy and atherosclerosis-prone aorta. host response to protozoan parasites. Immunol Rev.
Front Physiol. 2012;3:44. doi:10.3389/fphys.2012.00044. 2015;265(1):156–171. doi:10.1111/imr.12291.
27. De Paoli F, Staels B, Chinetti-Gbaguidi G. 41. Broz P, Dixit VM. Inflammasomes: mechanism of
Macrophage phenotypes and their modulation in assembly, regulation and signalling. Nat Rev Immunol.
atherosclerosis. Circ J. 2014;78(8):1775–1781. doi:10. 2016;16(7):407–420. doi:10.1038/nri.2016.58.
1253/circj.cj-14-0621. 42. Silva LM, de Sousa JR, Hirai KE, et al. The inflam-
28. de Sousa JR, Lucena Neto FD, Sotto MN, Quaresma masome in leprosy skin lesions: an immunohisto-
JAS. Immunohistochemical characterization of the chemical evaluation. Infect Drug Resist. 2018;11:
M4 macrophage population in leprosy skin lesions. 2231–2240. doi:10.2147/IDR.S172806.
BMC Infect Dis. 2018;18(1):576. doi:10.1186/s12879- 43. Yamamura M, Uyemura K, Deans RJ, et al. Defining
018-3478-x. protective responses to pathogens: cytokine profiles
29. Charrad R, Berraïes A, Hamdi B, Ammar J, in leprosy lesions. Science. 1991;254(5029):277–279.
Hamzaoui K, Hamzaoui A. Anti-inflammatory activ- doi:10.1126/science.1925582.
ity of IL-37 in asthmatic children: correlation with 44. Bleharski JR, Li H, Meinken C, et al. Use of genetic
inflammatory cytokines TNF-a, IL-b, IL-6 and IL- profiling in leprosy to discriminate clinical forms of
17A. Immunobiology. 2016;221(2):182–187. doi:10. the disease. Science. 2003;301(5639):1527–1530. doi:
1016/j.imbio.2015.09.009. 10.1126/science.1087785.
30. Ding VA, Zhu Z, Mantz AA, et al. The role of IL-37 45. Sampaio LH, Sousa AL, Barcelos MC, Reed SG,
in non-cancerous diseases. Pathol Oncol Res. 2017; Stefani MM, Duthie MS. Evaluation of various cyto-
23(3):463–470. doi:10.1007/s12253-016-0137-7. kines elicited during antigen-specific recall as poten-
31. Huang Z, Gao C, Chi X, et al. IL-37 expression is tial risk indicators for the differential development
upregulated in patients with tuberculosis and induces of leprosy. Eur J Clin Microbiol Infect Dis. 2012;
macrophages towards an M2-like phenotype. Scand J 31(7):1443–1451. doi:10.1007/s10096-011-1462-0.
Immunol. 2015;82(4):370–379. doi:10.1111/sji.12326. 46. Silva LM, Hirai KE, de Sousa JR, et al.
32. Montoya D, Cruz D, Teles RM, et al. Divergence of Immunohistochemical analysis of the expression of
macrophage phagocytic and antimicrobial programs cellular transcription NFjB (p65), AP-1 (c-Fos and c-
in leprosy. Cell Host Microbe. 2009;6(4):343–353. doi: Jun), and JAK/STAT in leprosy . Hum Pathol. 2015;
10.1016/j.chom.2009.09.002. 46(5):746–752. doi:10.1016/j.humpath.2015.01.015.
33. Krutzik SR, Tan B, Li H, et al. TLR activation trig- 47. Modlin RL. Th1-Th2 paradigm: insights from lep-
gers the rapid differentiation of monocytes into mac- rosy. J Invest Dermatol. 1994;102(6):828–832. doi:10.
rophages and dendritic cells. Nat Med. 2005;11(6): 1111/1523-1747.ep12381958.
653–660. doi:10.1038/nm1246. 48. Rendini T, Levis W. Leprosy is teaching us the
34. Kang TJ, Chae GT. Detection of Toll-like receptor 2 immunopathogenesis of inflammatory skin disease. J
(TLR2) mutation in the lepromatous leprosy Allergy Clin Immunol. 2015;136(6):1707. doi:10.1016/
patients. FEMS Immunol Med Microbiol. 2001;31(1): j.jaci.2015.08.029.
53–58. doi:10.1111/j.1574-695X.2001.tb01586.x. 49. Magombedze G, Reddy PB, Eda S, Ganusov VV.
35. Hirai KE, Aarao TL, Silva LM, et al. Langerhans cells Cellular and population plasticity of helper CD4(þ)
(CD1a and CD207), dermal dendrocytes (FXIIIa) T cell responses. Front Physiol. 2013;4:206. doi:10.
and plasmacytoid dendritic cells (CD123) in skin 3389/fphys.2013.00206.
lesions of leprosy patients. Microb Pathog. 2016;91: 50. Zhou W, Zhang F, Aune TM. Either IL-2 or IL-12 is
18–25. doi:10.1016/j.micpath.2015.11.013. sufficient to direct Th1 differentiation by nonobese
36. Spierings E, De Boer T, Zulianello L, Ottenhoff TH. diabetic T cells. J Immunol. 2003;170(2):735–740.
Novel mechanisms in the immunopathogenesis of lep- doi:10.4049/jimmunol.170.2.735.
rosy nerve damage: the role of Schwann cells, T cells 51. Walker SL, Lockwood DN. The clinical and
and Mycobacterium leprae. Immunol Cell Biol. 2000; immunological features of leprosy. Br Med Bull.
78(4):349–355. doi:10.1046/j.1440-1711.2000.00939.x. 2006;77–78:103–121. doi:10.1093/bmb/ldl010.
37. Spierings E, de Boer T, Wieles B, Adams LB, Marani 52. de Sousa JR, Pagliari C, de Almeida DSM, et al. Th9
E, Ottenhoff TH. Mycobacterium leprae-specific, cytokines response and its possible implications in the
HLA class II-restricted killing of human Schwann immunopathogenesis of leprosy. J Clin Pathol. 2017;
cells by CD4þ Th1 cells: a novel immunopathogenic 70(6):521–527. doi:10.1136/jclinpath-2016-204110.
mechanism of nerve damage in leprosy. J Immunol. 53. de Sousa JR, Quaresma JAS. The role of T helper 25
2001;166(10):5883–5888. doi:10.4049/jimmunol.166. (Th25) cells in the immune response to M. leprae. J
10.5883. Am Acad Dermatol. 2017;78(5):1009–1011. doi:10.
38. Andrade PR, Jardim MR, da Silva AC, et al. 1016/j.jaad.2017.11.025.
Inflammatory cytokines are involved in focal demye- 54. Tarique M, Saini C, Naqvi RA, Khanna N, Sharma
lination in Leprosy Neuritis. J Neuropathol Exp A, Rao DN. IL-12 and IL-23 modulate plasticity of
Neurol. 2016;75(3):272–283. doi:10.1093/jnen/nlv027. FoxP3þ regulatory T cells in human Leprosy. Mol
INTERNATIONAL REVIEWS OF IMMUNOLOGY 9

Immunol. 2017;83:72–81. doi:10.1016/j.molimm.2017. 67. de Sousa JR, de Sousa RPM, Aar~ao T. L d S, et al. In
01.008. situ expression of M2 macrophage subpopulation in
55. Amedei A, Munari F, Bella CD, et al. Helicobacter leprosy skin lesions. Acta Trop. 2016;157:108–114.
pylori secreted peptidyl prolyl cis, trans-isomerase doi:10.1016/j.actatropica.2016.01.008.
drives Th17 inflammation in gastric adenocarcin- 68. Sakaguchi S, Powrie F. Emerging challenges in regu-
oma. Intern Emerg Med. 2014;9(3):303–309. doi:10. latory T cell function and biology. Science. 2007;
1007/s11739-012-0867-9. 317(5838):627–629. doi:10.1126/science.1142331.
56. Attia EAS, Abdallah M, El-Khateeb E, et al. Serum 69. Quaresma JAS, Esteves PC, de Sousa Aar~ao TL, de
Th17 cytokines in leprosy: correlation with circulat- Sousa JR, da Silva Pinto D, Fuzii HT. Apoptotic
ing CD4(þ) CD25 (high)FoxP3 (þ) T-regs cells, as activity and Treg cells in tissue lesions of patients
well as down regulatory cytokines. Arch Dermatol with leprosy. Microb Pathog. 2014;76:84–88. doi:10.
Res. 2014;306(9):793–801. doi:10.1007/s00403-014- 1016/j.micpath.2014.07.005.
1486-2. 70. Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T
57. Saini C, Ramesh V, Nath I. CD4þ Th17 cells dis- cells: mechanisms of differentiation and function.
criminate clinical types and constitute a third subset Annu Rev Immunol. 2012;30:531–564. doi:10.1146/
of non Th1, Non Th2 T cells in human leprosy. annurev.immunol.25.022106.141623.
PLoS Negl Trop Dis. 2013;7(7):e2338. doi:10.1371/ 71. Belkaid Y. Regulatory T cells and infection: a dan-
journal.pntd.0002338. gerous necessity. Nat Rev Immunol. 2007;7(11):
58. Martiniuk F, Giovinazzo J, Tan AU, et al. Lessons of 875–888. doi:10.1038/nri2189.
leprosy: the emergence of TH17 cytokines during 72. Shevach EM. Mechanisms of foxp3þ T regulatory
type II reactions (ENL) is teaching us about T-cell cell-mediated suppression. Immunity. 2009;30(5):
plasticity. J Drugs Dermatol. 2012;11(5):626–630. 636–645. doi:10.1016/j.immuni.2009.04.010.
59. Aar~ao TL, de Sousa JR, Botelho BS, Fuzii HT, 73. Workman CJ, Szymczak-Workman AL, Collison LW,
Quaresma JA. Correlation between nerve growth factor Pillai MR, Vignali DA. The development and function
and tissue expression of IL-17 in leprosy. Microb of regulatory T cells. Cell Mol Life Sci. 2009;66(16):
Pathog. 2016;90:64–68. doi:10.1016/j.micpath.2015.11.019. 2603–2622. doi:10.1007/s00018-009-0026-2.
60. Kong Q, Wu W, Yang F, et al. Increased expressions 74. Sakaguchi S, Yamaguchi T, Nomura T, Ono M.
of IL-22 and Th22 cells in the coxsackievirus B3- Regulatory T cells and immune tolerance. Cell. 2008;
Induced mice acute viral myocarditis. Virol J. 2012;9: 133(5):775–787. doi:10.1016/j.cell.2008.05.009.
232. doi:10.1186/1743-422X-9-232. 75. Hori S, Nomura T, Sakaguchi S. Control of regula-
61. Eyerich S, Eyerich K, Pennino D, et al. Th22 cells tory T cell development by the transcription factor
represent a distinct human T cell subset involved in Foxp3. Science. 2003;299(5609):1057–1061. doi:10.
epidermal immunity and remodeling. J Clin Invest. 1126/science.1079490.
2009;119(12):3573–3585. 76. Palermo ML, Pagliari C, Trindade MAB, et al.
62. Furuzawa-Carballeda J, Bostock IC, Lima G, et al. Increased expression of regulatory T cells and down-
Immunophenotyping of peripheral immunoregula- regulatory molecules in lepromatous leprosy. Am J
tory as well as Th17A and Th22 cell subpopulations Trop Med Hyg. 2012;86(5):878–883. doi:10.4269/
in kidney transplant recipients under belatacept or ajtmh.2012.12-0088.
cyclosporine treatment. Transpl Immunol. 2014; 77. Fernandes C, Gonçalves HS, Cabral PB, Pinto HC,
30(2–3):107–113. doi:10.1016/j.trim.2014.02.002. Pinto MIM, C^amara LMC. Increased frequency of
63. Duhen T, Geiger R, Jarrossay D, Lanzavecchia A, CD4 and CD8 regulatory T cells in individuals under
Sallusto F. Production of interleukin 22 but not 15 years with multibacillary leprosy. PLoS One. 2013;
interleukin 17 by a subset of human skin-homing 8(11):e79072. doi:10.1371/journal.pone.0079072.
memory T cells. Nat Immunol. 2009;10(8):857–863. 78. Kumar S, Naqvi RA, Ali R, Rani R, Khanna N, Rao
doi:10.1038/ni.1767. DN. FoxP3 provides competitive fitness to
64. Fischer MK, Myer KA, Que SK, et al. CD4+CD25+T cells in leprosy patients via transcrip-
Pseudoepitheliomatous hyperplasia and transepider- tional regulation . Eur J Immunol. 2014;44(2):
mal elimination in lepromatous leprosy: does T-cell 431–439. doi:10.1002/eji.201343649.
plasticity play a role? J Drugs Dermatol. 2012;11(10): 79. Kumar S, Naqvi RA, Ali R, Rani R, Khanna N, Rao
1232–1235. DN. CD4 þ CD25þ T regs with acetylated FoxP3 are
65. de Lima Silveira E, de Sousa JR, de Sousa Aarao TL, associated with immune suppression in human lep-
et al. New immunologic pathways in the pathogen- rosy. Mol Immunol. 2013;56(4):513–520. doi:10.1016/
esis of leprosy: role for Th22 cytokines in the polar j.molimm.2013.04.015.
forms of the disease. J Am Acad Dermatol. 2015; 80. Saini C, Ramesh V, Nath I. Increase in TGF-beta
72(4):729–730. doi:10.1016/j.jaad.2014.11.023. secreting CD4(þ)CD25(þ) FOXP3(þ) T regulatory
66. de Sousa JR, de Sousa RPM, de Souza Aar~ao TL, cells in anergic lepromatous leprosy patients. PLoS
Dias LB, Oliveira Carneiro FR, Sim~oes Quaresma JA. Negl Trop Dis. 2014;8(1):e2639. doi:10.1371/journal.
Response of iNOS and its relationship with IL-22 pntd.0002639.
and STAT3 in macrophage activity in the polar 81. Sadhu S, Khaitan BK, Joshi B, Sengupta U, Nautiyal
forms of leprosy. Acta Trop. 2017;171:74–79. doi:10. AK, Mitra DK. Reciprocity between regulatory T
1016/j.actatropica.2017.03.016. cells and Th17 cells: relevance to polarized immunity
10 L. A. R. FROES ET AL.

in leprosy. PLoS Negl Trop Dis. 2016;10(1):e0004338. leprosy type 1 reactions. PLoS Negl Trop Dis. 2008;
doi:10.1371/journal.pntd.0004338. 2(12):e351. doi:10.1371/journal.pntd.0000351.
82. Abdallah M, Attia EAS, Saad AA, et al. Serum Th1/ 96. Chowdhry S, Shukla A, D’Souza P, Dhali T, Jaiswal
Th2 and macrophage lineage cytokines in leprosy; P. Treatment of severe refractory erythema nodosum
correlation with circulating CD4(þ) CD25(high) leprosum with tumor necrosis factor inhibitor
FoxP3(þ) T-regs cells. Exp Dermatol. 2014;23(10): Etanercept. Int J Mycobacteriol. 2016;5(2):223–225.
742–747. doi:10.1111/exd.12529. doi:10.1016/j.ijmyco.2016.02.002.
83. Chaves AT, Ribeiro-Junior AF, Lyon S, et al. 97. Little D, Khanolkar-Young S, Coulthart A, Suneetha
Regulatory T cells: friends or foe in human S, Lockwood DN. Immunohistochemical analysis of
Mycobacterium leprae infection? Immunobiology. 2018; cellular infiltrate and gamma interferon, interleukin-
223(4–5):397–404. doi:10.1016/j.imbio.2017.11.002. 12, and inducible nitric oxide synthase expression in
84. Quaresma JA, Aarao TL, Sousa JR, et al. T-helper 17 leprosy type 1 (reversal) reactions before and during
cytokines expression in leprosy skin lesions. Br J prednisolone treatment. Infect Immun. 2001;69(5):
Dermatol. 2015;173(2):565–567. doi:10.1111/bjd.13608. 3413–3417. doi:10.1128/IAI.69.5.3413-3417.2001.
85. Tarique M, Saini C, Naqvi RA, Khanna N, Rao DN. 98. Khanolkar-Young S, Rayment N, Brickell PM, et al.
Increased IL-35 producing Tregs and CD19(þ)IL- Tumour necrosis factor-alpha (TNF-alpha) synthesis
35(þ) cells are associated with disease progression in is associated with the skin and peripheral nerve
leprosy patients. Cytokine. 2017;91:82–88. doi:10. pathology of leprosy reversal reactions. Clin Exp
1016/j.cyto.2016.12.011. Immunol. 1995;99(2):196–202. doi:10.1111/j.1365-
86. Souza J, Sousa JR, Hirai KE, et al. E-selectin and P- 2249.1995.tb05532.x.
selectin expression in endothelium of leprosy skin 99. Kirkaldy AA, Musonda AC, Khanolkhar-Young S,
lesions. Acta Trop. 2015;149:227–231. doi:10.1016/j. Suneetha S, Lockwood DN. Expression of CC and
actatropica.2015.06.002. CXC chemokines and chemokine receptors in
87. de Sousa J, Sousa Aarao TL, Rodrigues de Sousa J, human leprosy skin lesions. Clin Exp Immunol.
et al. Endothelium adhesion molecules ICAM-1, 2003;134(3):447–453. doi:10.1111/j.1365-2249.2003.
ICAM-2, VCAM-1 and VLA-4 expression in leprosy.
02306.x.
Microb Pathog. 2017;104:116–124. doi:10.1016/j.mic-
100. Misch EA, Berrington WR, Vary JC, Jr, Hawn TR.
path.2017.01.021.
Leprosy and the human genome. Microbiol Mol Biol
88. Goulart IM, Penna GO, Cunha G. Immunopathology
Rev. 2010;74(4):589–620. doi:10.1128/MMBR.00025-10.
of leprosy: the complexity of the mechanisms of host
101. Polycarpou A, Walker SL, Lockwood DN. A system-
immune response to Mycobacterium leprae. Rev Soc
atic review of immunological studies of Erythema
Bras Med Trop. 2002;35(4):365–375. doi:10.1590/
nodosum leprosum. Front Immunol. 2017;8:233. doi:
s0037-86822002000400014.
10.3389/fimmu.2017.00233.
89. Mauri C, Ehrenstein MR. The ’short’ history of regu-
102. Wemambu SN, Turk JL, Waters MF, Rees RJ.
latory B cells. Trends Immunol. 2008;29(1):34–40.
Erythema nodosum leprosum: a clinical manifestation
doi:10.1016/j.it.2007.10.004.
90. Ochoa MT, Teles R, Haas BE, et al. A role for inter- of the arthus phenomenon. Lancet. 1969;2(7627):
leukin-5 in promoting increased immunoglobulin M 933–935. doi:10.1016/S0140-6736(69)90592-3.
at the site of disease in leprosy. Immunology. 2010; 103. Lee DJ, Li H, Ochoa MT, et al. Integrated pathways
131(3):405–414. doi:10.1111/j.1365-2567.2010.03314.x. for neutrophil recruitment and inflammation in lep-
91. Motta AC, Pereira KJ, Tarquinio DC, Vieira MB, rosy. J Infect Dis. 2010;201(4):558–569. doi:10.1086/
Miyake K, Foss NT. Leprosy reactions: coinfections 650318.
as a possible risk factor. Clinics (Sao Paulo). 2012; 104. Iyer A, Hatta M, Usman R, et al. Serum levels of
67(10):1145–1148. doi:10.6061/clinics/2012(10)05. interferon-gamma, tumour necrosis factor-alpha, sol-
92. Sousa AL, Fava VM, Sampaio LH, et al. Genetic and uble interleukin-6R and soluble cell activation
immunological evidence implicates interleukin 6 as a markers for monitoring response to treatment of lep-
susceptibility gene for leprosy type 2 reaction. J rosy reactions. Clin Exp Immunol. 2007;150(2):
Infect Dis. 2012;205(9):1417–1424. doi:10.1093/infdis/ 210–216. doi:10.1111/j.1365-2249.2007.03485.x.
jis208. 105. Faber WR, Jensema AJ, Goldschmidt WF. Treatment
93. Mastrangelo G, Marcer G, Cegolon L, et al. How to of recurrent erythema nodosum leprosum with
prevent immunological reactions in leprosy patients infliximab. N Engl J Med. 2006;355(7):739. doi:10.
and interrupt transmission of Mycobacterium leprae 1056/NEJMc052955.
to healthy subjects: two hypotheses. Med Hypotheses. 106. Ramien ML, Wong A, Keystone JS. Severe refractory
2008;71(4):551–563. doi:10.1016/j.mehy.2008.05.016. erythema nodosum leprosum successfully treated
94. Walker SL, Balagon M, Darlong J, Erythema with the tumor necrosis factor inhibitor etanercept.
Nodosum Leprosum International STudy Group, Clin Infect Dis. 2011;52(5):e133–e135. doi:10.1093/
et al. ENLIST 1: an international multi-centre cross- cid/ciq213.
sectional study of the clinical features of erythema 107. Schmitz V, Prata RB, Barbosa MG, et al. Expression
Nodosum Leprosum. PLoS Negl Trop Dis. 2015;9(9): of CD64 on circulating neutrophils favoring systemic
e0004065. doi:10.1371/journal.pntd.0004065. inflammatory status in erythema nodosum leprosum.
95. Walker SL, Nicholls PG, Butlin CR, et al. PLoS Negl Trop Dis. 2016;10(8):e0004955. doi:10.
Development and validation of a severity scale for 1371/journal.pntd.0004955.
INTERNATIONAL REVIEWS OF IMMUNOLOGY 11

108. Buckle AM, Hogg N. The effect of IFN-gamma and 121. Hauptmann G, Tappeiner G, Schifferli JA. Inherited
colony-stimulating factors on the expression of neu- deficiency of the fourth component of human com-
trophil cell membrane receptors. J Immunol. 1989; plement. Immunodefic Rev. 1988;1(1):3–22.
143(7):2295–2301. 122. Bjorvatn B, Barnetson RS, Kronvall G, Zubler RH,
109. Voorend CG, Post EB. A systematic review on the Lambert PH. Immune complexes and complement
epidemiological data of erythema nodosum lepro- hypercatabolism in patients with leprosy. Clin Exp
sum, a type 2 leprosy reaction. PLoS Negl Trop Dis. Immunol. 1976;26(3):388–396.
2013;7(10):e2440 doi:10.1371/journal.pntd.0002440. 123. Lahiri R, Sandoval FG, Krahenbuhl JL, Shannon EJ.
110. Wallach D, Cottenot F, Bach MA. Imbalances in T Activation of complement by Mycobacterium leprae
cell subpopulations in lepromatous leprosy. Int J requires disruption of the bacilli. Lepr Rev. 2008;
Lepr Other Mycobact Dis. 1982;50(3):282–290. 79(3):311–314.
111. Mshana RN, Haregewoin A, Belehu A. Thymus- 124. Duthie MS, Hay MN, Morales CZ, et al. Rational
dependent lymphocytes in leprosy. II. Effect of chemo- design and evaluation of a multiepitope chimeric
therapy on T-lymphocyte subpopulations. J Clin fusion protein with the potential for leprosy diagno-
Immunol. 1982;2(2):69–74. doi:10.1007/BF00916889. sis. Clin Vaccine Immunol. 2010;17(2):298–303. doi:
112. Narayanan RB, Laal S, Sharma AK, Bhutani LK, 10.1128/CVI.00400-09.
Nath I. Differences in predominant T cell pheno- 125. Duthie MS, Goto W, Ireton GC, et al. Use of protein
types and distribution pattern in reactional lesions of antigens for early serological diagnosis of leprosy.
tuberculoid and lepromatous leprosy. Clin Exp Clin Vaccine Immunol. 2007;14(11):1400–1408. doi:
Immunol. 1984;55(3):623–628. 10.1128/CVI.00299-07.
113. Wallach D, Flageul B, Cottenot F, Bach MA. Patients 126. Buhrer-Sekula S. PGL-I leprosy serology. Rev Soc
with erythema nodosum leprosum lack T-suppressor Bras Med Trop. 2008;41 (Suppl 2):3–5.
cells. Arch Dermatol. 1985;121(11):1379. 127. Wen Y, You YG, Yuan LC, et al. Evaluation of novel
114. Hussain T, Kulshreshtha KK, Yadav VS, Katoch K. tools to facilitate the detection and characterization
CD4þ, CD8þ, CD3þ cell counts and CD4þ/CD8þ of leprosy patients in China. Biomed Res Int. 2014;
ratio among patients with mycobacterial diseases 2014:371828. doi:10.1155/2014/371828.
(leprosy, tuberculosis), HIV infections, and normal 128. Hunter SW, Brennan PJ. A novel phenolic glycolipid
healthy adults: a comparative analysis of studies in
from Mycobacterium leprae possibly involved in
different regions of India. J Immunoassay
immunogenicity and pathogenicity. J Bacteriol. 1981;
Immunochem. 2015;36(4):420–443. doi:10.1080/
147(3):728–735. doi:10.1128/JB.147.3.728-735.1981.
15321819.2014.978082.
129. Buhrer-Sekula S, Smits HL, Gussenhoven GC, et al.
115. Vieira AP, Trindade MA, Pagliari C, Avancini J,
Simple and fast lateral flow test for classification of
et al. Development of Type 2, but not Type 1, lep-
leprosy patients and identification of contacts with
rosy reactions is associated with a severe reduction
high risk of developing leprosy. J Clin Microbiol.
of circulating and in situ regulatory T-cells. Am J
2003;41(5):1991–1995. doi:10.1128/jcm.41.5.1991-
Trop Med Hyg. 2016;94(4):721–727. doi:10.4269/
ajtmh.15-0673. 1995.2003.
116. de Azevedo MP, de Melo PH. A comparative study 130. Moura RS, Calado KL, Oliveira ML, Buhrer-Sekula
of the complementary activity of serum in the polar S. Leprosy serology using PGL-I: a systematic review.
forms of leprosy and in the leprosy reaction. Int J Rev Soc Bras Med Trop. 2008;41 (Suppl 2):11–18.
Lepr Other Mycobact Dis. 1966;34(1):34–38. doi:10.1590/s0037-86822008000700004.
117. Lange K, Wasserman E, Slobody LB. The signifi- 131. Iyer AM, Mohanty KK, van Egmond D, et al.
cance of serum complement levels for the diagnosis Leprosy-specific B-cells within cellular infiltrates in
and prognosis of acute and subacute glomeruloneph- active leprosy lesions. Hum Pathol. 2007;38(7):
ritis and lupus erythematosus disseminatus. Ann 1065–1073. doi:10.1016/j.humpath.2006.12.017.
Intern Med. 1960;53:636–646. doi:10.7326/0003-4819- 132. Marzano AV, Tosi D, Cusini M, Nunzi E, Massone
53-4-636. C, Crosti C. Facial reversal reaction: a dermatological
118. Ohi H, Tamano M. Decreased apolipoprotein levels emergency. J Dermatol. 2012;39(2):203–205. doi:10.
are associated with decreased complement levels in 1111/j.1346-8138.2011.01248.x.
acute glomerulonephritis. Nephron. 2001;88(4): 133. Roche PW, Theuvenet WJ, Britton WJ. Risk factors
389–390. doi:10.1159/000046028. for type-1 reactions in borderline leprosy patients.
119. de Messias IJ, Reis A, Brenden M, Queiroz-Telles F, Lancet. 1991;338(8768):654–657. doi:10.1016/0140-
Mauff G. Association of major histocompatibility 6736(91)91232-j.
complex class III complement components C2, BF, 134. Antunes DE, Araujo S, Ferreira GP, et al.
and C4 with Brazilian paracoccidioidomycosis. Identification of clinical, epidemiological and labora-
Complement Inflamm. 1991;8(5–6):288–293. doi:10. tory risk factors for leprosy reactions during and
1159/000463198. after multidrug therapy. Mem Inst Oswaldo Cruz.
120. Grevink ME, Horst G, Limburg PC, Kallenberg CG, 2013;108(7):901–908. doi:10.1590/0074-0276130222.
Bijl M. Levels of complement in sera from inactive 135. Brito M d F d M, Ximenes RAA, Gallo MEN,
SLE patients, although decreased, do not influence B€uhrer-Sekula S. Association between leprosy reac-
in vitro uptake of apoptotic cells. J Autoimmun. tions after treatment and bacterial load evaluated
2005;24(4):329–336. doi:10.1016/j.jaut.2005.03.004. using anti PGL-I serology and bacilloscopy. Rev Soc
12 L. A. R. FROES ET AL.

Bras Med Trop. 2008;41 (Suppl 2):67–72. doi:10. Infect Dis. 2014;79(2):233–239. doi:10.1016/j.diagmi-
1590/s0037-86822008000700014. crobio.2014.02.006.
136. Hungria EM, Oliveira RM, Penna GO, et al. Can 140. da Conceicao Oliveira Coelho Fabri A, Carvalho AP,
baseline ML flow test results predict leprosy reac- Araujo S, et al. Antigen-specific assessment of the
tions? An investigation in a cohort of patients immunological status of various groups in a leprosy
enrolled in the uniform multidrug therapy clinical endemic region. BMC Infect Dis. 2015;15:218. doi:10.
trial for leprosy patients in Brazil. Infect Dis Poverty. 1186/s12879-015-0962-4.
2016;5(1):110 doi:10.1186/s40249-016-0203-0. 141. Hungria EM, Buhrer-Sekula S, de Oliveira RM, et al.
137. Duthie MS, Ireton GC, Kanaujia GV, et al. Selection Leprosy reactions: the predictive value of
of antigens and development of prototype tests for Mycobacterium leprae-specific serology evaluated in
point-of-care leprosy diagnosis. Clin Vaccine a Brazilian cohort of leprosy patients (U-MDT/CT-
Immunol. 2008;15(10):1590–1597. doi:10.1128/CVI. BR). PLoS Negl Trop Dis. 2017;11(2):e0005396. doi:
00168-08. 10.1371/journal.pntd.0005396.
138. Duthie MS, Hay MN, Rada EM, et al. Specific IgG 142. Arantes CK, Garcia MLR, Filipe MS, et al. Avaliaç~ao
antibody responses may be used to monitor leprosy dos serviços de sa
ude em relaç~ao ao diagn
ostico pre-
treatment efficacy and as recurrence prognostic coce da hansenıase. Epidemiol Serv Sa ude. 2010;
markers. Eur J Clin Microbiol Infect Dis. 2011;30(10): 19(2):155–164.
1257–1265. doi:10.1007/s10096-011-1221-2. 143. Wang J, Wang Z, Yao Y, et al. The fibroblast growth
139. Duthie MS, Raychaudhuri R, Tutterrow YL, et al. A factor-2 arrests Mycobacterium avium sp. paratuber-
rapid ELISA for the diagnosis of MB leprosy based culosis growth and immunomodulates host response
on complementary detection of antibodies against a in macrophages. Tuberculosis (Edinb)). 2015;95(4):
novel protein-glycolipid conjugate. Diagn Microbiol 505–514. doi:10.1016/j.tube.2015.04.006.

You might also like