You are on page 1of 11

Microbial Pathogenesis 137 (2019) 103714

Contents lists available at ScienceDirect

Microbial Pathogenesis
journal homepage: www.elsevier.com/locate/micpath

Diet and nutrition: An important risk factor in leprosy T


a,∗∗ b c d e
Ved Prakash Dwivedi , Arindam Banerjee , Indraneel Das , Aparajita Saha , Malabika Dutta ,
Bhavya Bhardwaja, Saptarshi Biswasf, Debprasad Chattopadhyayg,8,∗
a
Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, 110067, India
b
Rafi Ahmed Dental College, Govt of West Bengal, Moulalai, Kolkata, 7600014, India
c
Declibac Technologies Private Limited, 24 B, Lake Road, Kolkata, 700 029, India
d
Nutri-Diet Kolkata, 34A Charu Avenue, Kolkata, 700033, India
e
Department of Dietetics, Kothari Medical Center, 8/3 Alipore Road, Kolkata, 700027, India
f
Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
g
ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi, 590010, India
8
ICMR-Virus Unit, ID & BG Hospital, General Block 4, 57 Dr Suresh C Banerjee Road, Beliaghata, Kolkata, 700010, India

A R T I C LE I N FO A B S T R A C T

Keywords: Leprosy, once considered as poor man's disease may cause severe neurological complications and physical
Leprosy disabilities. Classification of leprosy depends upon the cell mediated and humoral immune responses of the host,
Multi-drug therapy from tuberculoid to lepromatous stage. Current therapy to prevent the disease is not only very lengthy but also
Nutrition consists of expensive multiple antibiotics in combination. Treatment and the duration depend on the bacillary
Immune responses
loads, from six months in paucibacillary to a year in multibacillary leprosy. Although as per WHO re-
Cytokines
Cell mediated immunity
commendations, these antibiotics are freely available but still out of reach to patients of many rural areas of the
Humoral immunity world. In this review, we have focused on the nutritional aspect during the multi-drug therapy of leprosy along
with the role of nutrition, particularly malnutrition, on susceptibility of Mycobacterium leprae and development
of clinical symptoms. We further discussed the diet plan for the patients and how diet plans can affect the
immune responses during the disease.

1. Introduction Leprosy mainly affects the skin and peripheral nerves, along with
the mucosa of the upper respiratory tract and the eyes, leading to the
Leprosy is a chronic debilitating bacterial disease caused by in- progressive and permanent nerve damage to deformities to paralysis.
tracellular slow-growing gram-positive acid-fast bacillus Mycobacterium Based on bacillary load leprosy is classified as paucibacillary (PB) and
leprae (M. leprae), or rarely with M. lepromatosis, identified by Gerhard multibacillary (MB) forms. PB leprosy is a mild form with 2–5 pale or
Henrik Armauer Hansen in 1873. The oldest evidence for leprosy dates reddish skin lesions, whereas MB consists of more than five skin lesions,
back to 600 BCE in India, while genomic studies from skeletal remains nodules, plaques, thickened dermis or skin infiltration [2,3]. Histori-
suggest that lepromatous leprosy was present in India around 2000 B.C. cally, leprosy was ostracized by society for its incurable nature. Due to
Further, the presence of leprosy during the post-urban phase of the the absence of any cell or animal model, understanding about the
Indus civilization suggests that M. leprae evolved in Africa and migrated biology of leprosy bacillus largely remains unknown. Although M. le-
to India before the Late Holocene, during the third millennium B.C. The prae grow in mice footpads but failed to cause nerve damage; while it
incubation period of M. leprae varies from 3 to 5 years and is spread via causes nerve damage in armadillos, a rarely used animal model in re-
air droplets of the nasal and oral discharge from non-treated multi- search. The scenario has changed with the introduction of dapsone in
bacillary leprosy patients [1] and or via digital impregnation of skin. the 1940s and subsequently the multi-drug therapy (MDT) by the WHO
Interestingly over 99% of the population has adequate natural im- using a combination of dapsone, rifampin and clofazimine or clari-
munity against leprosy and 85% of the clinical cases are non-infectious. thromycin. The MDT treatment for PB and MB varies from 6 to 12
Moreover, an infectious person can become non-infectious within a months respectively, without relapses or recurrences or the develop-
week of the first dose of treatment [2]. ment of resistance, with an aim to reduce the grade two physical


Corresponding author. ICMR-National Institute of Traditional Medicine, Nehru Nagar, Belagavi, 590010, India.
∗∗
Corresponding author. Immunobiology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
E-mail addresses: vedprakashbt@gmail.com, ved@icgeb.res.in (V.P. Dwivedi), debprasadc@yahoo.co.in (D. Chattopadhyay).

https://doi.org/10.1016/j.micpath.2019.103714
Received 25 February 2019; Received in revised form 9 August 2019; Accepted 2 September 2019
Available online 04 September 2019
0882-4010/ © 2019 Elsevier Ltd. All rights reserved.
V.P. Dwivedi, et al. Microbial Pathogenesis 137 (2019) 103714

disabilities [5]. Recent studies by WHO estimated that the global immunity (IFN-β, IL-4, IL-10). TT leprosy has a hypopigmented center
burden of leprosy was dramatically decreased from 5.2 million in 1985 with raised erythematosus border; LL has macules, papules, and plaques
to 176,176 in 2015. with firm nodules; while Borderline (BT, BB, BL) forms have hypopig-
Being a poor man's disease, leprosy remains a public health problem mented macules. Molecular diagnosis of leprosy is usually done by 16S
in underdeveloped nations. The relation between the risk factors and ribosomal RNA gene polymerase chain reaction (PCR) assay, as most
development of leprosy is still unclear because it is difficult to in- skin lesions have no identifiable bacteria [16].
vestigate the causal relationships of infection and the onset of symp-
toms due to the long incubation period (3–5 years) of M. leprae [6]. 3. How leprosy cause nerve damage
Moreover, based on the [7] classification there are five clinical forms:
tuberculoid, lepromatous, borderline tuberculoid, mid-borderline, and One of the uniqueness of leprosy is axon demyelination that leads to
lepromatous borderline. The specific risk factors for contacting leprosy, peripheral neuropathy. The nerve damage in leprosy is caused by M.
except in endemic areas, include index patient, age of the contact, leprae after entering into the myelin sheath producing Schwann cells.
physical distance to the patient [8] and poverty. Although its associa- However, a recent study demonstrated that nerve damage is not directly
tion with susceptibility and clinical progress is unclear, many poor caused by M. leprae but through bacteria-specific phenolic glycolipid-1
countries have low prevalence rates; while economically developing (PGL-1) with myelinating glia. The M. leprae-induced nerve damage
Brazil had one of the highest new case detection rate [5]. Case-control was demonstrated in Zebrafish larvae where axonal demyelination is
studies on socioeconomic factors from Brazil [9] and Bangladesh [10] initiated by the bacteria-infected macrophages, and demyelination oc-
revealed that food shortage at any time of life was associated with le- curs in areas of intimate contact. Infected macrophages expressing PGL-
prosy. It was evident that when rice prices are high and income is low, 1 damage axons by producing neurotoxic response to nerve cells.
the family is forced to reduce the number of meals a day, or the intake Moreover, within infected macrophages, PGL-1 induces nitric oxide
of food [11]. Inadequate food intake leads to reduced intake of carbo- synthase that generates more reactive nitrogen species, and damages
hydrates, proteins, fats, vitamins, and minerals; and the nutritional axons by injuring mitochondria and inducing demyelination. Thus, M.
deficiency impairs the immune system against infections [12]. Al- leprae can hijack the important repair mechanism and excrete toxic
though the risk of subclinical infection is not always related to food chemicals to stop damage repairing process. Hence, leprosy is placed in
shortage, it helps in progressing the infection to the clinical disease. the same category of multiple sclerosis and Guillain-Barré syndrome
Another case-control study in Indonesia [3] and Bangladesh [11] also where the immune system can damage the myelin sheath [17].
showed that the differences in dietary intake between new patients and
control subjects are immunity related. Despite the use of chemopro- 4. Genetic susceptibility and metabolomic in leprosy
phylaxis and MDT, leprosy incidence is relatively stable indicating the
necessity of dietary diversity in high-prevalence areas [3]. Almost 2.5 Genetics is an important factor for leprosy susceptibility. Exploring
lakh new leprosy cases are detected each year, and 7% of these cases the mechanism of host genetic susceptibility and clinical manifestations
are grade-2 disabilities [5]. A recent review of 39 selected studies be- observational studies from Bangladesh and Indonesia showed that ge-
tween 2006 and 2016, from Brazil, India, and Bangladesh revealed that netic relationship as a risk factor for leprosy among household contacts
age, poor sanitation, socioeconomic conditions, past food shortage, [8,18] along with other factors [4,19,20]. Research on the relationship
food-insecurity, household contact, manual labor, crowded household, of human genetics, the M. leprae genome, and leprosy susceptibility is
inequality, poor health care, and low education are the risk markers for useful for developing a better understanding of the disease [21,22].
leprosy [13]. However, the welfare and primary health care policies on Detail interplays of genetics and leprosy susceptibility was outlined in a
poverty-related leprosy reduction are less known. While a close re- recent review [23]. It is known that PARK2/PACRG and NRAMP1 genes
lationship between social policies and health conditions of the people in on chromosome 6q25-q27 and 2q35 are associated with leprosy sus-
developing nations on a new case detection rate is evident from Brazil, ceptibility [24]. While increasing numbers of reports showed that the
using national databases of 1358 municipalities during 2004–2011, innate immune response has a key role in determining susceptibility to
Brazilian conditional cash transfer (Bolsa Família Program) and Pri- leprosy and its reactions with genetic regulation of the innate im-
mary Health Care (Family Health Program) revealed that the new case munity, presented by polymorphisms of the NOD2 gene, that linked
detection rate was significantly reduced in municipalities having in- with the increased susceptibility and development of leprosy reactions
termediate, high and BFP coverage. Thus, the primary health care was [23,25]. The genetic variability is also associated with Toll-like re-
effective to record increased new case detection rate as impact of ceptors (TLRs), particularly polymorphism in TLR-1 (T1805G variant)
conditional cash transfer; while the reduction of the new case detection that impairs intracellular signaling of mycobacteria and provides pro-
is due to the reduced disease incidence [14]. These findings indicated a tection against type 1 reactions. Genomic studies have confirmed a
logical relationship between leprosy and socioeconomic factors, sug- correlation among individual genetics and immunity with leprosy sus-
gesting that leprosy control policies must be targeted to the vulnerable ceptibility, as evident from the large-scale genome-wide association
groups. However, the underlying mechanism between food, poverty, study on 706 patients and 1225 controls in China. The study identified
and leprosy is still unclear. six genes (CCDC122, C13orf31, NOD2, TNFSF15, HLA-DR, RIPK2) that
are involved in innate immune response associated with leprosy sus-
2. Classification and diagnosis ceptibility; while variants of NOD2-mediated signaling pathway are
associated with M. leprae infection [25]. In lepromatous leprosy the
Based on immune response and pathology, leprosy has been clas- expression of galectin-3 is associated with the differentiation of
sified into five types [15]: Tuberculoid (TT), Borderline-Tuberculoid monocytes into macrophages; while the NOD2 expression is related to
(BT), Mid-borderline (BB), Borderline-Lepromatous (BL), and Lepro- the tuberculoid form by promoting monocyte differentiation into den-
matous (LL). Patient with one skin lesion is categorized as single lesion dritic cells. A recent report showed that three functional components of
paucibacillary (SLPB), having five or fewer skin lesions without bacilli the inflammatory response, associated with type 1 reactions, are
in smears; while more than six lesions with bacilli in skin smear re- regulated by a 44-gene set: (i) anti-inflammatory, (ii) proinflammatory,
present multibacillary (MB) form. Furthermore, TT leprosy is char- and (iii) arachidonic acid metabolism mediator regulators. People with
acterized by the presence of few bacilli, a minor loss of nerve with type 1 reactions had a defective regulation of inflammatory response
strong cell-mediated (IFN-γ, IL-2) and weak humoral immunity; while against M. leprae antigens, involving anti-inflammatory and proin-
LL leprosy exhibited a wide range of lesions containing multiple bac- flammatory components of the innate response. These findings can help
teria with skin and nerve involvement and strong cell-mediated to understand the pathogenesis and development of early diagnostic

2
V.P. Dwivedi, et al. Microbial Pathogenesis 137 (2019) 103714

markers to identify and predict the high-risk individual of leprosy re- International text book of Leprosy. Scollard DM, Gillis TP (Ed),
actions [25]. American Leprosy Missions. www.internationaltextbookofleprosy.org.
Although the exact mechanisms of disease susceptibility, onset, and It is known that IL-2 and IFN-γ are dominant in TT, whereas IL-4, IL-
progression of leprosy are still unclear, recent research focused on nu- 5, and IL-I0 in LL [41]. Thus, the mutual relationship between cell-
tritional, genetic and metabolic aspects of the disease, particularly fatty mediated immunity, antibody production and resistance or suscept-
acids, essential amino acids, glycoproteins, and nitric oxide synthetase. ibility to M. leprae depends on the differences in cytokine production by
Lipid metabolism is one of the major aspects of leprosy, as it helps to T-cells. T cell activation is regulated by CD1-mediated antigen pre-
distinguish the clinical forms. Fatty acids with anti-inflammatory and sentation pathway via major histocompatibility complex [42], influ-
proinflammatory role can serve as potential markers for susceptibility enced by the nutritional status of the individual.
and pathogenesis, as LL patients contain higher levels of omega-6 [26]. Host resistance against leprosy requires Th1-based responses, in-
The patients having omega-3 metabolites in serum and skin after itiated by CD4+ Th1 cells with the secretion of pro-inflammatory cy-
multidrug therapy showed anti-inflammatory and pro-resolving roles, tokines [34] IFN-γ and TNF-α, which synergistically activate micro-
suggesting that the host tolerance to M. leprae is a strategy to avoid bicidal mechanisms of macrophages. Predominantly LL patients secrete
tissue damage [27]. Similarly, the tryptophan metabolizing enzyme IL-10 and Th-2 producing cytokines IL-4, IL-5 and IL-13, leading to
indoleamine 2,3-dioxygenase is responsible for immunosuppressed antibody production with host defense dysregulation [43]. On the other
status of LL leprosy in a pathogen-specific manner and can serve as a hand, TT leprosy is characterized by the high level of Th-1 specific pro-
marker for diagnosis and prognosis [28]. Analysis of serum proteome of inflammatory cytokines IFN-γ, IL-15, IL-1β; Th-17 differentiating cy-
leprosy patients showed that differential expression of acute-phase tokines IL-6, TGF-β and IL-23 [44] like tuberculosis [45,46]. Thus, the
protein alpha-1-acid glycoprotein (AGP) isoforms and its changes in outcome of the host response to M. leprae is mainly determined by
serum level can act as putative biomarker for the diagnosis and mon- cytokines and chemokines, which can serve as a tool to predict the
itoring of ENL reactions [28]. In leprosy, the NO generation via nitric protection or progression of leprosy [47].
oxide synthases (NOS) are regulated by IFN-γ, and the expression of
inducible i-NOS, endothelial e-NOS, and neuronal n-NOS suggest that i- 6. Nutrition and leprosy
NOS expression can serve as a diagnostic marker for disease spectrum
[29]. The science that interprets the interaction of nutrients in food in
relation to growth, reproduction, and maintenance of health with pre-
5. Relationship of immunity and immune response in leprosy vention or counter diseases of an organism is termed as nutrition.
Nutrition includes the intake of food, its absorption, assimilation, bio-
Inter-individual variability in clinical presentation and cellular im- synthesis, catabolism, and excretion. The exact role of nutrition on
mune response in leprosy acts as a model of immunoregulatory disease susceptibility and development of clinical leprosy is not yet established
in humans [30–32]. About 95% of people exposed to Mycobacterium [48,49]. Recent reports on the role of micronutrients and the immune
develop natural resistance with higher innate and cellular immunity response in leprosy indicate a huge knowledge gap. Moreover, earlier
against M. leprae [33] and do not develop leprosy due to the low pro- studies are based on either blood analyses, or on diets of cured patients.
liferation rate and pathogenicity of the bacilli. Additionally, clinical Thus, it is difficult to determine whether leprosy is a cause or a con-
manifestation showed diverse immunological spectrum, as TT patients sequence of nutritional deficiencies [50–53]. As M. leprae is an in-
showed high cellular responses to M. leprae antigens, but low antibody tracellular bacterium, so host defense is based on cell-mediated im-
titer that develops localized granulomatous disease with few bacilli. On munity (Figs. 1 and 3). Previously it has been reported that protein
the other hand, the LL patients do not elicit M. leprae specific Th1 re- deficiency along with inadequate or low intake of vitamins and mi-
sponses, but high IgM titers [34]; while borderlines are im- nerals rich food from diverse food groups is linked to a reduced cell-
munologically unstable and prone to Type 1/reversal reactions (RR) mediated immunity [54], which put leprosy patient at risk of reduced
and Type 2/erythema nodosum leprosum (ENL) [35]. Thus, an im- immunity. Moreover, the phenomenon of “immune reconstitution”
balance between cellular and humoral immune response is responsible found in HIV cases after anti-retroviral therapy may have a role in le-
for the different clinical form of leprosy and varied immune responses prosy [55,56]. Immunosuppressed HIV patients suffer prolonged nu-
exhibit a variety of clinical features from TT to LL. Cell-mediated im- tritional deficiencies, and if such patients are supplied with good food
munity is the major defense against M. leprae (Fig. 1), and its outcome the body responds to infection by developing clinical disease. Com-
depends on host response [36]. pared to the normal population leprosy patients are financially poor
The intracellular life cycle of M. leprae initiates with the host cell and unable to spend on food, have less household food stocks and di-
entry. The PGL-1 epitope on the bacterial cell wall is recognised by verse diet. Thus, the patient had low consumption of proteins (such as
complement receptor (CR)-1,3,4 mediate phagocytosis [37], and the eggs, fish, meat, and milk), vitamin and mineral rich fruits and vege-
bacterium was recognised via molecular pattern recognition with the tables. It was observed that leprosy patients have less access to socio-
help of the complement and TLRs, expressed on dendritic cells (DCs) economic, health and nutritional factors compared to the control po-
and macrophages [38]. M. leprae invade and survive within macro- pulation [11]. The main factors for increased risk of leprosy include
phages, DC and Schwann cells. The stages of M. leprae infection are lower income, food expenditure, BMI, and dietary diversity score (DDS)
presented in Fig. 2. with an absence of household food stocks and past year food shortage
After recognition by TLRs, the NF-kB is activated to release several [11]. Collectively, the infection course depends on host genetic factors
pro-inflammatory chemokines and cytokines like GM-CSF, TNF-α, IL- and is influenced by environmental and nutritional status [24,57,58].
10, IL-1β, IL-12 that activate the migration of macrophages to the Nutritional deficiencies are common in leprosy endemic areas, so that
lymphoid organs to present M. leprae antigen to naïve T-cells (Fig. 3). there is a possibility that the clinical presentation is a result of nutri-
Cell-mediated immunity is elicited by T helper type-1 (Th-1) cells tional deficiencies along with environmental and genetic factors of the
with the secretion of IL-2 and IFN-γ; whereas Th-2 increases the hu- host. Nutrition and food supplements are known to influence the im-
moral immune response by secreting IL-4, IL-5, and IL-10. Based on mune response in many diseases. A deficiency of vitamins and trace
inhibitory/stimulatory signals, T-cell subsets CD4+ Th1 or Th2, elements affect the innate and adaptive immune response [12].
Cytotoxic T cell lymphocytes (CTL) or Th17 activate the regulatory T-
cells (Treg), which generate M. leprae-specific response in LL (Fig. 4) 7. Immune response and influence of nutrition in leprosy
patients [39,40].
Hooij A and Geluk A., 2016. Immunodiagnostics for Leprosy. In: The human body with uniform temperature provides a nutrition

3
V.P. Dwivedi, et al. Microbial Pathogenesis 137 (2019) 103714

Fig. 1. Immune responses in active leprosy [36]. Th1–Th2 paradigm: insights from leprosy. J Invest Dermatol 102: 828–832.

rich environment and serves as an attractive host for pathogens. only causes the suppression of immune responses, but several micro-
However, its natural barriers (mechanical, chemical, biological) and nutrients help to maintain the integrity of natural barriers, and the
immune response protects the body from pathogens. The skin, epithelial function of the immune system [61]. Clinical manifestations of infec-
and mucosal (respiratory and digestive mucosa) surfaces, with ciliary tion can also occur due to tissue inflammation mediated by the immune
and peristaltic movement constitute its mechanical barrier; while fatty response, as TNF-α, NO and reactive oxygen species (ROS) having mi-
acids, bactericidal lysozyme (saliva, sweat, tears), proteolytic enzymes crobicidal action can induce tissue damage. Thus, antioxidant sub-
(stomach and intestines), stomach acid and anti-bacterial peptides of stances (endogenous or nutritional) and cytokine modulations are es-
skin and intestine provide the chemical barrier. The microbiome (in- sential for a balanced immune response for host protection and
testinal flora) and epithelium receptors compete with pathogens for pathogen control [62]. The effect of malnutrition are variable and
nutrients and produce microbicidal substances [59]. Once a pathogen difficult to measure during infection; while a nutritional deficiency may
overcomes these barriers, host immune response gets activated, and the increase the susceptibility and aggravate the disease prognosis in TB
control of infection depends on the ability of hosts to produce a counter- and measles [63]. Moreover, persistent pathogens can cause auto-
response against the invading pathogen [60]. Nutritional deficiency not immune and inflammatory diseases, while most pathogens trigger an

Fig. 2. Different stages of Leprosy Infection. Treatment of Leprosy in the early 21st century. Dermatologic Therapy 22(6): 518–537.

4
V.P. Dwivedi, et al. Microbial Pathogenesis 137 (2019) 103714

Fig. 3. Innate and acquired immunity to Mycobacterium leprae infection [28]. Advances in leprosy immunology and the field application: A gap to bridge. Clinics
in Dermatology 34(1): 82–95.

vegetables, and fruits [9]. Studies from India and Brazil demonstrated
that an imbalance of dietary intake is related to feeding habits due to
the lack of knowledge about the nutritional value of foods [50,65]. Low
body weight or overweight people also have dietetic inadequacies while
overweight persons consume empty calories. A study in Brazil revealed
that 41.9% of leprosy patients are overweight to obese while 3.6% were
underweight [65]. The low quality of diet is associated with a higher
risk of leprosy due to low intake of antioxidants that impaired immune
defense against M. leprae [1,50,66]. Another study with 58 leprosy
patients showed that nutritional deficiency in different forms of leprosy,
leads to decreased serum levels of vitamin A (retinol), C (ascorbic acid),
D (cholecalciferol), and E (tocopherol), along with magnesium, sele-
nium, and zinc [66]. During leprae infection, macrophages get acti-
vated and kill the bacterium by the generation of ROS, NO, and RNS
which are known to kill intracellular microbes. However, these radicals
have oxidant activity and thus can cause tissue damage due to high
inflammation. Dietary antioxidants can counterbalance these effects of
oxidative stress, and thus a reduction of antioxidant levels can com-
plicate the treatment and disease progress [67].

8. Vitamin A in leprosy

Vitamin A along with its precursor's retinoic acid and β-carotene are
Fig. 4. Schematic representation of T cells involved in leprosy disease important antioxidants that interact with peroxyl free radicals and
spectrum. hydroperoxides by inhibiting lipid peroxidation [68], while carotenoids
quench the cellular singlet oxygen into less reactive species [69]. Beta-
inflammatory response. Thus, nutritional factors have a role to de- carotene act as an antioxidant in low partial oxygen pressures, while in
termine the outcome of the diseases [64]. high oxygen concentration vitamin E complement this action. As in-
The risk of contracting leprosy is reported to be associated with creased oxidative stress documented in leprosy patients [1] indicate the
poverty, dietetic inadequacy, reduced intake of calorie, proteins, requirement of vitamin A. Vitamin A is also required to regulate the
immune response of innate (cellular) and acquired (humoral) immunity

5
V.P. Dwivedi, et al. Microbial Pathogenesis 137 (2019) 103714

[70]. Thus, deficiency of vitamin A is associated with decreased pha- enzyme that inactivate vitamin D3 [85]. On the other hand, vitamin D3
gocytosis, oxidative burst of macrophages [61] and regulation of the NK helps to disseminate M. leprae by inducing dendritic and T-regulatory
cell population [71]. Moreover, vitamin A deficiency is reported to cells by inhibiting co-stimulatory molecules and inducing FOXp3 ex-
decrease IFN-γ production (Th1 response) and Th2 mediated humoral pressions and IL-10 production [86]. Infectious diseases are multi-
responses [12,72]. An Indonesian study showed that vitamin A defi- factorial but influenced by environmental and genetic factors. A poly-
cient children produce fewer IFN-γ and become susceptible to myco- morphism in the vitamin D receptor gene (codon 352 C/T) can regulate
bacterial diseases as IFN-γ producing Th1 cells provide protection the active metabolite of D3 and affect bone mineralization as well as
against intracellular pathogens [12]. Furthermore, retinoic acid in- Th1/Th2 immune response. Individuals with ‘tt’ homozygous alleles
duced IL-10 production as an anti-inflammatory response by inhibiting develop Th1 response, while the ‘TT’ homozygous alleles develop Th2
IL-12 and TNF-α. As IL-12 helps to induce Th1 differentiation, thus the response. In a case-control study it was shown that genotype ‘tt’ was
supply of vitamin A inhibits Th1 response, indicating that vitamin A less frequent in TB patients, while another study with 231 leprosy pa-
deficiency compromises Th2 responses and decreases IgG1 and IgE tients (107 TT and 124 LL) showed that genotype ‘tt’ was associated
antibody production [12,72]. The decrease in serum concentrations of with polymorphism in vitamin D receptor gene (codon 352 C/T) in
vitamin A in LL patients can reduce Th1 response and M. leprae re- tuberculoid leprosy, but genotype ‘TT’ was associated with LLs leprosy
plication in macrophages [1]. Another example is the induction of Treg while the resistance in developing leprosy is due to the heterozygous
cells by Schistosoma mansoni egg (antigen w1) depend on vitamin A that genotype ‘Tt’. These data suggest that VDR and vitamin D play an im-
down-regulate Th1 and Th2 responses to control inflammatory and portant role in TB and leprosy. Therefore, the evaluation of vitamin D
autoimmune diseases. deficiency and polymorphism of its receptor can help to predict the
clinical evolution and its dietary role in leprosy. TT patients trigger a
9. Vitamin C in leprosy protective vitamin D-mediated antimycobacterial innate immune re-
sponse by the production of cathelicidin and defensin beta (DEFB)-4
Vitamin C (ascorbic acid) is an enzymatic co-factor in many meta- [33]. Conversely, an LL patient lacks activation of the vitamin D-
bolic processes, especially in oxidation-reduction processes; thus es- mediated pathway and reduced expression of defensins. Thus, further
sential for the prevention of infections, and its deficiency affects the investigation is required to determine whether any of these mediators
immune response [74]. Vitamin C also helps in the absorption of iron can act as biomarkers in leprosy diagnosis.
and inactivation of free radicals [75]. As a non-enzymatic antioxidant,
vitamin C helps to control ROS-induced inflammation while killing 11. Vitamin E in leprosy
intracellular bacteria like M. leprae; along with the damage of the im-
mune system through oxidative attacks. ROS-induced oxidative stress Free radicals and lipid peroxidation lead to the suppression of im-
can also damage the cell membrane integrity, with alteration in mem- mune response. Vitamin E, a fat-soluble antioxidant, protects the lipid
brane permeability and cell-to-cell communication, which also affects membranes against peroxidation. In influenza-infected rats, vitamin E
the host immune responses [62,76]. Supplementation of vitamin C rich diet showed increased production of IL-2 and IFN-γ suggesting that
(20 mg/kg and 1–3g per day) in the food can enhance the activity of higher intake of vitamin E improves the immune response against in-
neutrophils and macrophages to kill the microbes. It has been reported fections [87]. Vitamin E in its most active form α ± -tocopherol, react
that vitamin C treatment enhances the phagocytotic activity of murine with phospholipids of mitochondria, plasma membrane and endoplas-
macrophages [74]. Thus, deficiency of vitamin C adversely affects le- matic reticulum as the first defense against peroxidation of membrane
prosy patients, as evident from the reduced serum level of ascorbic acid phospholipids. The tocopherols help to neutralize these reactions by
in TB patients [77,78]. Moreover, supplementation of vitamin C donating phenol hydrogen to the peroxyl free radical (Fig. 6).
(1.0 gm ascorbate) and vitamin E (600 mg) improved the plasma anti- This inhibition of lipid peroxidation by vitamin E usually aided by
oxidant capacity of pulmonary TB patients after a month of MDT [79]. vitamin C to reduced glutathione and NADPH, as the higher con-
centration of vitamin E has a pro-oxidant effect. A case-control study of
10. Vitamin D in leprosy leprosy patients, at the beginning of treatment, showed increased oxi-
dative stress with higher levels of lipid peroxidation due to free radicals
Mycobacterium leprae and M. tb are intracellular bacteria with si- formed during the immune-mediated killing of M. leprae. However,
milar host defense, and recent studies showed that vitamin D deficiency leprosy patients treated with MDT (dapsone, rifampicin, and clofazi-
leads to an increased incidence of TB [70,80]. For long, vitamin D was mine) and 400 IU of E daily for 12 months showed normal LPO levels
considered to be essential for mineralization of bones, but after the [88] similar to TB [89,90]. Compared to healthy controls TB patients
discovery of functional vitamin D receptors (VDR) in tissues that are not showed reduced level of vitamin C and E, but higher serum level of
involved in calcium and phosphate metabolism, another role of vitamin Malondialdehyde [90], while pulmonary TB patients receiving standard
D has been accepted. The VDRs, present in many tissues and cells, elicit chemotherapy when supplemented with 140 mg of E and 200 μg of
a variety of biological responses including immunomodulatory activity selenium for two months showed reduced oxidative stress with im-
by modulating antigen presenting cells and T cell function [75,81]. proved antioxidant status than TB patients treated only with che-
Serum samples from TB susceptible individuals showed a low level of motherapy [89].
25-hydroxyvitamin D and less efficiency to cathelicidin peptide [82].
Moreover, vitamin D can induce the production of antimicrobial pep- 12. Role of zinc in leprosy
tides in infected macrophages that kill Mycobacterium (Fig. 5). These
peptides (cathelicidins and defensins) modulate innate immune re- Zinc is known to help in the prevention of infections, and its defi-
sponse [83]. The cathelicidin peptide LL-3 recruits monocytes, T-cells ciencies can negatively affect the immune system [74] due to poor Th1
and neutrophils to the infection site; while LL-37 activates macrophages response with a reduced level of pro-inflammatory cytokines (IFN-γ, IL-
by binding to TLR to induce M.tb killing. A comparative study on ca- 2, and TNF-α) that help to control intracellular pathogens including M.
thelicidin and vitamin D3 in 29 leprosy patients with 19 healthy in- leprae, without affecting the level of IL-4, IL-6, and IL-10 [91]. While
dividuals showed that cathelicidin level is lower in leprosy patients prolonged zinc supplementation increased the production of IL-2
than healthy individuals [84]. The IFN-γ, secreted by Th1 cells is known leading to significantly decreased incidence of respiratory infections
to have a protective role against intracellular pathogens, and can po- [91]. The antioxidant role of zinc is associated with the regulation of
tentiate the effect of 1α-hydroxylase enzyme that converts vitamin D cysteine-rich low molecular weight protein metallothionein expression
into its active form 1α, 25-(OH)2D3, along with the inhibition of an during radiation, drugs and heavy metal exposures [92]. Moreover, zinc

6
V.P. Dwivedi, et al. Microbial Pathogenesis 137 (2019) 103714

Fig. 5. Roles of Vitamin D during Mycobacterium leprae infection [19]. Vitamin D and tuberculosis. Nutr Rev. 67: 289-293.

is a structural and catalytic component of superoxide dismutase (SOD), recommendation of nutrients, and their doses for the prevention and
which reduces the oxidative effect of ROS, transforming superoxide treatment of diseases.
(O2-, +O2-, +2H+) to hydrogen peroxide (H2O2 + O2) to minimize
the chain reaction during cellular injury. The cytoplasmic SOD contains 14. Food and leprosy management
copper-zinc, while mitochondrial forms have manganese [93]. Thus,
the loss of zinc in the cellular membrane can affect its function, flow, Food and its availability is a significant factor for leprosy prevention
hydro-osmotic balance and sodium-calcium transport channels of the and management. The majority of leprosy patients are from low socio-
cell. Further, zinc can stabilize the reduced form of sulfhydryl groups, economic strata, having acute to chronic lifelong food insecurity,
and protect cell membranes from the effects of lipid peroxidation [94]. having economic, social to physical dimensions. Even if leprosy patients
A study showed that Zn deficiency in HIV infected males may con- have economic access to food they may not have physical and social
tribute to secondary infections that can lead to increased morbidity and access to quality and quantity of food during treatment [5]. Although
mortality [95]. Government provides food grains for families living below the poverty
line (BPL), leprosy patient must have their own resources for nutritional
13. Selenium and immune response security. As leprosy patients are usually nutrient deficient, so a further
nutritional deficiency can impair their cellular immunity to poor de-
Essential micronutrient trace element selenium is strongly linked fense against infections. Moreover, the risk of contracting subclinical M.
with complex enzymatic and metabolic functions in the living system, leprae infection could facilitate the progression from infection to clinical
and the most important is its interaction with glutathione peroxides manifestation. Therefore, in order to make MDT successful, it is a must
(GPxs). Glutathione catalyzes the reduction of hydrogen peroxide and to promote low-cost high protein balanced diet among leprosy patients
organic hydroperoxides and also protects cell membrane and cellular receiving MDT, and important to make them understand that MDT
lipids from oxidative injuries [96]. Selenium affects the chemotactic along with locally available high protein diet is essential for leprosy
and microbicidal activities of innate immune components and mod- cure. Here we have provided general dietary guidelines for boosting
ulates leukotriene synthesis and peroxide regulation in immune cells immunity including the diverse food groups and types for people with
[96,97]. However, an optimal dose of selenium is required for phago- or contracting leprosy to keep their immune system function with a
cytosis and lymphocytic activity, but higher doses are inhibitory. low-cost diet chart for sufferers and their household (Table 1).
Clinical studies showed that TB, asthma, and HIV patients have a se- To recommend a specific diet plan for leprosy patients, we have
lenium deficiency, but its effects on leprosy infection are unknown considered their socio-economic condition, day-to-day physical activ-
[98,99]. It has been observed that high doses of any single antioxidant ities, and food preferences. The guidelines will be more useful if foods
might induce an opposite effect. Thus, it is very important to consider are consumed in natural form. (i) Sprouted pulses: It is better to con-
the biochemical, clinical and genetic aspects during the sume germinated or sprouted pulses instead of cooking dry pulses,

7
V.P. Dwivedi, et al. Microbial Pathogenesis 137 (2019) 103714

Fig. 6. Role of ROS and Vitamin E during Mycobacterium leprae infection. Vijayaraghavan R, Suribabu CS, Sekar B, Oommen PK, Kavithalakshmi SN,
Madhusudhanan N (2005). Protective role of vitamin E on the oxidative stress in Hansen's disease (Leprosy) patients. Eur J Clin Nutr 59(10): 1121–1128.

Table 1
Food groups and types with a low-cost Diet Chart for Leprosy (Approximately 2000 Calorie; Protein: 60 gm; Fat: 25 gm).
Food Group Quantity (gm) Types of food

Cereal 370–400 Brown rice, wheat, Ragi, Jawar, Bajra


Pulses 50 Mixed pulses, whole pulses,
Vegetables 350 Locally grown: Green leafy vegetables, Tomatoes, Brinjal, Bitter gourt, Pumpkin etc.
Roots-tubers 100 Carrot, Onion, Potatoes, Sweet potato, Tapioca, Yam
Oil 20 Cooking with Mustard, Sesame or Ground nut oil
Diary/meat/Fish 50 Egg or small Fishes for non vegetarian patients
Molasses 20 Instead of sugar
Nuts 15 Ground Nut or other locally available Nuts
Fruits 150 Amla, Guava, Lemon, Banana, Jamun, Berries, Mango and other locally grown fruits

No. Meal Plan Benefits

1 Early Morning Luke warm water (200 ml) + Curcumin + Black Pepper Curcumin and pepper have unique health benefits. Black pepper in combination
with turmeric enhanced the antiseptic and antibacterial properties of turmeric
with ideal wound healing and cleansing activities. Pepper increases the wound
healing ability of turmeric due to better absorption at small doses (¼ to half tsp
and gradually increases to one tsp three times daily).
2 Breakfast (After half Missi Roti (3 Pcs)/Chira (60 gm)/Mixed Veg (1 Bowl Nutrient dense Breakfast (30% of daily requirement). Need attractive, healthy and
an hour) −100 gm) + Dahi (100 gm) + one Boiled Egg tasty meal with Protein, Vitamin and Mineral from different food groups.
3 Mid-Morning Fruits (seasonal −150 gm) Micronutrients work with macronutrients to keep the body functioning and
maintain energy level, metabolisms, cellular function, physical and mental
wellbeing.
4 Lunch Rice (150 gm), Dal (25 gm), G.L.V (50 gm); Veg (200 gm), Soya/ Healthy nutrient packed lunch helps organs and tissue to work effectively.
Paneer (40 gm) or Fish/Chicken (65 gm) Without good nutrition body is more prone to disease, infection, fatigue and poor
performance.
5 Afternoon Pumpkin seed/Mixed seeds (15 gm) Pumpkin seeds are rich in antioxidant, iron, zinc, magnesium, carotenoids,
vitamin E that help to protect against disease and inflammation. Nuts are high in
healthy fats, protein and fibre.
6 Evening Badam/Chhola/Chhatu (25 gm) Snacking help to fit extra nutrients into diet and prevent overeating at meal times.
7 Dinner Wheat/Rice (100 gm) Need enough food from different food types. Meal with a variety of food types
Dal (25 gm); Veg (100 gm) helps to satisfy satiety and supply nutritious.

**Drinking Water: 2–2.5 L (10–12 glasses).

8
V.P. Dwivedi, et al. Microbial Pathogenesis 137 (2019) 103714

because sprouted pulse contains diverse micronutrients (copper, iron, nutrition, particularly the balance of protein and micronutrients need to
magnesium, phosphorus, potassium), vitamins (Thiamine or B1, be investigated, as the disease takes a long time to develop, and thus the
Riboflavin or B2, Niacin or B3, Pyridoxine or B6, Folic acid), protein, nutritional balance may reduce the risk of acquiring the disease.
carbohydrate (low level) and fibers. One bowl of sprouted moong dal Moreover, a reduction in antioxidant and nutrient levels leads to in-
(Vigna radiata) contains 1 g fat and only 30 Calories with overall health creased oxidative stress, along with skin and neurological disorders
benefits. (ii) Yogurt: a fermented food helps in digestion and improves during M. leprae infection. Thus, further studies on the role of nutrients,
pH balance in intestine to provide better digestive health. The particularly immuno-modulatory nutrients need to be re-investigated to
Lactobacillus sp helps in forming omega 3 fatty acids, essential for elucidate pathogenic mechanisms. Collectively, this review highlighted
strengthening the immune system. (iii) Poppy and sesame seed: the current status of leprosy and its immune mechanism with the role of
Consumption of poppy seeds is a traditional food practice in India, to food and nutrition in its management. The supply of nutritious food to
get good quality and quantity of protein. Both of these seeds are rich in all members of a leprosy patient household is the most important factor
the essential minerals magnesium and calcium; help in converting LDL- along with related parameters including BMI, per capita food ex-
cholesterol to healthy HDL-cholesterol (iv) Zinc: Zinc has been used penditure, food storage, etc. Poverty alone leads to low consumption of
since ancient Egyptian times to enhance wound healing. Leprosy pa- protein foods, fruits, and vegetables leading to nutritional deficiency,
tients may be benefited from food rich in zinc. Normally meat, fish, egg, and prolonged nutritional deficiencies lead to an impaired immune
nuts, legumes and whole grains are good sources of zinc. Thus, these response, leading to clinical leprosy. Thus, future research should be
foods are helpful for leprosy patients to heal their ulcers faster. directed on the association of leprosy and nutrition, and the role of the
Although zinc is considered safe at the recommended dosages, more immunoregulatory pathway for leprosy management.
studies are required for a definitive conclusion [100]. (v) Omega-3 fatty
acid: It is a highly nutritious vital food that provides an essential Conflicts of interest
polyunsaturated fatty acid. Sea fish, flax seeds, soya bean, peanut, and
other nuts produce omega 3. Fish oil contains both forms of Omega 3: All authors have declared no conflict of interest.
docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA). Omega-
3 fatty acid may help to treat leprosy symptoms but well-designed Funding statement
studies with clear evidence are lacking. An omega-3 fatty acid is con-
sidered safe if taken in doses of recommended dietary allowance (RDA) There is no financial support for this study.
that also helps in blood clotting, making cell membranes, and prevent
inflammation [101]. Omega-3 and omega 6 mediators are found to be Acknowledgment
involved in the regulation of M. leprae-specific inflammatory and im-
mune responses, as these mediators are present in different forms of DC conceived this review and drafted the manuscript. VPD finalizes
leprosy-before and after MDT and during T1R as described in a recent the review with DC; BB helped in figure preparation. AS and MD help
review comparing the role of lipid mediators in leprosy including cy- DC in preparing the nutrition part, while AB, ID and SB help to compile
steinyl leukotrienes, leukotriene B4, prostaglandin E2 and D2, lipoxin this review. All authors revised, read and approved the final review.
A4 and resolvin D1 on erythema nodosum leprosum [101].
References
15. Discussion and conclusion
[1] E.S. Lima, A. Roland Ide, F. Maroja Mde, J.L. Marcon, Vitamin A and lipid per-
Leprosy displays a variety of clinical features with complex pre- oxidation in patients with different forms of leprosy, Rev. Inst. Med. Trop. Sao
Paulo 49 (4) (2007) 211–214.
sentation and multifactorial risk, thus its management is a huge chal- [2] WHO-SEARO/department of control of neglected tropical diseases, in:
lenge. The majority of individuals is neither infected nor develops le- E.A. Cooreman (Ed.), Operational Manual: Global Leprosy Strategy 2016-2020,
prosy following regular exposure due to their natural immunity. Even 2016, pp. 1–62.
[3] S. Oktaria, N.S. Hurif, W. Naim, H.B. Thio, T.E.C. Nijsten, J.H. Richardus, Dietary
people with latent M. leprae infection may develop only a single self- diversity and poverty as risk factors for leprosy in Indonesia: a case-control study,
healing lesion, but if untreated or unhealed it may progress into the PB PLoS Neglected Trop. Dis. 12 (3) (2018) e0006317.
or MB [102]. Recent studies showed that NOD2-mediated signaling [4] A. Alter, A. Grant, L. Abel, A. Alcais, E. Schurr, Leprosy as a genetic disease,
Mamm. Genome 22 (2011) 19–31.
[103], vitamin D-dependent antimicrobial pathways [104], interferons [5] World Health Organization, Global leprosy situation: update on the 2012 situa-
[105], T-regulatory Th-17/IL-17A/IL-17F cytokines, CD163 and ga- tion, Wkly. Epidemiol. Rec. 88 (2013) 365–380.
lectin-3 [106] have role in leprosy control. However, the intracellular [6] S.M. van Beers, M. Hatta, P.R. Klatser, Patient contact is the major determinant in
incident leprosy: implications for future control, Int. J. Lepr. Other Mycobact. Dis.
bacterium M. leprae could avoid immune surveillance, despite the in-
67 (1999) 119–128.
nate immune response by the skin epithelium and keratinocytes during [7] D.S. Ridley, W.H. Jopling, Classification of leprosy according to immunity. A five-
bacterial entry into the skin and peripheral nerves, while keratinocyte group system, Int. J. Lepr. Other Mycobact. Dis. 34 (1966) 255–273.
elicits an immune response by mannose-binding receptors, TLRs, cy- [8] F.J. Moet, D. Pahan, R.P. Schuring, L. Oskam, J.H. Richardus, Physical distance,
genetic relationship, age, and leprosy classification are independent risk factors for
tokines, chemokines and antimicrobial peptides [107]. Moreover, leprosy in contacts of patients with leprosy, J. Infect. Dis. 193 (2006) 346–353.
human keratinocytes have the phagocytic ability with the expression of [9] L.R. Kerr-Pontes, M.L. Barreto, C.M. Evangelista, L.C. Rodrigues, J. Heukelbach,
antimicrobial factors CD80, CD209, cathelicidin, TNF-α and IL-1β; and H. Feldmeier, Socioeconomic, environmental, and behavioural risk factors for le-
prosy in North-east Brazil: results of a case-control study, Int. J. Epidemiol. 35
M. leprae induce cytokines and chemokines in keratinocytes to kill the (2006) 994–1000.
bacterium with the help of LCs, DCs, T-cells, and neutrophils [108]. So [10] S.G. Feenstra, Q. Nahar, D. Pahan, L. Oskam, J.H. Richardus, Recent food shortage
the best way to prevent the spread of leprosy is early diagnosis and is associated with leprosy disease in Bangladesh: a case-control study, PLoS
Neglected Tropic. Dis. 5 (2011) e1029.
treatment. Immediate and annual examinations are necessary for at [11] I. Wagenaar, L. van Muiden, K. Alam, R. Bowers, Md A. Hossain, K. Kispotta,
least five years after last contact with an infectious patient with ade- J.H. Richardus, Diet-related risk factors for leprosy: a case-control study, PLoS
quate nutritious diets to the victim, family members, and close contacts Neglected Trop. Dis. 9 (5) (2015) e0003766.
[12] E.S. Wintergerst, S. Maggini, D.H. Hornig, Contribution of selected vitamins and
to improve/strengthen immunity. trace elements to immune function, Ann. Nutr. Metab. 51 (2007) 301–323.
Leprosy can serve as a spectral model of innate and adaptive im- [13] J.M. Pescarini, A. Strina, J. Silva Nery, L.M. Skalinski, K.V. Freitas de Andrade,
mune response so far protection and pathogenesis are concerned but are M.L.F. Penna, E.B. Brickley, L.C. Rodrigues, M.L. Barreto, G.O. Penna,
Socioeconomic risk markers of leprosy in high-burden countries: a systematic re-
relatively understudied in relation to its impact on nutritional factors.
view and meta-analysis, PLoS Neglected Trop. Dis. 12 (7) (2018) e0006622.
Leprosy is known to be influenced by genetic and environmental factors [14] J.S. Nery, S.M. Pereira, D. Rasella, M.L.F. Penna, R. Aquino, L.C. Rodrigues,
and the patient's reduced antioxidant status. The importance of M.L. Barreto, G.O. Penna, Effect of the Brazilian conditional cash transfer and

9
V.P. Dwivedi, et al. Microbial Pathogenesis 137 (2019) 103714

primary health care programs on the new case detection rate of leprosy, PLoS D. Chattopadhyay, G. Das, Early secreted antigen ESAT-6 of Mycobacterium tu-
Neglected Trop. Dis. 8 (11) (2014) e3357. berculosis promotes protective T helper 17 cell responses in a toll-like receptor-2-
[15] World Health Organization, WHO expert committee on leprosy, World Health dependent manner, PLoS Pathog. 7 (11) (2011) e1002378.
Organ Tech Rep Ser 968 (2012) 1–61. [46] V.P. Dwivedi, D. Bhattacharya, S. Chatterjee, D. Chattopadhyay, L. Van Kaer,
[16] S. Veena, P. Kumar, P. Shashikala, H. Gurubasavaraj, H.R. Chandrasekhar, W.R. Bishai, G. Das, Mycobacterium tuberculosis directs T helper 2 cell differ-
Murugesh, Significance of histopathology in leprosy patients with 1-5 skin lesions entiations by inducing interleukin-1β production in dendritic cells, J. Biol. Chem.
with relevance to therapy, J Lab Physicians 3 (1) (2011) 21–24. 287 (40) (2012) 33656–33663.
[17] C.A. Madigan, C.J. Cambier, K.M. Kelly-Scumpia, A. Saqasti, R.L. Modlin, [47] A. Hooij, A. Geluk, Immunodiagnostics for leprosy, in: D.M. Scollard, T.P. Gillis
L. Ramakrishnan, A macrophage response to Mycobacterium leprae phenolic gly- (Eds.), International Text Book of Leprosy, American Leprosy Missions, 2016,
colipid initiates nerve damage in leprosy, Cell 170 (5) (2017) P973–985E10. www.internationaltextbookofleprosy.org.
[18] M.I. Bakker, L. May, M. Hatta, A. Kwenang, P.R. Klatser, L. Oskam, J.J. Houwing- [48] G.T. Keusch, The history of nutrition: malnutrition, infection and immunity, J.
Duistermaat, Genetic, household and spatial clustering of leprosy on an island in Nutr. 133 (2003) 336S–340S.
Indonesia: a population-based study, BMC Med. Genet. 6 (2005) 40. [49] N.S. Scrimshaw, J.P. SanGiovanni, Synergism of nutrition, infection, and im-
[19] M.O. Moraes, C.C. Cardoso, P.R. Vanderborght, A.G. Pacheco, Genetics of host munity: an overview, AJCN (Am. J. Clin. Nutr.) 66 (1997) 464S–477S.
response in leprosy, Lepr. Rev. 77 (2006) 189–202. [50] S.Y. Oh, H.Y. Paik, D. Ju, Dietary habits, Food intake and functional outcomes in
[20] Wong SH, Hill AV, Vannberg FO, India-Africa-United Kingdom Leprosy Genetics C those with a history of Hansen's disease in Korea, Int. J. Lepr. Other Mycobact. Dis.
(2010). Genomewide association study of leprosy. N. Engl. J. Med. 362:1446- 66 (1) (1998) 34–42.
1447; 1447-1448.. [51] B. Diffey, M. Vaz, M.J. Soares, A.J. Jacob, L.S. Piers, The effect of leprosy-induced
[21] A. Alter, A. Grant, L. Abel, A. Alcais, E. Schurr, Leprosy as a genetic disease, deformity on the nutritional status of index cases and their household members in
Mamm. Genome 22 (2011) 19–31. rural South India: a socio-economic perspective, Eur. J. Clin. Nutr. 54 (2000)
[22] V. Shinde, P. Marcinek, D.S. Rani, S.R. Sunder, A. Sunder, S. Jain, S. Arun, S. Jain, 643–649.
I. Nath, T. Kumarasamy, T.P. Velavan, V. Valluri, Genetic evidence of TAP1 gene [52] T. Khadapani, B.K. Mishra, Health problems and nutritional status of selected le-
variant as a susceptibility factor in Indian leprosy patients, Hum. Immunol. 74 prosy victims of Burla town, Orissa, India, Curr. Res. J. Soc. Sci. 2 (2010) 350–357.
(2013) 803–807. [53] P.S.S. Rao, A.S. John, Nutritional status of leprosy patients in India, Indian J. Lepr.
[23] C. White, C. Franco-Paredes, Leprosy in the 21st century, Clin. Microbiol. Rev. 28 84 (2012) 17–22.
(2015) 80–94. [54] A. Ferguson, G.E. Griffin, Nutrition and the immune system, in: J.S. Garrow,
[24] D.M. Scollard, L.B. Adams, T.P. Gillis, J.L. Krahenbuhl, R.W. Truman, W.P.T. James, A. Ralph (Eds.), Human Nutrition and Dietetics, Elsevier Science
D.L. Williams, The continuing challenges of leprosy, Clin. Microbiol. Rev. 19 (2) Ltd, London, 2000, pp. 747–764.
(2006) 338-338. [55] H.H. Hirsch, G. Kaufmann, P. Sendi, M. Battegay, Immune reconstitution in HIV-
[25] M. Orlova, A. Cobat, N.T. Houng, N.N. Ba, N. Van Thuc, J. Spencer, Y. Nedelec, infected patients, Clin. Infect. Dis. 38 (2004) 1159–1166.
L. Barreiro, V.H. Thai, L. Abel, A. Alcais, E. Schurr, Gene set signature of reversal [56] S.D. Lawn, L.G. Bekker, R.F. Miller, Immune reconstitution disease associated with
reaction type 1 in leprosy patients, PLoS Genet. 9 (2013) e1003624. mycobacterial infections in HIV-infected individuals receiving antiretrovirals,
[26] R. Al-Mubarak, J. Vander Heiden, C.D. Broeckling, Serum metabolomics reveals Lancet Infect. Dis. 5 (2005) 361–373.
higher levels of polyunsaturated fatty acids in lepromatous leprosy: potential [57] W.J. Britton, D.N. Lockwood, Leprosy, Lancet 363 (9416) (2004) 1209–1219.
markers for susceptibility and pathogenesis, PLoS Neglected Trop. Dis. 5 (2011) [58] C.M. Passos Vázquez, R.S. Mendes Netto, K.B. Ferreira Barbosa, T.R. Rodrigues de
e1303. Moura, R.P. de Almeida, M.S. Duthie, A.R. Ribeiro de Jesus, Micronutrients in-
[27] J.J. Amaral, L.C. Antunes, C.S. de Macedo, K.A. Mattos, J. Han, J. Pan, fluencing the immune response in leprosy, Nutr. Hosp. 29 (1) (2014) 26–36.
A.L. Candéa, Md Henriques, M. Ribeiro-Alves, C.H. Borchers, E.N. Sarno, [59] C.G. Buffie, E.G. Pamer, Microbiota-mediated colonization resistance against in-
P.T. Bozza, B.B. Finlay, M.C. Pessolani, Metabonomics reveals drastic changes in testinal pathogens, Nat. Rev. Immunol. 13 (2013) 790–801.
anti-inflammatory/pro-resolving polyunsaturated Fatty acids-derived lipid med- [60] B. Alberts, A. Johnson, J. Lewis, M. Raff, K. Roberts, P. Walter, Biologia molecular
iators in leprosy disease, PLoS Neglected Trop. Dis. 7 (8) (2013) e2381. da célula (Molecular biology of the cell), fourth ed., Artmed; New York: Garland
[28] V.N. Britode Souza, A.M. Iyer, D.A. Lammas, B. Naafs, P.K. Das, Advances in le- Science, Porto Alegre, 2004.
prosy immunology and its field application: a gap to bridge, Clin. Dermatol. 34 (1) [61] S. Maggini, E.S. Wintergerst, S. Beveridge, D.H. Hornig, Selected vitamins and
(2016) 82–95. trace elements support immune function by strengthening epithelial barriers and
[29] D.N. Lockwood, L. Suneetha, K.D. Sagili, et al., Cytokine and protein markers of cellular and humoral immune responses, Br. J. Nutr. 98 (Suppl. 1) (2007)
leprosy reactions in skin and nerves: baseline results for the North Indian INFIR S29–S35.
cohort, PLoS Neglected Trop. Dis. 5 (2011) e1327. [62] P.C. Calder, A.A. Jackson, Under nutrition, infection and immune function, Nutr.
[30] A. Geluk, Biomarkers for leprosy: would you prefer T (cells)? Lepr. Rev. 84 (2013) Res. Rev. 13 (1) (2000) 3–29.
3–12. [63] Y. Shapira, N. Agmon-Levin, Y. Shoenfeld, Mycobacterium tuberculosis, auto-
[31] B. Beiguelman, The genetics of resistance to leprosy, Int. J. Lepr. 33 (1965) immunity, and vitamin D, Clin. Rev. Allergy Immunol. 38 (2–3) (2010) 169–177.
808–812. [64] R.K. Chandra, S. Kumari, Nutrition and immunity: an overview, J. Nutr. 124 (8
[32] A. Rotberg, Some aspects of immunity on leprosy and their importance in epide- Supl) (1994) 1433S–1435S.
miology, pathogenesis and classification of forms of the disease. Based on 1529 [65] R.M.N. Montenegro, E. Zandonade, M.D.C.B. Molina, L.M. Diniz, Reactional state
lepromin tested cases, Rev. Bras. Leprol. 5 (1937) 45–97. and nutritional profile among leprosy patients in the primary health care system,
[33] D. Montoya, R.L. Modlin, Learning from leprosy: insight into the human innate Greater Vitória, Espírito Santo State, Brazil, Cad. Saúde Pública 28 (1) (2012)
immune response, Adv. Immunol. 105 (2010) 1–24. 31–38.
[34] T.H. Ottenhoff, New pathways of protective and pathological host defense to [66] P. Jyothi, N. Riyaz, G. Nandakumar, M.P. Binitha, A study of oxidative stress in
mycobacteria, Trends Microbiol. 20 (2012) 419–428. paucibacillary and multibacillary leprosy, Indian J. Dermatol. Venereol. Leprol. 74
[35] L.C. Rodrigues, D.N. Lockwood, Leprosy now: epidemiology, progress, challenges, (1) (2008) 80.
and research gaps, Lancet Infect. Dis. 11 (2011) 464–470. [67] E. Azzini, A. Polito, A. Fumagalli, F. Intorre, E. Venneria, Mediterranean diet ef-
[36] R.L. Modlin, Th1-Th2 paradigm: insights from leprosy, J. Investig. Dermatol. 102 fect: an Italian picture, Nutr. J. 10 (2011) 125.
(1994) 828–832. [68] V.P. Palace, N. Khaper, Q. Qin, P.K. Singal, Antioxidant potentials of vitamin A
[37] L.S. Schlesinger, M.A. Horwitz, Phenolic glycolipid-1 of Mycobacterium leprae and carotenoids and their relevance to heart disease, Free Radic. Biol. Med. 26
binds complement component C3 in serum and mediates phagocytosis by human (5–6) (1999) 746–761.
monocytes, J. Exp. Med. 174 (1991) 1031–1038. [69] H. Siest, W. Stahl, Vitamins E and C, beta-carotene, and other carotenoids as an-
[38] I. Nath, C. Saini, V.L. Valluri, Immunology of leprosy and diagnostic challenges, tioxidants, Am. J. Clin. Nutr. 62 (6 Suppl) (1995) 1315S–1321S.
Clin. Dermatol. 33 (2015) 90–98. [70] Q. Wang, A. Ma, I.C. Bygbjerg, X. Han, Y. Liu, S. Zhao, J. Cai, Rationale and design
[39] M.L. Palermo, C. Pagliari, M.A. Trindade, T.M. Yamashitafuji, A.J. Duarte, of a randomized controlled trial of the effect of retinol and vitamin D supple-
C.R. Cacere, G. Benard, Increased expression of regulatory T cells and down-reg- mentation on treatment in active pulmonary tuberculosis patients with diabetes,
ulatory molecules in lepromatous leprosy, Am. J. Trop. Med. Hyg. 86 (2012) BMC Infect. Dis. 13 (2013) 104.
878–883. [71] H.D. Dawson, N.Q. Li, K.L. DeCicco, J.A. Nibert, A.C. Ross, Chronic marginal vi-
[40] K. Bobosha, L. Wilson, K.E. van Meijgaarden, Y. Bekele, M. Zewdie, J.J. van der tamin A status reduces natural killer cell number and function in aging Lewis rats,
Ploeg-van Schip, M. Abebe, J. Hussein, S. Khadge, K.D. Neupane, D.A. Hagge, J. Nutr. 129 (8) (1999) 1510–1517.
E.S. Jordanova, A. Aseffa, T.H. Ottenhoff, A. Geluk, T-cell regulation in lepro- [72] X. Wang, C. Allen, M. Ballow, Retinoic acid enhances the production of IL-10 while
matous leprosy, PLoS Neglected Trop. Dis. 8 (2014) e2773. reducing the synthesis of IL-12 and TNF-alpha from LPS-stimulated monocytes/
[41] P. Salgame, J.S. Abrams, C. Clayberger, H. Goldstein, J. Convit, R.L. Modlin, macrophages, J. Clin. Immunol. 27 (2) (2007) 193–200.
B.R. Bloom, Differing lymphokine profiles of functional subsets of human CD4 and [74] E.S. Wintergerst, S. Maggini, D.H. Hornig, Immune-enhancing role of vitamin C
CD8 T cell clones, Science 254 (5029) (1991) 279–282. and zinc and effect on clinical conditions, Ann. Nutr. Metab. 50 (2) (2006) 85–94.
[42] S. Sasaki, F. Takeshita, K. Okuda, N. Ishii, Mycobacterium leprae and leprosy: a [75] Y.B. Shaik-Dasthagirisaheb, G. Varvara, G. Murmura, A. Saggini, A. Caraffa,
compendium, Microbiol. Immunol. 45 (2001) 729–736. P. Antinolfi, S. Tete, Role of vitamins D, E and C in immunity and inflammation, J.
[43] C. Saini, M. Tarique, R. Rai, A. Siddiqui, N. Khanna, A. Sharma, T helper cells in Biol. Regul. Homeost. Agents 27 (2) (2013) 291–295.
leprosy: an update, Immunol. Lett. 184 (2017) 61–66. [76] B.N. Ames, M.K. Shigenaga, T.M. Hagen, Oxidants, antioxidants, and the degen-
[44] S. Chaitanya, M. Lavania, R.P. Turankar, S.R. Karri, U. Sengupta, Increased serum erative diseases of aging, Proc. Natl. Acad. Sci. U.S.A. 90 (17) (1993) 7915–7922.
circulatory levels of interleukin 17F in Type 1 reactions of leprosy, J. Clin. [77] V.V. Bakaev, A.P. Duntau, Ascorbic acid in blood serum of patients with pul-
Immunol. 32 (2012) 1415–1420. monary tuberculosis and pneumonia, Int. J. Tuberc. Lung Dis. 8 (2) (2004)
[45] S. Chatterjee, V.P. Dwivedi, Y. Singh, I. Siddiqui, P. Sharma, L.V. Kaer, 263–266.

10
V.P. Dwivedi, et al. Microbial Pathogenesis 137 (2019) 103714

[78] M.L. Plit, A.J. Theron, H. Fickl, C.E. van Rensburg, S. Pendel, R. Anderson, 16 (4) (2003) 433–441.
Influence of antimicrobial chemotherapy and smoking status on the plasma con- [94] R.J. Wood, Assessment of marginal zinc status in humans, J. Nutr. 130 (5S Suppl)
centrations of vitamin C, vitamin E, beta-carotene, acute phase reactants, iron and (2000) 1350S–1354S.
lipid peroxides in patients with pulmonary tuberculosis, Int. J. Tuberc. Lung Dis. 2 [95] H.I. Afridi, T.G. Kazi, N. Kazi, G.A. Kandhro, J.A. Baig, A.Q. Shah, Evaluation of
(7) (1998) 590–596. zinc in scalp hair and blood samples of tuberculosis and diarrhea male human
[79] J. Kowalski, B. Janiszewska-Drobinska, L. Pawlicki, T. Ceglinski, R. Irzmanski, immunodeficiency virus patients, Clin. Lab. 57 (3–4) (2011) 171–181.
Plasma antioxidative activity in patients with pulmonary TB, Pol. Merkur. Lek. 16 [96] S.J. Fairweather-Tait, Y. Bao, M.R. Broadley, R. Collings, D. Ford, J.E. Hesketh,
(92) (2004) 119–122. Selenium in human health and disease, Antioxidants Redox Signal. 14 (7) (2011)
[80] S.-J. Huang, X.-H. Wang, Z.-D. Liu, W.-L. Cao, Y. Han, A.-G. Ma, S.-F. Xu, Vitamin 1337–1383.
D deficiency and the risk of tuberculosis: a meta-analysis, Drug Des. Dev. Ther. 11 [97] J. Nève, Selenium in nutrition and therapeutics, Princ. Med. Biol. 8 (1997)
(2017) 91–102. 985–994.
[81] G. Siest, E. Jeannesson, J.B. Marteau, A. Samara, B. Marie, M. Pfister, [98] M. Van Lettow, C.E. West, J.W. van der Meer, F.T. Wieringa, R.D. Semba, Low
Transcription factor and drug-metabolizing enzyme gene expression in lympho- plasma selenium concentrations, high plasma HIV load and high IL-6 concentra-
cytes from healthy human subjects, Drug Metab. Dispos. 36 (1) (2008) 182–189. tions are risk factors associated with anemia in adults presenting with pulmonary
[82] P.T. Liu, S. Stenger, H. Li, L. Wenzel, B.H. Tan, S.R. Krutzik, Toll-like receptor tuberculosis in Zomba district, Malawi, Eur. J. Clin. Nutr. 59 (4) (2005) 526–532.
triggering of a vitamin D-mediated human antimicrobial response, Science 311 [99] K. Ramakrishnan, R. Shenbagarathai, K. Kavitha,
(5768) (2006) 1770–1773. P. Thirumalaikolundusubramanian, R. Rathinasabapati, Selenium levels in persons
[83] B. Rivas-Santiago, R. Hernandez-Pando, C. Carranza, E. Juarez, J.L. Contreras, with HIV/tuberculosis in India, Madurai City, Clin. Lab. 58 (1–2) (2012) 165–168.
D. Aguilar-Leon, Expression of cathelicidin LL-37 during Mycobacterium tubercu- [100] A.P. Ustianowski, D.N. Lockwood, Leprosy: current diagnostic and treatment
losis infection in human alveolar macrophages, monocytes, neutrophils, and epi- approaches, Curr. Opin. Infect. Dis. 16 (5) (2003) 421–427.
thelial cells, Infect. Immun. 76 (3) (2008) 935–941. [101] C.S. Macedo, F.M. Carvalho, J.J. Amaral, S.M. Ochs, E.F. Assis, E.N. Sarno,
[84] M. Matzner, A.R. Al Samie, H.M. Winkler, J. Nemeth, A. Grasnek, A. Indra, Low P.T. Bozza, M.C. VidalPessolani, Leprosy and its reactional episodes: serum levels
serum levels of cathelicidin LL-37 in leprosy, Acta Trop. 117 (1) (2011) 56–59. and possible roles of omega-3 and omega-6-derived lipid mediators, Cytokines 112
[85] A.R. Martineau, K.A. Wilkinson, S.M. Newton, R.A. Floto, A.W. Norman, (2018) 87–94.
K. Skolimowska, IFN-gamma- and TNF-independent vitamin D-inducible human [102] R.R. Jacobson, J.L. Krahenbuhl, Leprosy, Lancet 353 (1999) 655–660.
suppression of mycobacteria: the role of cathelicidin LL-37, J. Immunol. 178 (11) [103] M.G. Netea, B.J. Kullberg, J.W. van der Meer, Genome wide association study of
(2007) 7190–7198. leprosy, N. Engl. J. Med. 362 (2010) 1447–1448.
[86] J. Mora, M. Iwata, U. Andrian, Vitamin effects on the imune system: vitamins A [104] P.T. Liu, M. Wheelwright, R. Teles, E. Komisopoulou, K. Edfeldt, B. Ferguson,
and D take centre stage, Nat. Rev. Immunol. 8 (2008) 685–698. M.D. Mehta, A. Vazirnia, T.H. Rea, E.N. Sarno, T.G. Graeber, R.L. Modlin,
[87] M. Meydani, Nutrition, immune cells, and atherosclerosis, Nutr. Rev. 56 (1Pt 2) MicroRNA-21 targets the vitamin D-dependent antimicrobial pathway in leprosy,
(1998) S177–S182. Nat. Med. 18 (2) (2012) 267–273.
[88] R. Vijayaraghavan, C.S. Suribabu, B. Sekar, P.K. Oommen, S.N. Kavithalakshmi, [105] R.M. Teles, T.G. Graeber, S.R. Krutzik, D. Montoya, M. Schenk, D.J. Lee,
N. Madhusudhanan, Protective role of vitamin E on the oxidative stress in Hansen's E. Komisopoulou, K. Kelly-Scumpia, R. Chun, S.S. Iyer, E.N. Sarno, T.H. Rea,
disease (Leprosy) patients, Eur. J. Clin. Nutr. 59 (10) (2005) 1121–1128. M. Hewison, J.S. Adams, S.J. Popper, D.A. Relman, S. Stenger, B.R. Bloom,
[89] E. Seyedrezazadeh, A. Ostadrahimi, S. Mahboob, Y. Assadi, J. Ghaemmagami, G. Cheng, R.L. Modlin, Type I interferon suppresses type II interferon-triggered
M. Pourmogaddam, Effect of vitamin E and selenium supplementation on oxida- human anti-mycobacterial responses, Science 339 (6126) (2013) 1448–1453.
tive stress status in pulmonary tuberculosis patients, Respirology 13 (2) (2008) [106] A. Polycarpou, S.L. Walker, D.N. Lockwood, New findings in the pathogenesis of
294–298. leprosy and implications for the management of leprosy, Curr. Opin. Infect. Dis. 26
[90] M. Lamsal, N. Gautam, N. Bhatta, B.D. Toora, S.K. Bhattacharya, N. Baral, (2013) 413-456 419.
Evaluation of lipid peroxidation product, nitrite and antioxidant levels in newly [107] T. Mutis, M. De Bueger, A. Bakker, T.H.H.L.A. Ottenhoff, Class II+ human kera-
diagnosed and two months follow-up patients with pulmonary tuberculosis, tinocytes present Mycobacterium leprae antigens to CD4+ Th1-like cells, Scand. J.
Southeast Asian J. Trop. Med. Public Health 38 (4) (2007) 695–703. Immunol. 37 (1993) 43–51.
[91] A.S. Prasad, Effects of zinc deficiency on Th1 and Th2 cytokine shifts, J. Infect. Dis. [108] E.C. Lyrio, I.C. Campos-Souza, L.C. Corrêa, G.C. Lechuga, M. Verícimo,
182 (Suppl. 1) (2000) S62–S68. H.C. Castro, S.C. Bourguignon, S. Côrte-Real, N. Ratcliffe, W. Declercq,
[92] M. Wolfgang, The function of zinc metallothionein: a link between cellular zinc D.O. Santos, Interaction of Mycobacterium leprae with the HaCaT human kerati-
and redox state, J. Nutr. 130 (2000) 1455–1458. nocyte cell line: new frontiers in the cellular immunology of leprosy, Exp.
[93] J.C. Koury, C.M. Donangelo, Zinc, oxidative stress and physical activity, Rev. Nutr. Dermatol. 24 (7) (2015) 536–542.

11

You might also like