24,537 research outputs found

    Abstracts of the 7th International Conference on Lactoferrin / Résumés de la 7e Conférence internationale sur la lactoferrine

    Get PDF
    The article presents abstracts on lactoferrin research. They include "Oral administration of lactoferrin raises NK cell activity in mice,"Comparison of antimicrobial activity between lactoferricin B 15 derivative and antibiotics" and "Batch extraction of lactoferrin from raw whole milk.

    Characterization of recombinant human lactoferrin N-glycans expressed in the milk of transgenic cows.

    Get PDF
    Lactoferrin (LF) is one of the most abundant bioactive glycoproteins in human milk. Glycans attached through N-glycosidic bonds may contribute to Lactoferrin functional activities. In contrast, LF is present in trace amounts in bovine milk. Efforts to increase LF concentration in bovine milk led to alternative approaches using transgenic cows to express human lactoferrin (hLF). This study investigated and compared N-glycans in recombinant human lactoferrin (rhLF), bovine lactoferrin (bLF) and human lactoferrin by Nano-LC-Chip-Q-TOF Mass Spectrometry. The results revealed a high diversity of N-glycan structures, including fucosylated and sialylated complex glycans that may contribute additional bioactivities. rhLF, bLF and hLF had 23, 27 and 18 N-glycans respectively with 8 N-glycan in common overall. rhLF shared 16 N-glycan with bLF and 9 N-glycan with hLF while bLF shared 10 N-glycan with hLF. Based on the relative abundances of N-glycan types, rhLF and hLF appeared to contain mostly neutral complex/hybrid N-glycans (81% and 52% of the total respectively) whereas bLF was characterized by high mannose glycans (65%). Interestingly, the majority of hLF N-glycans were fucosylated (88%), whereas bLF and rhLF had only 9% and 20% fucosylation, respectively. Overall, this study suggests that rhLF N-glycans share more similarities to bLF than hLF

    Enteral lactoferrin supplementation for very preterm infants: a randomised placebo-controlled trial

    Get PDF
    Background Infections acquired in hospital are an important cause of morbidity and mortality in very preterm infants. Several small trials have suggested that supplementing the enteral diet of very preterm infants with lactoferrin, an antimicrobial protein processed from cow's milk, prevents infections and associated complications. The aim of this large randomised controlled trial was to collect data to enhance the validity and applicability of the evidence from previous trials to inform practice. Methods In this randomised placebo-controlled trial, we recruited very preterm infants born before 32 weeks' gestation in 37 UK hospitals and younger than 72 h at randomisation. Exclusion criteria were presence of a severe congenital anomaly, anticipated enteral fasting for longer than 14 days, or no realistic prospect of survival. Eligible infants were randomly assigned (1:1) to receive either enteral bovine lactoferrin (150 mg/kg per day; maximum 300 mg/day; lactoferrin group) or sucrose (same dose; control group) once daily until 34 weeks' postmenstrual age. Web-based randomisation minimised for recruitment site, gestation (completed weeks), sex, and single versus multifetal pregnancy. Parents, caregivers, and outcome assessors were unaware of group assignment. The primary outcome was microbiologically confirmed or clinically suspected late-onset infection (occurring >72 h after birth), which was assessed in all participants for whom primary outcome data was available by calculating the relative risk ratio with 95% CI between the two groups. The trial is registered with the International Standard Randomised Controlled Trial Number 88261002. Findings We recruited 2203 participants between May 7, 2014, and Sept 28, 2017, of whom 1099 were assigned to the lactoferrin group and 1104 to the control group. Four infants had consent withdrawn or unconfirmed, leaving 1098 infants in the lactoferrin group and 1101 in the sucrose group. Primary outcome data for 2182 infants (1093 [99·5%] of 1098 in the lactoferrin group and 1089 [99·0] of 1101 in the control group) were available for inclusion in the modified intention-to-treat analyses. 316 (29%) of 1093 infants in the intervention group acquired a late-onset infection versus 334 (31%) of 1089 in the control group. The risk ratio adjusted for minimisation factors was 0·95 (95% CI 0·86–1·04; p=0·233). During the trial there were 16 serious adverse events for infants in the lactoferrin group and 10 for infants in the control group. Two events in the lactoferrin group (one case of blood in stool and one death after intestinal perforation) were assessed as being possibly related to the trial intervention. Interpretation Enteral supplementation with bovine lactoferrin does not reduce the risk of late-onset infection in very preterm infants. These data do not support its routine use to prevent late-onset infection and associated morbidity or mortality in very preterm infants. Funding UK National Institute for Health Research Health Technology Assessment programme (10/57/49)

    Lactoferrin's anti-cancer properties. Safety, selectivity, and wide range of action

    Get PDF
    Despite recent advances in cancer therapy, current treatments, including radiotherapy, chemotherapy, and immunotherapy, although beneficial, present attendant side effects and long-term sequelae, usually more or less affecting quality of life of the patients. Indeed, except for most of the immunotherapeutic agents, the complete lack of selectivity between normal and cancer cells for radio- and chemotherapy can make them potential antagonists of the host anti-cancer self-defense over time. Recently, the use of nutraceuticals as natural compounds corroborating anti-cancer standard therapy is emerging as a promising tool for their relative abundance, bioavailability, safety, low-cost effectiveness, and immuno-compatibility with the host. In this review, we outlined the anti-cancer properties of Lactoferrin (Lf), an iron-binding glycoprotein of the innate immune defense. Lf shows high bioavailability after oral administration, high selectivity toward cancer cells, and a wide range of molecular targets controlling tumor proliferation, survival, migration, invasion, and metastasization. Of note, Lf is able to promote or inhibit cell proliferation and migration depending on whether it acts upon normal or cancerous cells, respectively. Importantly, Lf administration is highly tolerated and does not present significant adverse effects. Moreover, Lf can prevent development or inhibit cancer growth by boosting adaptive immune response. Finally, Lf was recently found to be an ideal carrier for chemotherapeutics, even for the treatment of brain tumors due to its ability to cross the blood-brain barrier, thus globally appearing as a promising tool for cancer prevention and treatment, especially in combination therapies

    Crystallographic and physicochemical studies on anion-binding and deglycosylation of human lactoferrin : a thesis presented in partial fulfilment of the requirements for the degree of Master of Philosophy in Chemistry at Massey University

    Get PDF
    Lactoferrin, a monomeric 80 kDa glycoprotein, is a major component of human milk and is found in many other exocrine secretions as well as in the neutrophilic granules of white blood cells. A member of the transferrin family of iron-binding proteins, lactoferrin has the ability to bind tightly but reversibly 2Fe3+ ions with the concomitant binding of 2CO32- ions. Crystal structure studies clearly demonstrate that the polypeptide chain is folded into two similar lobes, representing the N- and the C-terminal halves of the protein, and that each lobe contains one of the two very similar iron-binding sites. Transferrins also show considerable versatility in their binding properties, being able to bind many metal ions in place of Fe3+ and anions in place of CO32-. Differences between the two sites become more pronounced, however, with the substitution of non-native metals and anions, and the origins of this inequivalence have long been debated. To investigate the means by which lactoferrin can accommodate anions larger than carbonate, the diferricdioxalatolactoferrin (Fe2(C2O4)2Lf) complex was prepared and crystallised. The crystals, which were isomorphous with those of Fe2(CO3)2Lf, were used to collect a complete 2.4 Ǻ data set at the Photon Factory (Japan) synchrotron source. The structure was refined by restrained least squares methods to a final R factor of 0.196 for all 31758 reflections in the resolution range 8.0 to 2.4 Ǻ. The polypeptide folding and domain closure were identical to those of the native Fe2(CO3)2Lf. In contrast to the carbonate complex, however, in which the two binding sites appear almost identical, with the carbonate coordinating in a symmetrical bidentate mode to each iron, when oxalate is the anion, the coordination around the metal differs between the N- and the C-lobe. In the C-lobe, the oxalate has a symmetrical 1,2-bidentate coordination to the iron, but in the N-lobe this coordination is quite asymmetric (O1ox-Fe = 1.87 Ǻ, O2ox-Fe = 2.55 Ǻ). Analysis of the structure indicates that the stereochemistry of the oxalate coordination to the iron is influenced by the position of the anion-binding arginine. The position this arginine can adopt in each lobe is, in turn, influenced by residues more remote from the iron site and which differ between the N- and C-lobes. All lactoferrins so far characterised are glycoproteins, but the importance of the glycan chains for structure and/or function has yet to be established. Enzymatic methods were used to deglycosylate human and bovine lactoferrins, and the native deglycosylated forms of the human protein were compared with respect to CD spectra, iron binding and release, stability to proteolysis and heat stability. Deglycosylation was carried out at pH 6.0 on the iron-free form of lactoferrin, using an endoglycosidase preparation from Flavobacterium meningosepticum, comprising PNGase F and Endo F. Deglycosylation was rapid for human lactoferrin, being essentially complete within 12-24 hr. Only partial deglycosylation of bovine lactoferrin could be achieved under the same conditions, however, and this is attributed to the relative inaccessibility of at least one of the glycosylation sites. The CD spectra of native and deglycosylated human lactoferrins were found to be essentially identical in the range 250-350 nm, implying the same three dimensional structures. Both also bind iron in identical fashion; 2 Fe3+ ions are bound and binding is complete within 1 minute. The release of iron as the pH was lowered from 8.0 to 2.0 also showed no significant difference, the pH at which 50% release had taken place being 3.2 and 3.0 respectively for native and deglycosylated proteins. Susceptibility to proteolytic digestion by bovine trypsin over a period of 24 hr showed similar fragmentation patterns and a similar time course for the reaction for both species. Iron binding ability as a function of temperature was used as a measure of heat stability; melting temperatures derived from these experiments were 64°C for native and 63°C for deglycosylated lactoferrin. Comparison of the three dimensional structures of glycosylated iron-lactoferrin with deglycosylated apo-lactoferrin are consistent with these results, showing only a small increase in flexibility near the glycosylation site, when the carbohydrate is removed. Conclusions are that the in vitro physicochemical properties of lactoferrin are unaffected by the presence or absence of its glycan chains. In vivo studies may be necessary to establish the importance, if any, of glycosylation

    Lactoferrin. A natural glycoprotein involved in iron and inflammatory homeostasis

    Get PDF
    Human lactoferrin (hLf), an iron-binding multifunctional cationic glycoprotein secreted by exocrine glands and by neutrophils, is a key element of host defenses. HLf and bovine Lf (bLf), possessing high sequence homology and identical functions, inhibit bacterial growth and biofilm dependently from iron binding ability while, independently, bacterial adhesion to and the entry into cells. In infected/inflamed host cells, bLf exerts an anti-inflammatory activity against interleukin-6 (IL-6), thus up-regulating ferroportin (Fpn) and transferrin receptor 1 (TfR1) and down-regulating ferritin (Ftn), pivotal actors of iron and inflammatory homeostasis (IIH). Consequently, bLf inhibits intracellular iron overload, an unsafe condition enhancing in vivo susceptibility to infections, as well as anemia of inflammation (AI), re-establishing IIH. In pregnant women, affected by AI, bLf oral administration decreases IL-6 and increases hematological parameters. This surprising effect is unrelated to iron supplementation by bLf (80 µg instead of 1-2 mg/day), but to its role on IIH. AI is unrelated to the lack of iron, but to iron delocalization: cellular/tissue overload and blood deficiency. BLf cures AI by restoring iron from cells to blood through Fpn up-expression. Indeed, anti-inflammatory activity of oral and intravaginal bLf prevents preterm delivery. Promising bLf treatments can prevent/cure transitory inflammation/anemia/oral pathologies in athletes

    Lactoferrin efficiently counteracts the inflammation-induced changes of the iron homeostasis system in macrophages

    Get PDF
    Human lactoferrin (hLf), an 80-kDa multifunctional iron-binding cationic glycoprotein, is constitutively secreted by exocrine glands and by neutrophils during inflammation. hLf is recognized as a key element in the host immune defense system. The in vitro and in vivo experiments are carried out with bovine Lf (bLf), which shares high sequence homology and identical functions with hLf, including anti-inflammatory activity. Here, in "pure" M1 human macrophages, obtained by stimulation with a mixture of 10 pg/ml LPS and 20 ng/ml IFN-γ, as well as in a more heterogeneous macrophage population, challenged with high-dose of LPS (1 µg/ml), the effect of bLf on the expression of the main proteins involved in iron and inflammatory homeostasis, namely ferroportin (Fpn), membrane-bound ceruloplasmin (Cp), cytosolic ferritin (Ftn), transferrin receptor 1, and cytokines has been investigated. The increase of IL-6 and IL-1β cytokines, following the inflammatory treatments, is associated with both upregulation of cytosolic Ftn and downregulation of Fpn, membrane-bound Cp, and transferrin receptor 1. All these changes take part into intracellular iron overload, a very unsafe condition leading in vivo to higher host susceptibility to infections as well as iron deficiency in the blood and anemia of inflammation. It is, therefore, of utmost importance to counteract the persistence of the inflammatory status to rebalance iron levels between tissues/secretions and blood. Moreover, levels of the antiinflammatory cytokine IL-10 were increased in cells treated with high doses of LPS. Conversely, IL-10 decreased when the LPS/IFN-γ mix was used, suggesting that only the inflammation triggered by LPS high doses can switch on an anti-inflammatory response in our macrophagic model. Here, we demonstrate that bLf, when included in the culture medium, significantly reduced IL-6 and IL-1β production and efficiently prevented the changes of Fpn, membrane-bound Cp, cytosolic Ftn, and transferrin receptor 1 in "pure" M1 macrophages, as well as in the more heterogeneous macrophage population. In addition, the decrease of IL-10 induced by the LPS/IFN-γ mix was counteracted by bovine lactoferrin. Several drugs capable of modulating macrophagic phenotypes are emerging as attractive molecules for treating inflammation, and in this sense, bovine lactoferrin is no exception
    corecore