Skip to main content
Advertisement
  • Loading metrics

Lactoferrin is a dynamic protein in human melioidosis and is a TLR4-dependent driver of TNF-α release in Burkholderia thailandensis infection in vitro

  • Shelton W. Wright,

    Roles Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Writing – original draft, Writing – review & editing

    Affiliation Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, United States of America

  • Lara Lovelace-Macon,

    Roles Data curation, Writing – review & editing

    Affiliation Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States of America

  • Deirdre Ducken,

    Roles Data curation, Writing – review & editing

    Affiliation Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States of America

  • Sarunporn Tandhavanant,

    Roles Data curation, Methodology, Writing – review & editing

    Affiliation Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand

  • Prapit Teparrukkul,

    Roles Methodology, Writing – review & editing

    Affiliation Department of Internal Medicine, Sunpasitthiprasong Hospital, Ubon Ratchathani, Thailand

  • Viriya Hantrakun,

    Roles Data curation, Methodology, Writing – review & editing

    Affiliation Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand

  • Direk Limmathurotsakul,

    Roles Formal analysis, Funding acquisition, Supervision, Writing – review & editing

    Affiliations Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand

  • Narisara Chantratita,

    Roles Data curation, Funding acquisition, Methodology, Supervision, Writing – review & editing

    Affiliations Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand

  • T. Eoin West

    Roles Conceptualization, Formal analysis, Funding acquisition, Methodology, Supervision, Writing – review & editing

    tewest@uw.edu

    Affiliation Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States of America

Abstract

Melioidosis is an often-severe tropical infection caused by Burkholderia pseudomallei (Bp) with high associated morbidity and mortality. Burkholderia thailandensis (Bt) is a closely related surrogate that does not require BSL-3 conditions for study. Lactoferrin is an iron-binding glycoprotein that can modulate the innate inflammatory response. Here we investigated the impact of lactoferrin on the host immune response in melioidosis. Lactoferrin concentrations were measured in plasma from patients with melioidosis and following ex vivo stimulation of blood from healthy individuals. Bt growth was quantified in liquid media in the presence of purified and recombinant human lactoferrin. Differentiated THP-1 cells and human blood monocytes were infected with Bt in the presence of purified and recombinant human lactoferrin, and bacterial intracellular replication and cytokine responses (tumor necrosis factor-α (TNF-α), interleukin-1β and interferon-γ) were measured. In a cohort of 49 melioidosis patients, non-survivors to 28 days had significantly higher plasma lactoferrin concentrations compared to survivors (median (interquartile range (IQR)): 326 ng/ml (230–748) vs 144 ng/ml (99–277), p<0.001). In blood stimulated with heat-killed Bp, plasma lactoferrin concentration significantly increased compared to unstimulated blood (median (IQR): 424 ng/ml (349–479) vs 130 ng/ml (91–214), respectively; p<0.001). Neither purified nor recombinant human lactoferrin impaired growth of Bt in media. Lactoferrin significantly increased TNF-α production by differentiated THP-1 cells and blood monocytes after Bt infection. This phenotype was largely abrogated when Toll-like receptor 4 (TLR4) was blocked with a monoclonal antibody. In sum, lactoferrin is produced by blood cells after exposure to Bp and lactoferrin concentrations are higher in 28-day survivors in melioidosis. Lactoferrin induces proinflammatory cytokine production after Bt infection that may be TLR4 dependent.

Author summary

Melioidosis is a severe tropical infection caused by the bacterium Burkholderia pseudomallei. Despite antibiotics, mortality in some regions remains very high, necessitating the need for alternative treatment strategies, including targeting the immune system. Lactoferrin is an iron-binding protein with a variety of different functions. In this study, we wanted to test whether lactoferrin alters how the immune system responds during melioidosis. To achieve this, we first tested the blood of melioidosis patients and found that patients who later died had higher lactoferrin levels compared to those who survived. We also stimulated blood obtained from healthy individuals with B. pseudomallei and found that lactoferrin levels increase. We next analyzed whether lactoferrin impaired how the bacteria grows and found that the growth of Burkholderia thailandensis, a closely related bacterium, was not affected by the addition of lactoferrin to the media. When human immune cells, called monocytes, were infected with B. thailandensis, we found that levels of a specific inflammatory protein, TNF-α, increased after adding lactoferrin and that this effect was related to a specific immune recognition pathway called Toll-like receptor 4. These findings provide new data about the role of lactoferrin in modulating the immune response in melioidosis.

Introduction

Melioidosis is a tropical bacterial infection which can cause pneumonia and sepsis. The etiologic agent of melioidosis, Burkholderia pseudomallei (Bp), a Gram-negative saprophytic bacteria, causes disease following inoculation by inhalation, ingestion or cutaneous exposure [1]. Endemic to southeast Asia and parts of northern Australia, melioidosis has mortality rates upwards of 40% in certain regions, including northeast Thailand, despite appropriate antibiotic therapy [2,3]. Due to the difficulty in eradicating the facultatively intracellular pathogen, treatment regimens last several months [1]. Given its pathogenicity, Bp is classified as a Tier 1 select agent by the US Centers for Disease Control and Prevention. Importantly, the worldwide prevalence of melioidosis is unknown and may represent a burgeoning global threat with over 160,000 cases worldwide annually [4].

Lactoferrin is an iron-binding glycoprotein typically found in human milk, saliva and other mucosal secretions and is produced by multiple cells, including neutrophils, during inflammation [5,6]. This apparent enrichment in secretions has been postulated to be related to lactoferrin’s broad antimicrobial properties [7]. Historically, these defense mechanisms were ascribed to lactoferrin’s iron sequestration properties [8,9]. However, more recent evidence suggests that lactoferrin may play a direct antimicrobial role by preventing bacterial biofilm formation, activating proteolysis of bacterial virulence factors and blocking the adhesion of bacteria to host immune cell membranes [6,10]. In fact, lactoferrin’s non-iron binding protein domains have exhibited frequent evolvement of polymorphisms, suggesting an importance of lactoferrin’s other moieties [11]. Taking advantage of these properties, bovine lactoferrin, sharing 77% amino acid homology with human lactoferrin, has been studied as an adjuvant therapy in several clinical trials for patients with sepsis [12,13].

Iron metabolism may play a role in Bp’s virulence as iron supplementation enhances bacterial growth in soil and Bp-infected mice [14,15]. Bp can also capture iron from lactoferrin and transferrin in vivo through a specific siderophore, malleobactin [16]. However, in contrast to several other Gram-negative bacteria, lactoferrin supplementation does not impede Bp growth in media [17]. Lactoferrin may also directly regulate innate immunity by interacting with intracellular signaling pathways, including NF-κB [1820]. The immune response to Bp infection is dependent on multiple Toll-like receptor (TLR) pathways, including TLR4 [21]. However, whether lactoferrin regulates the immune response during Bp infection is unknown.

We hypothesized that human lactoferrin modulates the innate immune response in melioidosis. We initially tested our hypothesis by measuring plasma lactoferrin in patients with melioidosis as well as Bp-stimulated blood of healthy individuals. We then assessed the immunomodulatory effects of lactoferrin experimentally in vitro using B. thailandensis (Bt). Bt is a closely related bacterium to Bp which does not require high containment laboratory conditions but induces a similar innate inflammatory response in vivo and in vitro [2224].

Methods and materials

Ethics statement

The Ethical Review Committee for Research in Human Subjects, Ministry of Public Health, Thailand; the Ethics Committee of the Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand (MUTM2012-024-01 and MUTM2015-002-01); the Ethical Review Committee for Research in Human Subjects, Sunpasitthiprasong Hospital, Ubon Ratchathani, Thailand (039/2556); the Oxford Tropical Medicine Ethics Committee, Oxford UK (OXTREC172-12); and the University of Washington Human Subjects Division Institutional Review Board (42988) approved the studies involving human subjects research. Written informed consent for enrollment in the clinical studies was obtained from subjects or their representatives at the time of enrollment.

Melioidosis patient cohort

Subjects aged 18 years or older admitted to Sunpasitthiprasong Hospital, Ubon Ratchathani, Thailand with suspected infection were prospectively enrolled between 2013 through 2017. Inclusion criteria were enrollment within the first 24 hours of admission to the hospital and the presence of at least three documented systemic manifestations of infection, as proposed by the 2012 Surviving Sepsis Campaign [25]. Patients suspected of having a hospital-acquired infection, those hospitalized in the 30 days prior to the current hospitalization or those transferred from another healthcare facility after more than 72 hours were excluded. This cohort, and subsets of it, have previously been described [2629]. For this study, we analyzed plasma, obtained at the time of enrollment, from the subset of 49 subjects with melioidosis, defined by any growth of Bp in any clinical specimen submitted for culture, recruited during the first year of the study. Subjects were followed until 28 days after enrollment to determine survival.

Healthy donor whole blood stimulation

Three hundred healthy Thai adults presenting to donate blood at Sunpasitthiprasong Hospital, Ubon Ratchathani, Thailand were recruited. After subjects provided informed consent, a peripheral blood sample was obtained and stimulated as described previously [30]. For this study, we analyzed blood stimulated with heat-killed Bp and unstimulated blood. Blood was mixed 1:1 with RPMI media and 380μl were added to a pre-prepared 96-well plate containing 20μl heat-killed Bp K96243 (to achieve a final concentration of 2.5 x 106 CFU/ml) or 20μl media. Plates were then incubated for 6 hours at 37°C under 5% CO2 before being spun down, and plasma was removed and frozen.

Lactoferrin assay

Frozen plasma samples were thawed and human lactoferrin concentrations measured by ELISA (EMD Millipore, San Diego, CA) according to the manufacturer’s directions. Lactoferrin levels in all samples were within the manufacturer’s stated range of detection.

Bacterial culture

B. thailandensis E264 [24] was cultured at 1 x 103 CFU/ml at 37°C in either Luria broth (LB), tryptic soy broth (TSB) or M9, minimal salt, media. LB media was prepared by mixing 20g of Difco LB broth (BD Biosciences, Franklin Lakes, NJ) with 1 liter of water. TSB media was prepared by mixing 30g of TSB broth (Sigma-Aldrich, St. Louis, MO) with 1 liter of water and 50 mM glutamate and 1% glycerol. M9 media was prepared by combining 6g Na2HPO4, 3g KH2PO4, 0.5g NaCl, 1g NH4Cl, 20 ml 20% glucose, 1ml 1M MgSO4, 0.1 ml 1M CaCl2 in 1L of water [31]. Partially iron-saturated recombinant human lactoferrin (rhLTF; L4040, 0.07% Fe saturation, expressed in rice, Sigma-Aldrich, St. Louis, MO) or partially saturated purified human lactoferrin (phLTF; L0520, 0.006% Fe saturation, purified from human milk, Sigma-Aldrich) were added to the culture media at varying concentrations. Bacterial colony forming units in the broth were quantified using serial dilutions at 3, 6 and 24 hours after inoculation.

Cell culture

Human monocytic THP-1 cells were purchased and cultured according to manufacturer specifications (ATCC TIB-202; Manassas, VA). THP-1 cells were cultured in RPMI 1640 with 10% FBS, 1% L-glutamine, 1% HEPES and 0.05 mM 2-mercaptoethanol. For differentiation to macrophages, THP-1 cells were stimulated with 100 nM Vitamin D3 for 24 hours in a 48-well polystyrene plate at a density of 2 x 105 cells/well prior to infection.

Peripheral blood samples were obtained from healthy subjects at Harborview Medical Center, Seattle, USA. Peripheral blood was collected in three 8 ml sodium citrate-containing vacutainers (BD Biosciences, Franklin Lakes, NJ). Peripheral blood monocytes were isolated with centrifugation within 1 hour of collection. Monocyte isolation was performed using a microbead monocyte separation kit (Monocyte Isolation Kit II) according to the manufacturer’s specifications (Miltenyi Biotec, Bergisch Gladbach, Germany). Monocytes were then plated on a 48-well polystyrene plate at 2x105 cells/well, suspended in RPMI 1640 with 10% fetal bovine serum (FBS), 1% L-glutamine and 1% 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) overnight prior to infection.

Monocyte infection

Both peripheral blood monocytes and differentiated THP-1 cells were infected with Bt at a multiplicity of infection (MOI) of 1 and 10 in 48-well plates. Plates were centrifuged for 10 minutes at 300 x g and incubated at 37°C for 6 hours. At 1 hour after infection, rhLTF or phLTF were added at 100 μg/ml or 500 μg/ml. At 6 hours, supernatants were removed and stored at -30°C pending protein measurement. In samples designated for assessing the role of TLR4, 20 μg/ml of monoclonal mouse IgG1 for TLR4 or IgG1 isotype control (InvivoGen, San Diego, CA) were added to cell cultures one hour prior to infection. As a control condition, designated cells were exposed to either 10 or 50 ng/ml of E. coli K12-derived LPS (Ultrapure LPS-EK, InvivoGen, San Diego, CA). In samples designated for intracellular bacterial measurement, at 1 hour after infection 100 μg/ml of kanamycin was added. At 6 hours after infection, cells were washed with Hank’s balanced salt solution and lysed with 0.1% Triton-X100 (Promega, Madison, WI) and lysed supernatants were plated and incubated at 37°C for 24 hours, at which point colonies were counted. The endotoxin concentration of the rhLTF and phLTF was quantified by a limulus amebocyte lysate assay (Thermo Scientific, Waltham, MA). The final concentration of endotoxin from lactoferrin within each cell culture was determined to be 0.02 endotoxin units/ml for both rhLTF and phLTF conditions. In samples designated for cytokine measurement, uninfected cells were additionally exposed to either media or 100 μg/ml of rhLTF or phLTF, in order to determine effects from direct stimulation by lactoferrin supplementation.

Cytokine assays

Concentrations of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interferon-γ (IFN- γ) were measured in cell supernatants by ELISA according to manufacturer specifications (R&D, Minneapolis, MN). Absorbance of each sample was determined at 450 nm by microplate reader and final cytokine levels were determined using a standard curve for each experiment.

Statistical analysis

Combined data following a normal distribution are reported as mean ± SD. The Student’s t-test or ANOVA was used to compare groups with normally distributed data. Data following a non-normal distribution are presented as medians with interquartile range (IQR). Correlations of non-normal data were assessed by Spearman’s correlation coefficient. Lactoferrin concentrations were log10-transformed for statistical analysis. Comparison of means between individuals were performed using the paired t-test and between unpaired groups using an unpaired t-test. Categorical data were analyzed using the chi-square test. Analyses were performed using Stata version 14.2 (College Station, TX) or GraphPad Prism (San Diego, CA). A two-sided P-value ≤ 0.05 was considered to be significant.

Results

Blood lactoferrin concentrations are associated with survival in melioidosis and increase after Bp exposure ex vivo

To quantify lactoferrin levels in human melioidosis and determine whether they differ based on clinical outcome, we measured plasma lactoferrin concentrations in 49 patients with melioidosis. Subject characteristics are listed in Table 1. Non-survivors to 28 days had significantly higher lactoferrin concentrations compared to survivors (median (IQR): 326 ng/ml (230–748) vs 144 ng/ml (99–277), p<0.001) (Fig 1). As neutrophils can produce lactoferrin, we evaluated whether neutrophil count was correlated with lactoferrin concentration. Neutrophil count was not significantly higher in non-survivors compared to survivors (p = 0.06) and was only moderately correlated with lactoferrin concentration in all subjects with melioidosis (Spearman’s rank correlation coefficient 0.43, p<0.01).

thumbnail
Fig 1. Plasma lactoferrin concentrations are higher in patients who do not survive melioidosis.

Plasma lactoferrin concentrations, obtained at study enrollment, in survivors (N = 19) compared to 28-day non-survivors (30) of melioidosis. Data presented as individual values with median and IQR. Comparisons are made following log10-transformation of data using unpaired t-test.

https://doi.org/10.1371/journal.pntd.0008495.g001

To evaluate whether blood cells produce lactoferrin in response to Bp, we compared lactoferrin concentrations in whole blood from 26 healthy individuals stimulated ex vivo with heat-killed Bp to concentrations in unstimulated blood from the same individuals. The average lactoferrin concentration in Bp-stimulated blood was significantly higher compared to the average concentration in unstimulated blood (median (IQR): 424 ng/ml (349–479) vs 130 ng/ml (91–214), respectively; p<0.001) (Fig 2). The median neutrophil count in the healthy subject cohort was 3,000 with an IQR of 2,200–3,600. Neutrophil count was not correlated with lactoferrin concentration before stimulation (Spearman’s rank correlation coefficient 0.12, p = 0.33) but was more strongly correlated after stimulation (Spearman’s rank correlation coefficient 0.67, p<0.01). These results taken together suggest that early plasma lactoferrin levels are increased in melioidosis patients who do not survive to 28 days, and that Bp induces lactoferrin production by blood cells.

thumbnail
Fig 2. Lactoferrin is produced by blood cells of healthy individuals after stimulation with Bp.

Plasma lactoferrin concentrations of matched, unstimulated (naïve) blood from healthy individuals and blood stimulated with heat-killed Bp K96243 (hkBp) at 2.5 x 106 CFU/ml (N = 26). Data presented as individual values with median and IQR. Comparisons are made following log10-transformation of data using paired t-test.

https://doi.org/10.1371/journal.pntd.0008495.g002

Lactoferrin does not impair Bt growth in culture

Lactoferrin has iron-sequestering capabilities that inhibit bacterial growth as well as microbicidal effects against some bacteria [32]. In order to evaluate whether lactoferrin impairs bacterial growth, Bt was cultured in liquid media in the presence of rhLTF. To account for lactoferrin’s ability to sequester iron, three different medias were used: LB, minimal M9 and TSB media. For each of the three medias used, the addition of rhLTF up to 100 μg/ml did not consistently impair growth of Bt over 24 hours (Fig 3A–3C). In selected experiments, rhLTF concentrations of 500–1000 μg/ml did not impair Bt growth in any media. Similarly, the addition of phLTF (Fig 3D) showed no reduction in Bt growth in TSB media at 24 hours.

thumbnail
Fig 3. Lactoferrin does not impair growth of B. thailandensis in media.

In the presence of varying concentrations of rhLTF, B. thailandensis (Bt) growth was quantified over 24 hours in A) LB, B) M9 or C) TSB media. In the presence of varying concentrations of phLTF, Bt growth was quantified over 24 hours in D) TSB media. Each graph is representative of at least two independent experiments.

https://doi.org/10.1371/journal.pntd.0008495.g003

Lactoferrin does not inhibit intracellular replication of Bt in differentiated THP-1 cells or blood monocytes

Bp and Bt are facultative intracellular pathogens. This intracellular lifestyle may be an important contributor to subversion or avoidance of the host response and eradication of the bacterium [33]. To determine whether lactoferrin alters intracellular bacterial replication in monocytic cells, we infected differentiated THP-1 cells with Bt at an MOI of 10. After one hour either 100 μg/ml of rhLTF or phLTF was added and an antibiotic protection assay was performed. Six hours after infection, no differences in intracellular Bt counts were noted between cells exposed to 100 μg/ml of either rhLTF or phLTF compared to control cells (Fig 4A). We repeated this experiment infecting fresh human peripheral blood monocytes with Bt at an MOI of 10. We observed no difference in the six-hour intracellular Bt counts between cells treated with rhLTF, phLTF or control cells (Fig 4B). Therefore, supplemental lactoferrin does not alter intracellular replication of Bt in differentiated THP-1 cells or primary blood monocytes.

thumbnail
Fig 4. Lactoferrin does not alter intracellular replication of Bt in differentiated THP-1 cells or blood monocytes.

Differentiated THP-1 cells (A) and primary blood monocytes (B) were infected with Bt at an MOI of 10 and 1 hour later were treated with either media or 100 μg/ml of rhLTF or phLTF, along with kanamycin (100 μg/ml). After 6 hours, cells were washed, lysed and intracellular bacteria were cultured and quantified in CFU/ml at 24 hours. Means ± standard deviations of duplicate or triplicate conditions are displayed. rhLTF vs media and phLTF vs media groups were compared by t-test (p>0.05 for all comparisons). One representative example of two or three independent experiments is shown.

https://doi.org/10.1371/journal.pntd.0008495.g004

Lactoferrin increases TNF-α production by monocytic cells infected with Bt

To test whether lactoferrin modulates monocytic cell cytokine production in response to Bt infection, differentiated THP-1 cells were infected with Bt at an MOI of 1 or an MOI of 10. After one hour, cells were treated with either rhLTF or phLTF. Levels of TNF-α, IL-1β and IFN-γ in cell supernatants were measured by ELISA at 6 hours. At an MOI of 1, infected differentiated THP-1 cells exposed to rhLTF produced significantly more TNF-α compared to infected cells without lactoferrin (p = 0.01; Fig 5A). At an MOI of 10, infected differentiated THP-1 cells exposed to rhLTF or phLTF produced significantly more TNF-α compared to unexposed infected cells (p = 0.01, p = 0.008, respectively; Fig 5A). This effect did not appear to be related to direct stimulation from either rhLTF or phLTF as uninfected cells exposed to either media, rhLTF or phLTF had TNF-α concentrations below the level of detection for the assay. No differences in IL-1β (Fig 5B) or IFN-γ expression (S1 Fig) were noted following differentiated THP-1 cell infection.

thumbnail
Fig 5. Lactoferrin increases proinflammatory cytokine production by differentiated THP-1 cells or blood monocytes infected with Bt.

Differentiated THP-1 cells (A-B) and primary blood monocytes (C) were infected with Bt at an MOI of 1 (THP-1 cells) and an MOI of 10 (THP-1 cells and primary blood monocytes), and 1 hour later were treated with either media, or 100 μg/ml of rhLTF or phLTF. At 6 hours after infection, cell supernatants were collected and TNF-α (A and C) or IL-1β (B) were measured by ELISA. Means ± standard deviations of duplicate or triplicate conditions are displayed. rhLTF vs media and phLTF vs media groups were compared for each MOI by t-test (* p<0.05; ** p<0.01); The concentrations of cytokines in uninfected THP-1 cells, for all conditions, were below the level of detection. One representative example of two or three independent experiments is shown.

https://doi.org/10.1371/journal.pntd.0008495.g005

The effect of lactoferrin on cytokine production was next tested in human primary blood monocytes infected with Bt. Similar to the response noted in infected differentiated THP-1 cells, blood monocytes infected with Bt at MOI 10 produced significantly more TNF-α when exposed to either rhLTF (p = 0.02) or phLTF (p = 0.01) compared to no lactoferrin (Fig 5C). While uninfected peripheral blood monocytes had low detectable levels of TNF-α, no difference in concentration was noted between cells exposed to rhLTF or phLTF (S2 Fig). Together, these observations indicate that in monocytic cells infected with Bt, lactoferrin promotes TNF-α production.

The effect of lactoferrin on TNF-α is partially dependent on TLR4 in Bt monocyte infection

Lactoferrin may modulate LPS-TLR4 pathways by direct binding of LPS and by nuclear inhibition of NF-κB activation [18,19,34,35]. However, the lactoferrin-LPS complex may also be proinflammatory under certain conditions [36]. In order to determine whether lactoferrin-induced activation of TLR4 may be responsible for the increased TNF-α production by monocytic cells noted in the presence of lactoferrin, we investigated whether the addition of a TLR4-blocking antibody (TLR4-ab) attenuates production of this cytokine. Differentiated THP-1 cells were exposed to either 20 μg/ml of TLR4-ab or an IgG isotype control prior to infection by Bt and subsequent treatment with lactoferrin. In control experiments, the addition of TLR4-ab markedly reduced TNF-α production after differentiated THP-1 cells were exposed to E. coli K12-derived LPS, with or without rhLTF or phLTF treatment (Fig 6A). Following infection of differentiated THP-1 cells with Bt at an MOI of 1, treatment with TLR4-ab–compared to an isotype control–significantly reduced supernatant TNF-α concentrations in the presence of rhLTF (p = 0.03) or phLTF (p = 0.008), but not media alone (Fig 6B). A comparable effect was noted for Bt infection at an MOI of 10 (Fig 6B). The TLR4-ab-dependent reduction in TNF-α after lactoferrin treatment approximated the baseline TNF-α levels in infected cells not treated with lactoferrin. These results suggest that TLR4 contributes substantially to the lactoferrin-dependent induction of TNF-α production during Bt infection.

thumbnail
Fig 6. Lactoferrin-induced TNF-α production following Bt infection of differentiated THP-1 cells is attenuated by blockade of TLR4.

Differentiated THP-1 cells were exposed to E. coli K12 LPS at 10 ng/ml or 50 ng/ml (A) or infected with Bt at an MOI of 1 and 10 (B). At one hour, cells were treated with media or with 100 μg/ml of either rhLTF or phLTF and concurrently with either 20 μg/ml of TLR4-ab or IgG isotype control. After six hours, cell supernatants were collected and TNF-α was measured by ELISA. Means ± standard deviations of duplicate or triplicate conditions are displayed. TLR4-ab vs isotype groups were compared for each condition by t-test (* p<0.05; ** p<0.01; ***p<0.001). The concentration of cytokines in uninfected cells, for all conditions, was below the level of detection. One representative example of two independent experiments is shown.

https://doi.org/10.1371/journal.pntd.0008495.g006

Discussion

Melioidosis remains an important cause of sepsis and death in subtropical regions [2] and elucidating the immune response to Bp may facilitate the development of novel therapeutics. The main findings of our study are that lactoferrin concentrations are increased in 28-day non-survivors with melioidosis compared to survivors and that lactoferrin concentrations in blood from healthy subjects increase following ex vivo stimulation with heat-killed Bp. Furthermore, while lactoferrin does not impair Bt growth in culture or intracellular bacterial replication within monocytic cells, lactoferrin drives TNF-α production in Bt-infected monocytic cells in a TLR4-dependent manner.

Iron sequestration is likely one mechanism by which lactoferrin exerts antimicrobial properties, including in Bp infection [15,16]. However, few reports describe or discern between iron-saturation levels when assessing lactoferrin’s protective effects during bacterial infection. Furthermore, significant variance exists regarding the utilization of human or bovine and recombinant or purified lactoferrin. Here, we specifically analyzed the effect of human lactoferrin, utilizing both recombinant and human milk-purified versions. Iron-saturated lactoferrin–also known as holo-lactoferrin–may become quickly partially unsaturated and quantifying saturation levels can be challenging [37,38]. Our objective was to study lactoferrin-dependent immune modulation rather than antimicrobial properties. In order to limit possible confounding of our results by lactoferrin-dependent iron sequestration, we chose to use partially-iron saturated lactoferrin in our studies after establishing that partially-iron saturated lactoferrin does not impair Bt replication. Others have found similarly little effect of supplemental lactoferrin on the growth of Bp in culture [17]. However, the degree of iron saturation of rhLTF and phLTF may differ in culture and so this report does not directly address the role of iron saturation on the immunoregulatory properties of lactoferrin.

One of the pathogenic mechanisms of Bp and Bt is to invade intracellularly, replicate, and ultimately lyse the host cell or infect nearby cells [3941]. The spread of Bp to adjacent host cells results in the formation of multinuclear giant cells which eventually lyse resulting in localized spread [33,42]. Bp has variable sensitivity to two potentially bactericidal amino acid residues on bovine lactoferrin [4345]. Bovine lactoferrin has also been previously described as enhancing phagocytic killing in Staphylococcus aureus infection but recently was reported not to interfere with monocytic bacterial uptake [46,47]. In our studies, we found no difference in intracellular bacteria replication when Bt-infected, differentiated THP-1 cells were exposed to lactoferrin. These findings suggest that lactoferrin neither enhances, nor interferes with, the intracellular replication of Bt.

The initial inflammatory response to Bp is characterized by a robust activation of the innate immune system [4850]. This activation relies, in part, on multiple Toll-like receptor (TLR) pathways, including TLR4 and TLR5 [21,51,52]. Recent population genetic studies have identified TLR genetic variants associated with outcomes in melioidosis, further suggesting an important role of these pathways [51,53]. Besides iron-sequestration and other direct antimicrobial properties, lactoferrin may have immunomodulatory properties as well, though its role in TLR4 signaling is unclear. For example, human lactoferrin may promote TLR4-mediated activation through its carbohydrate chains [35]. Lactoferrin can also bind and sequester LPS or even inhibit LPS-binding, decreasing TLR4 intracellular signaling [5456]. Conversely, neonatal and adult human monocyte-derived macrophages, differentiated in the presence of lactoferrin, express reduced TLR4 activation and signaling, even after LPS stimulation [57]. Our findings support a TLR4-mediated mechanism of increased TNF-α production in the presence of lactoferrin.

Even in the absence of LPS, lactoferrin increases IL-8 and TNF-α mRNA expression in monocytes [58]. However, lactoferrin may alter LPS-induced intracellular cytokine production through NF-κB interference [18]. In vitro studies of methicillin-resistant Staphylococcus aureus (MRSA)-infected monocytes treated with lactoferrin demonstrated an increase in IFN-γ and IL-2 but a decrease in TNF-α, IL-1β and IL-6 [59]. TLR4 activates NF-κB, but so do multiple other pathways, including NF-κB-inducing kinase (NIK). Although lactoferrin does not have a known receptor on monocytes, it can stimulate NIK porcine monocyte-derived macrophages [58]. Lactoferrin can also activate NF-κB through TLR4-independent pathways as well, potentially accounting for the partial attenuation of lactoferrin’s TNF-α production by TLR4-blockade in our study [60].

Our study has several limitations. The association we observed between lower lactoferrin levels and survival in melioidosis patients was not adjusted for potential confounders. Our in vitro experiments may not represent the effects of lactoferrin in a systemic infection model. We also cannot rule out an effect related to the iron-saturation state of our lactoferrin preparations. Complete iron-depletion of lactoferrin is difficult, requiring acidic buffers which may affect protein function [37]. Finally, many other reports regarding the effects of lactoferrin on host response to bacterial infection have used bovine lactoferrin [10,46,58,61]. In our report, we limited our assessment to human-derived lactoferrin and so comparisons are limited. Finally, our infection model utilized Bt, a BSL-1 organism, as a surrogate for Bp infection. While monocyte infection with Bt may yield similar in vitro inflammatory cytokine patterns, further studies are needed to confirm a similar pattern in Bp infection [23]. Importantly, we studied the role of lactoferrin on monocyte infection; other cells, including neutrophils, may play a critical role in lactoferrin production during infection [62]. Finally, while we chose to focus on the TLR4 pathway due to its importance in melioidosis, other TLR pathways, including TLR2, may be modulated by lactoferrin [63,64].

In conclusion, our results implicate lactoferrin as a dynamic protein in melioidosis. We show that lactoferrin does not impair growth of Bt in culture or intracellular replication in monocytic cells but that it does enhance activation of specific proinflammatory pathways in monocytic cells. Furthermore, we have identified TLR4 as a contributor to this proinflammatory cytokine activation. These investigations add to our growing knowledge of host defense in melioidosis.

Supporting information

S1 Fig. Lactoferrin does not alter IFN-γ production by THP-1 cells infected with Bt.

THP-1 cells were infected with Bt at an MOI of 1 and 10, and 1 hour later were treated with either media, or 100 μg/ml of rhLTF or phLTF. At 6 hours after infection, cell supernatants were collected and IFN-γ was measured by ELISA. Means ± standard deviations of duplicate conditions are displayed. rhLTF vs media and phLTF vs media groups were compared for each MOI by t-test. The concentrations of cytokines in uninfected cells, for all conditions, were below the level of detection. One representative example of two or three independent experiments is shown.

https://doi.org/10.1371/journal.pntd.0008495.s001

(TIF)

S2 Fig. Lactoferrin does not alter TNF-α production by uninfected peripheral blood monocytes.

Peripheral blood monocytes were treated with either media, 100 μg/ml of rhLTF, or 100 μg/ml of phLTF. At 6 hours after treatment, cell supernatants were collected and TNF-α was measured by ELISA. Means ± standard deviations of triplicate conditions are displayed. rhLTF vs media and phLTF vs media groups were compared by t-test and both comparisons were above the limit of statistical significance (p>0.3 for both). One representative example of two independent experiments is shown.

https://doi.org/10.1371/journal.pntd.0008495.s002

(TIF)

Acknowledgments

The authors thank the patients and staff at Sunpasitthiprasong Hospital and Mahidol-Oxford Tropical Medicine Research Unit.

References

  1. 1. Wiersinga WJ, Currie BJ, Peacock SJ. Melioidosis. N Engl J Med. 2012;367: 1035–1044. pmid:22970946
  2. 2. Limmathurotsakul D, Wongratanacheewin S, Teerawattanasook N, Wongsuvan G, Chaisuksant S, Chetchotisakd P, et al. Increasing incidence of human melioidosis in northeast Thailand. Am J Trop Med Hyg. 2010;82: 1113–1117. pmid:20519609
  3. 3. Chewapreecha C, Holden MTG, Vehkala M, Välimäki N, Yang Z, Harris SR, et al. Global and regional dissemination and evolution of Burkholderia pseudomallei. Nat Microbiol. 2017;2: 1–8. pmid:28112723
  4. 4. Limmathurotsakul D, Golding N, Dance DAB, Messina JP, Pigott DM, Moyes CL, et al. Predicted global distribution of Burkholderia pseudomallei and burden of melioidosis. Nat Microbiol. 2016;1: 6–10. pmid:26877885
  5. 5. Valenti P, Antonini G. Lactoferrin: An important host defence against microbial and viral attack. Cell Mol Life Sci. 2005;62: 2576–2587. pmid:16261253
  6. 6. Sharma D, Murki A, Murki S, Pratap OT. Use of lactoferrin in the newborn: Where do we stand? J Matern Neonatal Med. 2015;28: 1774–1778. pmid:25245229
  7. 7. Sharma D, Shastri S, Sharma P. Role of lactoferrin in neonatal care: a systematic review. J Matern Neonatal Med. 2017;30: 1920–1932. pmid:27593940
  8. 8. Ling JML, Schryvers AB. Perspectives on interactions between lactoferrin and bacteria. Biochem Cell Biol. 2006;84: 275–281. pmid:16936797
  9. 9. Ellison RT, Giehl TJ. Killing of gram-negative bacteria by lactoferrin and lysozyme. J Clin Invest. 1991;88: 1080–1091. pmid:1918365
  10. 10. Wakabayashi H, Yamauchi K, Kobayashi T, Yaeshima T, Iwatsuki K, Yoshie H. Inhibitory effects of lactoferrin on growth and biofilm formation of Porphyromonas gingivalis and Prevotella intermedia. Antimicrob Agents Chemother. 2009;53: 3308–3316. pmid:19451301
  11. 11. Barber MF, Kronenberg Z, Yandell M, Elde NC. Antimicrobial Functions of Lactoferrin Promote Genetic Conflicts in Ancient Primates and Modern Humans. PLoS Genet. 2016;12: 1–15. pmid:27203426
  12. 12. Guntupalli K, Dean N, Morris PE, Bandi V, Margolis B, Rivers E, et al. A phase 2 randomized, double-blind, placebo-controlled study of the safety and efficacy of talactoferrin in patients with severe sepsis. Crit Care Med. 2013;41: 706–716. pmid:23425819
  13. 13. Manzoni P, Rinaldi M, Cattani S, Pugni L, Romeo MG, Messner H. Bovine Lactoferrin for Prevention of Late-Onset Sepsis in Very Low-Birth-Weight Neonates. J Am Med Assoc. 2013;302: 1421–1428.
  14. 14. Wang-ngarm S, Chareonsudjai S, Chareonsudjai P. Physicochemical factors affecting the growth of Burkholderia pseudomallei in soil microcosm. Am J Trop Med Hyg. 2014;90: 480–485. pmid:24445210
  15. 15. Schmidt IHE, Gildhorn C, Böning MAL, Kulow VA, Steinmetz I, Bast A. Burkholderia pseudomallei modulates host iron homeostasis to facilitate iron availability and intracellular survival. PLoS Negl Trop Dis. 2018;12: 1–25. pmid:29329289
  16. 16. Kvitko BH, Goodyear A, Propst KL, Dow SW, Schweizer HP. Burkholderia pseudomallei known siderophores and hemin uptake are dispensable for lethal murine melioidosis. PLoS Negl Trop Dis. 2012;6. pmid:22745846
  17. 17. Tandhavanant S, Thanwisai A, Limmathurotsakul D, Korbsrisate S, Day NP, Peacock SJ, et al. Effect of colony morphology variation of Burkholderia pseudomallei on intracellular survival and resistance to antimicrobial environments in human macrophages in vitro. BMC Microbiol. 2010;10. pmid:21114871
  18. 18. Håversen L, Ohlsson BG, Hahn-Zoric M, Hanson LÅ, Mattsby-Baltzer I. Lactoferrin down-regulates the LPS-induced cytokine production in monocytic cells via NF-κB. Cell Immunol. 2002;220: 83–95. pmid:12657243
  19. 19. Latorre D, Puddu P, Valenti P, Gessani S. Reciprocal interactions between lactoferrin and bacterial endotoxins and their role in the regulation of the immune response. Toxins (Basel). 2010;2: 54–68. pmid:22069546
  20. 20. Hwang SA, Kruzel ML, Actor JK. Recombinant human lactoferrin modulates human PBMC derived macrophage responses to BCG and LPS. Tuberculosis. 2016;101: S53–S62. pmid:27727130
  21. 21. West TE, Ernst RK, Jansson-Hutson MJ, Skerrett SJ. Activation of Toll-like receptors by Burkholderia pseudomallei. BMC Immunol. 2008;9: 1–10. pmid:18211710
  22. 22. Haraga A, West TE, Brittnacher MJ, Skerrett SJ, Miller SI. Burkholderia thailandensis as a model system for the study of the virulence-associated type III secretion system of burkholderia pseudomallei. Infect Immun. 2008;76: 5402–5411. pmid:18779342
  23. 23. West TE, Hawn TR, Skerrett SJ. Toll-like receptor signaling in airborne Burkholderia thailandensis infection. Infect Immun. 2009;77: 5612–5622. pmid:19797072
  24. 24. West TE, Frevert CW, Liggitt HD, Skerrett SJ. Inhalation of Burkholderia thailandensis results in lethal necrotizing pneumonia in mice: a surrogate model for pneumonic melioidosis. Trans R Soc Trop Med Hyg. 2008;102: S119–S126. pmid:19121672
  25. 25. Seymour CW, Liu VX, Iwashyna TJ, Brunkhorst FM, Rea TD, Scherag A, et al. Assessment of clinical criteria for sepsis for the third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA—J Am Med Assoc. 2016;315: 762–774. pmid:26903335
  26. 26. Hantrakun V, Somayaji R, Teparrukkul P, Boonsri C, Rudd K, Day NPJ, et al. Clinical epidemiology and outcomes of community acquired infection and sepsis among hospitalized patients in a resource limited setting in Northeast Thailand: A prospective observational study (Ubon-sepsis). PLoS One. 2018;13: 1–14. pmid:30256845
  27. 27. Teparrukkul P, Hantrakun V, Day NPJ, West TE, Limmathurotsakul D. Management and outcomes of severe dengue patients presenting with sepsis in a tropical country. PLoS One. 2017;12: 1–13. pmid:28437459
  28. 28. Rudd KE, Seymour CW, Aluisio AR, Augustin ME, Bagenda DS, Beane A, et al. Association of the quick sequential (sepsis-related) organ failure assessment (qSOFA) score with excess hospital mortality in adults with suspected infection in low- and middle-income countries. JAMA—J Am Med Assoc. 2018;319: 2202–2211. pmid:29800114
  29. 29. Rudd KE, Hantrakun V, Somayaji R, Booraphun S, Boonsri C, Fitzpatrick AL, et al. Early Management of Sepsis in Medical Patients in Rural Thailand: a Single-Center Prospective Observational Study. J Intensive Care. 2019;7: 1–8.
  30. 30. Chantratita N, Tandhavanant S, Myers ND, Seal S, Arayawichanont A, Kliangsa-ad A, et al. Survey of innate immune responses to Burkholderia pseudomallei in human blood identifies a central role for lipopolysaccharide. PLoS One. 2013;8: 1–11. pmid:24303060
  31. 31. Maniatis T, Fritsch EF, Sambrook J. Molecular Cloning. A Laboratory Manual. Cold Spring Harbor, NY; 1982.
  32. 32. Arnold RR, Russell JE, Champion WJ, Brewer M, Gauthier JJ. Bactericidal activity of human lactoferrin: Differentiation from the stasis of iron deprivation. Infect Immun. 1982;35: 792–799. pmid:6802759
  33. 33. Wiersinga WJ, Virk HS, Torres AG, Currie BJ, Peacock SJ, Dance DAB, et al. Melioidosis. Nat Rev Dis Prim. 2018;4: 1–22. pmid:29930242
  34. 34. Appelmelk BJ, An YQ, Geerts M, Thijs BG, De Boer HA, MacLaren DM, et al. Lactoferrin is a lipid A-binding protein. Infect Immun. 1994;62: 2628–2632. pmid:8188389
  35. 35. Ando K, Hasegawa K, Shindo KI, Furusawa T, Fujino T, Kikugawa K, et al. Human lactoferrin activates NF-κB through the Toll-like receptor 4 pathway while it interferes with the lipopolysaccharide-stimulated TLR4 signaling. FEBS J. 2010;277: 2051–2066. pmid:20345905
  36. 36. Yong JN, Sang BH, Jong SK, Yeo DY, Park SK, Hwan MK, et al. Lactoferrin works as a new LPS-binding protein in inflammatory activation of macrophages. Int Immunopharmacol. 2004;4: 1187–1199. pmid:15251114
  37. 37. Majka G, Śpiewak K, Kurpiewska K, Heczko P, Stochel G, Strus M, et al. A high-throughput method for the quantification of iron saturation in lactoferrin preparations. Anal Bioanal Chem. 2013;405: 5191–5200. pmid:23604471
  38. 38. Majka G, Więcek G, Śróttek M, Śpiewak K, Brindell M, Koziel J, et al. The impact of lactoferrin with different levels of metal saturation on the intestinal epithelial barrier function and mucosal inflammation. BioMetals. 2016;29: 1019–1033. pmid:27757565
  39. 39. Jones AL, Beveridge TJ, Woods DE. Intracellular survival of Burkholderia pseudomallei. Infect Immun. 1996;64: 782–790. pmid:8641782
  40. 40. Whiteley L, Meffert T, Haug M, Weidenmaier C, Hopf V, Bitschar K, et al. Entry, intracellular survival, and multinucleated-giant-cell-forming activity of Burkholderia pseudomallei in human primary phagocytic and nonphagocytic cells. Infect Immun. 2017;85: 1–12. pmid:28760929
  41. 41. Jenner D, Ducker C, Clark G, Prior J, Rowland CA. Using multispectral imaging flow cytometry to assess an in vitro intracellular Burkholderia thailandensis infection model. Cytometry A. 2016;89: 328–337. pmid:26841315
  42. 42. French CT, Toesca IJ, Wu T-H, Teslaa T, Beaty SM, Wong W, et al. Dissection of the Burkholderia intracellular life cycle using a photothermal nanoblade. Proc Natl Acad Sci. 2011;108: 12095–12100. pmid:21730143
  43. 43. Kanthawong S, Nazmi K, Wongratanacheewin S, Bolscher JGM, Wuthiekanun V, Taweechaisupapong S. In vitro susceptibility of Burkholderia pseudomallei to antimicrobial peptides. Int J Antimicrob Agents. 2009;34: 309–314. pmid:19577435
  44. 44. Kanthawong S, Puknun A, Bolscher JGM, Nazmi K, Van Marle J, De Soet JJ, et al. Membrane-active mechanism of LFchimera against Burkholderia pseudomallei and Burkholderia thailandensis. BioMetals. 2014;27: 949–956. pmid:24961697
  45. 45. Puknun A, Kanthawong S, Anutrakunchai C, Nazmi K, Tigchelaar W, Hoeben KA, et al. Ultrastructural effects and antibiofilm activity of LFchimera against Burkholderia pseudomallei. World J Microbiol Biotechnol. 2016;32: 1–10. pmid:26596268
  46. 46. Miyauchi H, Hashimoto SI, Nakajima M, Shinoda I, Fukuwatari Y, Hayasawa H. Bovine lactoferrin stimulates the phagocytic activity of human neutrophils: Identification of its active domain. Cell Immunol. 1998;187: 34–37. pmid:9682001
  47. 47. Kai K, Komine KI, Komine Y, Kuroishi T, Kozutsumi T, Kobayashi J, et al. Lactoferrin stimulates a Staphylococcus aureus killing activity of bovine phagocytes in the mammary gland. Microbiol Immunol. 2002;46: 187–194. pmid:12008928
  48. 48. Lauw FN, Simpson AJH, Prins JM, Smith MD, Kurimoto M, van Deventer SJH, et al. Elevated Plasma Concentrations of Interferon (IFN)–γ and the IFN‐γ–Inducing Cytokines Interleukin (IL)–18, IL‐12, and IL‐15 in Severe Melioidosis. J Infect Dis. 1999;180: 1878–1885. pmid:10558944
  49. 49. Friedland JS, Suputtamongkol Y, Remick DG, Chaowagul W, Strieter RM, Kunkel SL, et al. Prolonged elevation of interleukin-8 and interleukin-6 concentrations in plasma and of leukocyte interleukin-8 mRNA levels during septicemic and localized Pseudomonas pseudomallei infection. Infect Immun. 1992;60: 2402–2408. pmid:1375198
  50. 50. Krishnananthasivam Shivankari; Sathkumara Harindra; Corea Enoka; Natesan Mohan; De Silva A. Gene Expression Profile of Human Cytokines in Response to Burkholderia pseudomallei Infection. mSphere. 2017;2: e00121–17. https://doi.org/10.1128/mSphere.00121-17. pmid:28435890
  51. 51. West TE, Chantratita N, Chierakul W, Limmathurotsakul D, Wuthiekanun V, Myers ND, et al. Impaired TLR5 Functionality Is Associated with Survival in Melioidosis. J Immunol. 2013;190: 3373–3379. pmid:23447684
  52. 52. Sengyee S, Yoon SH, Paksanont S, Yimthin T, Wuthiekanun V, Limmathurotsakul D, et al. Comprehensive analysis of clinical Burkholderia pseudomallei isolates demonstrates conservation of unique lipid A structure and TLR4-dependent innate immune activation. 2018; 1–24. pmid:29474381
  53. 53. West TE, Chierakul W, Chantratita N, Limmathurotsakul D, Wuthiekanun V, Emond MJ, et al. Toll-like receptor 4 region genetic variants are associated with susceptibility to melioidosis. Genes Immun. 2011;13: 38–46. pmid:21776015
  54. 54. Puddu P, Latorre D, Valenti P, Gessani S. Immunoregulatory role of lactoferrin-lipopolysaccharide interactions. BioMetals. 2010;23: 387–397. pmid:20191308
  55. 55. Baveye S, Elass E, Mazurier J, Legrand D. Lactoferrin inhibits the binding of lipopolysaccharides to L-selectin and subsequent production of reactive oxygen species by neutrophils. FEBS Lett. 2000;469: 5–8. pmid:10708745
  56. 56. Kruzel ML, Harari Y, Mailman D, Actor JK, Zimecki M. Differential effects of prophylactic, concurrent and therapeutic lactoferrin treatment on LPS-induced inflammatory responses in mice. Clin Exp Immunol. 2002;130: 25–31. pmid:12296849
  57. 57. Wisgrill L, Wessely I, Spittler A, Förster-Waldl E, Berger A, Sadeghi K. Human lactoferrin attenuates the proinflammatory response of neonatal monocyte-derived macrophages. Clin Exp Immunol. 2018;192: 315–324. pmid:29393509
  58. 58. Zemankova N, Chlebova K, Matiasovic J, Prodelalova J, Gebauer J, Faldyna M. Bovine lactoferrin free of lipopolysaccharide can induce a proinflammatory response of macrophages. BMC Vet Res. 2016;12: 1–9. pmid:26728767
  59. 59. Hwang Shen-An; Kruzel Marian; Actor J. Immunomodulatory effects of recombinant lactoferrin during MRSA infection. Int Immunopharmacol. 2014;20: 157–163. pmid:24613206
  60. 60. Curran CS, Demick KP, Mansfield JM. Lactoferrin activates macrophages via TLR4-dependent and -independent signaling pathways. Cell Immunol. 2006;242: 23–30. pmid:17034774
  61. 61. Bhimani RS, Vendrov Y, Furmanski P. Influence of lactoferrin feeding and injection against systemic staphylococcal infections in mice. J Appl Microbiol. 1999;86: 135–144. pmid:10030017
  62. 62. Lepanto MS, Rosa L, Paesano R, Valenti P, Cutone A. Lactoferrin in aseptic and septic inflammation. Molecules. 2019;24: 1–27. pmid:30987256
  63. 63. He Y, Lawlor NT, Newburg DS. Human Milk Components Modulate Toll-Like Receptor–Mediated Inflammation. Adv Nutr. 2016;7: 102–11. pmid:26773018
  64. 64. Zheng Y, Qin Z, Ye Q, Chen P, Wang Z, Yan Q, et al. Lactoferrin suppresses the Epstein-Barr virus-induced inflammatory response by interfering with pattern recognition of TLR2 and TLR9. Lab Investig. 2014;94: 1188–1199. pmid:25068657