Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.997
Filter
1.
Lett Appl Microbiol ; 77(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39009015

ABSTRACT

Lactoferrin is an antimicrobial glycoprotein that demonstrates a broad-spectrum of activity against a wide variety of clinical pathogens. This study investigated the potential of bovine lactoferrin (bLf) against multidrug resistant Staphylococcus capitis (S. capitis) strains. Growth curve analysis and time-kill curves demonstrated that at 750 µg ml-1 lactoferrin significantly inhibited (50.6%, P < 0.05) the growth of most isolates tested (90%), and this effect was based on a bacteriostatic mechanism. At the same concentration, bLf also significantly inhibited (30%, P < 0.05) biofilm formation in 40% of strains tested. Combinations of bLf with selected antibiotics were assessed for enhanced antimicrobial activity using growth curves. BLf combined with ß-lactam antibiotics reduced the growth of S. capitis strains, however, the effects were not significant. BLf displays antimicrobial effects against multidrug resistant S. capitis isolates, but with strain-specific effects.


Subject(s)
Anti-Bacterial Agents , Biofilms , Lactoferrin , Microbial Sensitivity Tests , Staphylococcus capitis , Lactoferrin/pharmacology , Animals , Cattle , Biofilms/drug effects , Biofilms/growth & development , Anti-Bacterial Agents/pharmacology , Staphylococcus capitis/drug effects , Drug Resistance, Multiple, Bacterial , Staphylococcal Infections/microbiology
3.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39000442

ABSTRACT

Human defensins are cysteine-rich peptides (Cys-rich peptides) of the innate immune system. Defensins contain an ancestral structural motif (i.e., γ-core motif) associated with the antimicrobial activity of natural Cys-rich peptides. In this study, low concentrations of human α- and ß-defensins showed microbicidal activity that was not associated with cell membrane permeabilization. The cell death pathway was similar to that previously described for human lactoferrin, also an immunoprotein containing a γ-core motif. The common features were (1) cell death not related to plasma membrane (PM) disruption, (2) the inhibition of microbicidal activity via extracellular potassium, (3) the influence of cellular respiration on microbicidal activity, and (4) the influence of intracellular pH on bactericidal activity. In addition, in yeast, we also observed (1) partial K+-efflux mediated via Tok1p K+-channels, (2) the essential role of mitochondrial ATP synthase in cell death, (3) the increment of intracellular ATP, (4) plasma membrane depolarization, and (5) the inhibition of external acidification mediated via PM Pma1p H+-ATPase. Similar features were also observed with BM2, an antifungal peptide that inhibits Pma1p H+-ATPase, showing that the above coincident characteristics were a consequence of PM H+-ATPase inhibition. These findings suggest, for the first time, that human defensins inhibit PM H+-ATPases at physiological concentrations, and that the subsequent cytosolic acidification is responsible for the in vitro microbicidal activity. This mechanism of action is shared with human lactoferrin and probably other antimicrobial peptides containing γ-core motifs.


Subject(s)
Cell Membrane , Proton-Translocating ATPases , Humans , Cell Membrane/metabolism , Cell Membrane/drug effects , Proton-Translocating ATPases/metabolism , Proton-Translocating ATPases/antagonists & inhibitors , Cell Membrane Permeability/drug effects , Anti-Infective Agents/pharmacology , Defensins/pharmacology , Defensins/metabolism , Hydrogen-Ion Concentration , Saccharomyces cerevisiae/metabolism , beta-Defensins/metabolism , beta-Defensins/pharmacology , Lactoferrin/pharmacology , Lactoferrin/metabolism , Potassium/metabolism , Microbial Sensitivity Tests , Candida albicans/drug effects
4.
Curr Gene Ther ; 24(5): 410-421, 2024.
Article in English | MEDLINE | ID: mdl-39005063

ABSTRACT

BACKGROUND: Injury systemically disrupts the homeostatic balance and can cause organ failure. LF mediates both iron-dependent and iron-independent mechanisms, and the role of LF in regulating iron homeostasis is vital in terms of metabolism. OBJECTIVES: In this study, we evaluated the organ-level effect and gene expression change of bLf in the cutaneous repair process. MATERIALS AND METHODS: An excisional full-thickness skin defect (FTSD) wound model was created in male Sprague Dawley rats (180-250 g) (n = 48) fed a high-fat diet (HFD) and the PHGPx, SLC7A11 and SLC40A1 genes and iron metabolism were evaluated. The animals were randomly divided into 6 groups: 1- Control, 2- bLf (200 mg/kg/day, oral), 3- FTSD (12 mm in diameter, dorsal), 4- HFD + bLf, 5- HFD + FTSD, 6- HFD + FTSD + bLf. Histologically, iron accumulation was demonstrated by Prussian blue staining in the liver, kidney, and intestinal tissues. Gene expression analysis was performed with qPCR. RESULTS: Histologically, iron accumulation was demonstrated by Prussian blue staining in the liver, kidney, and intestinal tissues. Prussian blue reactions were detected in the kidney. PHPGx and SLC7A11 genes in kidney and liver tissue were statistically significant (P < 0.05) except for the SLC40A1 gene (P > 0.05). Expression changes of the three genes were not statistically significant in analyses of rat intestinal tissue (P = 0.057). CONCLUSION: In the organ-level ferroptotic damage mechanism triggered by wound formation. BLf controls the expression of three genes and manages iron deposition in these three tissues. In addition, it suppressed the increase in iron that would drive the cell to ferroptosis and anemia caused by inflammation, thereby eliminating iron deposition in the tissues.


Subject(s)
Homeostasis , Iron , Lactoferrin , Rats, Sprague-Dawley , Wound Healing , Animals , Iron/metabolism , Rats , Male , Homeostasis/drug effects , Lactoferrin/pharmacology , Lactoferrin/genetics , Wound Healing/drug effects , Wound Healing/genetics , Cattle , Multiple Organ Failure/genetics , Multiple Organ Failure/metabolism , Multiple Organ Failure/drug therapy , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Gene Expression Regulation/drug effects , Liver/metabolism , Liver/drug effects
5.
Molecules ; 29(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38998965

ABSTRACT

In this study, a self-responsive fluorescence aptasensor was established for the determination of lactoferrin (Lf) in dairy products. Herein, the aptamer itself functions as both a recognition element that specifically binds to Lf and a fluorescent signal reporter in conjunction with fluorescent moiety. In the presence of Lf, the aptamer preferentially binds to Lf due to its specific and high-affinity recognition by folding into a self-assembled and three-dimensional spatial structure. Meanwhile, its reduced spatial distance in the aptamer-Lf complex induces a FRET phenomenon based on the quenching of 6-FAM by amino acids in the Lf protein, resulting in a turn-off of the fluorescence of the system. As a result, the Lf concentration can be determined straightforwardly corresponding to the change in the self-responsive fluorescence signal. Under the optimized conditions, good linearities (R2 > 0.99) were achieved in an Lf concentration range of 2~10 µg/mL for both standard solutions and the spiked matrix, as well as with the desirable detection limits of 0.68 µg/mL and 0.46 µg/mL, respectively. Moreover, the fluorescence aptasensor exhibited reliable recoveries (89.5-104.3%) in terms of detecting Lf in three commercial samples, which is comparable to the accuracy of the HPCE method. The fluorescence aptasensor offers a user-friendly, cost-efficient, and promising sensor platform for point-of-need detection.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , Dairy Products , Lactoferrin , Lactoferrin/analysis , Lactoferrin/chemistry , Aptamers, Nucleotide/chemistry , Biosensing Techniques/methods , Dairy Products/analysis , Fluorescence , Limit of Detection , Spectrometry, Fluorescence/methods , Food Analysis/methods , Fluorescence Resonance Energy Transfer/methods
6.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39000056

ABSTRACT

The lack of specific biological materials and biomarkers limits our knowledge of the mechanisms underlying intrauterine regulation of iron supply to the fetus. Determining the meconium content of proteins commonly used in the laboratory to assess the transport, storage, and distribution of iron in the body may elucidate their roles in fetal development. Ferritin, transferrin, haptoglobin, ceruloplasmin, lactoferrin, myeloperoxidase (MPO), neutrophil gelatinase-associated lipocalin (NGAL), and calprotectin were determined by ELISA in meconium samples obtained from 122 neonates. There were strong correlations between the meconium concentrations of haptoglobin, transferrin, and NGAL (p < 0.05). Meconium concentrations of ferritin were several-fold higher than the concentrations of the other proteins, with the exception of calprotectin whose concentration was approximately three-fold higher than that of ferritin. Meconium ceruloplasmin concentration significantly correlated with the concentrations of MPO, NGAL, lactoferrin, and calprotectin. Correlations between the meconium concentrations of haptoglobin, transferrin, and NGAL may reflect their collaborative involvement in the storage and transport of iron in the intrauterine environment in line with their recognized biological properties. High meconium concentrations of ferritin may provide information about the demand for iron and its utilization by the fetus. The associations between ceruloplasmin and neutrophil proteins may indicate the involvement of ceruloplasmin in the regulation of neutrophil activity in the intrauterine environment.


Subject(s)
Ceruloplasmin , Haptoglobins , Iron , Lipocalin-2 , Meconium , Humans , Iron/metabolism , Meconium/metabolism , Infant, Newborn , Ceruloplasmin/metabolism , Female , Haptoglobins/metabolism , Lipocalin-2/metabolism , Transferrin/metabolism , Transferrin/analysis , Ferritins/metabolism , Leukocyte L1 Antigen Complex/metabolism , Lactoferrin/metabolism , Lactoferrin/analysis , Male , Peroxidase/metabolism , Biomarkers/metabolism , Adult
7.
Nutrients ; 16(14)2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39064803

ABSTRACT

Lactoferrin (LF) is a major component of human milk. LF supplementation (currently bovine) supports the immune system and helps maintain iron homeostasis in adults. No recombinant human lactoferrin (rhLF) is available for commercial food use. To determine the extent to which rhLF (Effera™) produced by Komagataella phaffii digests similarly to hmLF, a validated in vitro digestion protocol was carried out. Bovine LF (bLF) was used as an additional control, as it is approved for use in various food categories. This study compared the extent of intact protein retention and the profile of peptides released in hmLF, bLF and rhLF (each with low and high iron saturation) across simulated adult gastric and intestinal digestion using gel electrophoresis, ELISA and LC-MS. Intact LF retention across digestion was similar across LF types, but the highest iron-saturated hmLF had greater retention in the simulated gastric fluid than all other sample types. Peptides identified in digested hmLF samples strongly correlated with digested rhLF samples (0.86 < r < 0.92 in the gastric phase and 0.63 < r < 0.70 in the intestinal phase), whereas digested bLF samples were significantly different. These findings support the potential for rhLF as a food ingredient for human consumption.


Subject(s)
Digestion , Lactoferrin , Milk, Human , Recombinant Proteins , Lactoferrin/metabolism , Humans , Cattle , Animals , Milk, Human/chemistry , Peptides , Iron/metabolism
8.
Nutrients ; 16(14)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39064821

ABSTRACT

BACKGROUND: Postmenopausal dyspareunia and vulvar pain are common complaints, affecting about 60% of women within a few years after hormone levels begin to decline (such as estrogen and androgen). Atrophic changes mainly located in the vulvar vestibule and vulnerability to vulvovaginal infections in postmenopause could be predisposing factors to the development of vulvar burning/pain and introital dyspareunia (vestibulodynia secondary to atrophy). Tibolone is the most effective and safe alternative for treating menopausal symptoms. The role of Lactobacilli and lactoferrin shows its effectiveness in the treatment of vaginal microbiota dysbiosis. The aim of the present study was to assess the efficacy of the combination of tibolone and an oral-specific Lactobacilli mixture in combination with bovine lactoferrin as synergistic therapy for the treatment of vestibulodynia related to atrophy. METHODS: In this study, we included 35 postmenopausal women with at least 1 year of amenorrhea, affected by vulvar burning/pain and introital dyspareunia. All participants received treatment with open-label, oral Tibolone 2.5 mg and Lactobacilli mixture (5 × 109 CFU per capsule) in combination with bovine lactoferrin (Respecta®). Each product was taken once daily for 90 days. RESULTS: After 90 d of therapy with TIB+ Respecta®, in 30 women that completed the treatment, there was a statistically significant decrease from the baseline in the mean of the Visual Analog Scale for vulvar burning/pain and a reduction in scores in the pain evaluation test. CONCLUSIONS: This study provides evidence that the combination of TIB+ Respecta® was effective in reducing symptoms related to vestibular pain and hypersensitivity in a postmenopausal setting.


Subject(s)
Lactobacillus , Lactoferrin , Norpregnenes , Postmenopause , Female , Humans , Lactoferrin/administration & dosage , Middle Aged , Norpregnenes/administration & dosage , Vulvodynia/drug therapy , Vulvodynia/therapy , Probiotics/administration & dosage , Treatment Outcome , Dyspareunia/drug therapy , Dyspareunia/therapy , Vulva/microbiology
9.
Chem Biol Drug Des ; 104(1): e14574, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38958121

ABSTRACT

To develop novel bovine lactoferrin (bLF) peptides targeting bLF-tumour necrosis factor (TNF) receptor-associated factor 6 (TRAF6) binding sites, we identified two peptides that could target bLF-TRAF6 binding sites using structural analysis. Moreover, another peptide that could bind to the TRAF6 dimerization area was selected from the bLF sequence. The effects of each peptide on cytokine expression in lipopolysaccharide (LPS)-stimulated osteoblasts (ST2) and on osteoclastogenesis were examined using an LPS-treated co-culture of primary bone marrow cells (BMCs) with ST2 cells and a single culture of osteoclast precursor cells (RAW-D) treated with soluble receptor activator of NF-κB ligand. Finally, the effectiveness of these peptides against LPS-induced alveolar bone destruction was assessed. Two of the three peptides significantly suppressed LPS-induced TNF-α and interleukin-1ß expression in ST2 cells. Additionally, these peptides inhibited and reversed LPS-induced receptor activator of NF-κB ligand (RANKL) upregulation and osteoprotegerin (OPG) downregulation, respectively. Furthermore, both peptides significantly reduced LPS-induced osteoclastogenesis in the BMC-ST2 co-culture and RANKL-induced osteoclastogenesis in RAW-D cells. In vivo, topical application of these peptides significantly reduced the osteoclast number by downregulating RANKL and upregulating OPG in the periodontal ligament. It is indicated that the novel bLF peptides can be used to treat periodontitis-associated bone destruction.


Subject(s)
Lactoferrin , Lipopolysaccharides , Osteoclasts , Peptides , Animals , Lactoferrin/pharmacology , Lactoferrin/chemistry , Lactoferrin/metabolism , Lipopolysaccharides/pharmacology , Rats , Peptides/pharmacology , Peptides/chemistry , Osteoclasts/drug effects , Osteoclasts/metabolism , RANK Ligand/metabolism , Male , Alveolar Bone Loss/drug therapy , Alveolar Bone Loss/metabolism , Alveolar Bone Loss/pathology , Cattle , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/cytology , Rats, Sprague-Dawley , Osteogenesis/drug effects , Tumor Necrosis Factor-alpha/metabolism , Binding Sites , Coculture Techniques , Osteoprotegerin/metabolism , Disease Models, Animal
10.
Mol Med Rep ; 30(2)2024 08.
Article in English | MEDLINE | ID: mdl-38873986

ABSTRACT

Chronic low­grade inflammation defines obesity as a metabolic disorder. Alterations in the structure of gut flora are strongly associated with obesity. Lactoferrin (LF) has a biological function in regulating intestinal flora. The present study aimed to investigate the therapeutic and anti­-inflammatory effects of LF in obese mice based on intestinal flora. A total of 30 C57BL/6 mice were divided into three groups consisting of 10 mice each. Subsequently, one group was fed a normal diet (Group K), another group was fed a high­fat diet (Group M) and the remaining group switched from regular drinking to drinking 2% LF water (Group Z2) after 2 weeks of high­fat diet; all mice were fed for 12 weeks. After the experiment, the mouse blood lipid and lipopolysaccharide levels, levels of inflammatory factors and intestinal tight junction proteins were assessed. Mouse stool samples were analyzed using 16S ribosomal RNA sequencing. The results showed that LF reduced serum total cholesterol, triglycerides and low­density lipoprotein levels, elevated high­density lipoprotein levels, suppressed metabolic endotoxemia and attenuated chronic low­grade inflammatory responses in obese mice. In addition, LF upregulated zonula occludens­1 and occludin protein expression levels in the intestine, thereby improving intestinal barrier integrity. LF altered the intestinal microbial structure of obese mice, reduced the ratio of Firmicutes and an elevated ratio of Bacteroidota, modifying the bacterial population to the increased relative abundance of Alistipes, Acidobacteriota, Psychrobacter and Bryobacter.


Subject(s)
Diet, High-Fat , Gastrointestinal Microbiome , Inflammation , Lactoferrin , Mice, Inbred C57BL , Mice, Obese , Obesity , Animals , Lactoferrin/pharmacology , Gastrointestinal Microbiome/drug effects , Mice , Obesity/metabolism , Obesity/drug therapy , Male , Inflammation/drug therapy , Inflammation/metabolism , Diet, High-Fat/adverse effects , Occludin/metabolism , Occludin/genetics , Lipopolysaccharides
11.
Food Chem Toxicol ; 190: 114817, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38880466

ABSTRACT

Human milk lactoferrin (hmLF) is a glycoprotein with well-known effects on immune function. Helaina Inc. has used a glycoengineered yeast, Komatagaella phaffii, to produce recombinant human lactoferrin (Helaina rhLF, Effera™) that is structurally similar to hmLF with intended uses as a food ingredient. However, earlier FDA reviews of rhLF were withdrawn due to insufficient safety data and unanswered safety questions the experts and FDA raised about the immunogenicity/immunotoxicity risks of orally ingested rhLF. Helaina organized a panel of leading scientists to build and vet a safety study roadmap containing the studies and safety endpoints needed to address these questions. Panelists participated in a one-day virtual workshop in June 2023 and ensuing discussions through July 2023. Relevant workshop topics included physicochemical properties of LF, regulatory history of bovine LF and rhLF as food ingredients in the FDA's generally recognized as safe (GRAS) program, and synopses of publicly available studies on the immunogenicity/alloimmunization, immunotoxicology, iron homeostasis, and absorption, distribution, metabolism, and excretion of rhLF. Panelists concluded that the safety study roadmap addresses the unanswered safety questions and the intended safe use of rhLF as a food ingredient for adults and agreed on broad applications of the roadmap to assess the safety and support GRAS of other recombinant milk proteins with immunomodulatory functions.


Subject(s)
Lactoferrin , Recombinant Proteins , Humans , Recombinant Proteins/toxicity , Animals , Food Safety , Saccharomycetales/genetics , Saccharomycetales/metabolism , United States Food and Drug Administration , United States , Cattle , Food Ingredients
12.
Nutrients ; 16(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38931261

ABSTRACT

The prevalence of allergic diseases has dramatically increased among children in recent decades. These conditions significantly impact the quality of life of allergic children and their families. Lactoferrin, a multifunctional glycoprotein found in various biological fluids, is emerging as a promising immunomodulatory agent that can potentially alleviate allergic diseases in children. Lactoferrin's multifaceted properties make it a compelling candidate for managing these conditions. Firstly, lactoferrin exhibits potent anti-inflammatory and antioxidant activities, which can mitigate the chronic inflammation characteristic of allergic diseases. Secondly, its iron-binding capabilities may help regulate the iron balance in allergic children, potentially influencing the severity of their symptoms. Lactoferrin also demonstrates antimicrobial properties, making it beneficial in preventing secondary infections often associated with respiratory allergies. Furthermore, its ability to modulate the immune response and regulate inflammatory pathways suggests its potential as an immune-balancing agent. This review of the current literature emphasises the need for further research to elucidate the precise roles of lactoferrin in allergic diseases. Harnessing the immunomodulatory potential of lactoferrin could provide a novel add-on approach to managing allergic diseases in children, offering hope for improved outcomes and an enhanced quality of life for paediatric patients and their families. As lactoferrin continues to capture the attention of researchers, its properties and diverse applications make it an intriguing subject of study with a rich history and a promising future.


Subject(s)
Hypersensitivity , Lactoferrin , Respiratory Tract Diseases , Child , Humans , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Hypersensitivity/drug therapy , Lactoferrin/therapeutic use , Quality of Life , Respiratory Tract Diseases/drug therapy
13.
Food Chem ; 456: 139818, 2024 Oct 30.
Article in English | MEDLINE | ID: mdl-38878531

ABSTRACT

This study aimed to develop complex coacervates utilizing lactoferrin (LF) and chia seed mucilage (CSM) for promoting intestinal delivery of quercetin (Q) and fortification of set yogurt. Three cross-linkers, including calcium chloride (CC), transglutaminase (TG), and polyphenolic complex (HP), were used to further reinforce the coacervate network. Cross-linked coacervates had higher values of coacervate yield, encapsulation efficiency, and loading capacity. They efficiently preserved Q under gastric condition (⁓87%-99%), with CSM-TG-Q-LF being most effective for intestinal delivery of Q. Moreover, digested pellets of the cross-linked coacervates displayed better antioxidant activity than the uncross-linked coacervates with CSM-TG-Q-LF pellets showing maximum bioactivity. The Q-loaded coacervates demonstrated superior assembly in the yogurt matrix compared to the unencapsulated Q. Moreover, the coacervate systems, especially CSM-TG-Q-LF significantly improved the textural properties of yogurt and the stability of Q in it. Therefore, CSM-TG-LF is a promising carrier to promote intestinal delivery and food application of hydrophobic molecules.


Subject(s)
Lactoferrin , Quercetin , Seeds , Yogurt , Seeds/chemistry , Yogurt/analysis , Lactoferrin/chemistry , Lactoferrin/metabolism , Quercetin/chemistry , Plant Mucilage/chemistry , Humans , Chenopodium quinoa/chemistry , Food, Fortified/analysis , Intestinal Mucosa/metabolism , Drug Delivery Systems/instrumentation
14.
Food Chem ; 456: 140056, 2024 Oct 30.
Article in English | MEDLINE | ID: mdl-38878546

ABSTRACT

In this study, carrageenan (CG), xanthan gum (XG) and locust bean gum (LBG), which can be used in infant formulas in China national standards, were selected to prepare LF-polysaccharide complexes to improve the stability of lactoferrin. The results showed that LF interacted more strongly with polysaccharides and did not affect the LF structure to a large extent when the pH and protein/polysaccharide mass ratio were 7 and 10:1 for LF-CG, 8 and 5:1 for LF-XG, 7 and 15:1 for LF-LBG. The zeta potential and fluorescence intensity of the LF-polysaccharide complexes displayed a decreasing trend with the increase in pH. When pH < 6, LF-CG and LF-XG exhibited precipitation and increased UV absorbance. Complexation between LF and CG/XG mainly attributed to electrostatic interactions, while LF and LBG form complexes based on hydrogen bonding or hydrophobic interactions. This study could provide a reference for the practical application of LF in infant formula.


Subject(s)
Infant Formula , Lactoferrin , Polysaccharides , Lactoferrin/chemistry , Hydrogen-Ion Concentration , Polysaccharides/chemistry , Infant Formula/chemistry , Galactans/chemistry , Polysaccharides, Bacterial/chemistry , Plant Gums/chemistry , Mannans/chemistry , Humans , Carrageenan/chemistry
15.
Sheng Wu Gong Cheng Xue Bao ; 40(6): 1895-1908, 2024 Jun 25.
Article in Chinese | MEDLINE | ID: mdl-38914499

ABSTRACT

Human lactoferrin (HLF), an essential nutrient found in breast milk, possesses antibacterial, anti-inflammatory, and immune-enhancing properties. In this study, the effects of three constitutive promoters (P21, P43, and Pveg) and three inducible promoters (Pgrac100, PxylA, and Ptet*) on the expression of HLF were compared using Bacillus subtilis G601 as the host strain. The results showed that the highest expression of HLF, reaching 651.57 µg/L, was achieved when regulated by the Ptet* promoter. Furthermore, the combinational optimization of ribosome binding site (RBS) and signal peptides was investigated, and the optimal combination of RBS6 and SPyycP resulted in increased HLF expression to 1 099.87 µg/L, with 498.68 µg/L being secreted extracellularly. To further enhance HLF secretion, the metal cations-related gene dltD was knocked out, leading to an extracellular HLF level of 637.28 µg/L. This study successfully demonstrated the secretory expression of HLF in B. subtilis through the selection and optimization of expression elements, laying the foundation for the development of efficient B. subtilis cell factories for lactoprotein synthesis.


Subject(s)
Bacillus subtilis , Lactoferrin , Promoter Regions, Genetic , Bacillus subtilis/metabolism , Bacillus subtilis/genetics , Lactoferrin/genetics , Lactoferrin/metabolism , Lactoferrin/biosynthesis , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
16.
Sci Rep ; 14(1): 14505, 2024 06 24.
Article in English | MEDLINE | ID: mdl-38914667

ABSTRACT

Identification of an early biomarker and effective testing device to differentiate dry eye disease secondary to autoimmune disease (Sjögren's syndrome dry eye disease) from non-Sjögren's dry eye disease are prerequisites for appropriate treatment. We aimed to demonstrate the capacity of a new photo-detection device to evaluate tear lactoferrin levels as a tool for differentiating systemic conditions associated with dry eye disease. Patients with non-Sjögren's and Sjögren's syndrome dry eye disease (n = 54 and n = 52, respectively) and controls (n = 11) were enrolled. All participants completed the Ocular Surface Disease Index questionnaire. Tear collection was performed with Schirmer test, and tear break-up time was examined using a slit lamp. Tear lactoferrin was evaluated using our newly developed photo-detection device. The average lactoferrin concentration was significantly lower in samples from patients with non-Sjögren's dry eye disease (0.337 ± 0.227 mg/mL, n = 54) and Sjögren's syndrome dry eye disease (0.087 ± 0.010 mg/mL, n = 52) than in control samples (1.272 ± 0.54 mg/mL, n = 11) (p < 0.0001). Further, lactoferrin levels were lower in patients with Sjögren's syndrome dry eye disease than in those with non-Sjögren's dry eye disease (p < 0.001). Our cost-effective, antibody-free, highly sensitive photo-detection device for evaluating tear lactoferrin levels can assist ophthalmologists in differentiating different types of dry eye diseases.


Subject(s)
Dry Eye Syndromes , Lactoferrin , Sjogren's Syndrome , Tears , Lactoferrin/analysis , Lactoferrin/metabolism , Humans , Tears/chemistry , Tears/metabolism , Sjogren's Syndrome/diagnosis , Sjogren's Syndrome/metabolism , Female , Middle Aged , Dry Eye Syndromes/diagnosis , Dry Eye Syndromes/metabolism , Male , Adult , Biomarkers/analysis , Diagnosis, Differential , Aged , Fluorescence
17.
Vet Microbiol ; 295: 110153, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38889618

ABSTRACT

Bovine leukemia virus (BLV) is a widespread virus that decreases milk production and quality in dairy cows. As crucial components of BLV, BLV-encoded microRNAs (BLV-miRNAs) affect BLV replication and may impact the synthesis of Lactoferrin (LTF), Lactoperoxidase (LPO), Alpha-lactalbumin (alpha-LA), and Beta-lactoglobulin (beta-LG). In this study, we investigated the targeting relationship between BLV-miRNAs and LTF, LPO, alpha-LA, and beta-LG in cow's milk. Additionally, we investigated the possible mechanisms by which BLV reduces milk quality. The results showed that cow's milk had significantly lower levels of LTF, LPO, and alpha-LA proteins in BLV-positive cows than in BLV-negative cows. BLV-△miRNAs (miRNA-deleted BLV) enhanced the reduction of LPO, alpha-LA, and beta-LG protein levels caused by BLV infection. Multiple BLV-miRNAs have binding sites with LTF and LPO mRNA; however, only BLV-miR-B1-5 P has a targeting relationship with LPO mRNA. The results revealed that BLV-miR-B1-5 P inhibits LPO protein expression by targeting LPO mRNA. However, BLV does not directly regulate the expression of LTF, alpha-LA, or beta-LG proteins through BLV-miRNAs.


Subject(s)
Lactalbumin , Lactoferrin , Lactoglobulins , Lactoperoxidase , Leukemia Virus, Bovine , MicroRNAs , Milk , Animals , Lactoferrin/genetics , Lactoferrin/metabolism , Lactoperoxidase/metabolism , Lactoperoxidase/genetics , Lactalbumin/genetics , Lactalbumin/metabolism , Cattle , Lactoglobulins/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Leukemia Virus, Bovine/genetics , Female , Enzootic Bovine Leukosis/virology , Enzootic Bovine Leukosis/genetics
18.
Int J Pharm ; 660: 124308, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38848800

ABSTRACT

In this paper, electrospinning and supercritical impregnation were coupled to produce polyurethane fibrous membranes loaded with mesoglycan and lactoferrin. The proposed methodology allowed the production of three skin wound healing bilayer systems: a first system containing mesoglycan loaded through electrospinning and lactoferrin loaded by supercritical impregnation, a second system where the use of the two techniques was reversed, and a third sample where the drugs were both encapsulated through a one-step process. SEM analysis demonstrated the formation of microfibers with a homogeneous drug distribution. The highest loadings were 0.062 g/g for mesoglycan and 0.013 g/g for lactoferrin. Then, hydrophilicity and liquid retention analyses were carried out to evaluate the possibility of using the manufacturers as active patches. The kinetic profiles, obtained through in vitro tests conducted using a Franz diffusion cell, proved that the diffusion of the active drugs followed a double-step release before attaining the equilibrium after about 30 h. When the electrospun membranes were placed in contact with HUVEC, HaCaT, and BJ cell lines, as human endothelial cells, keratinocytes, and fibroblasts, respectively, no cytotoxic events were assessed. Finally, the capacity of the most promising system to promote the healing process was performed by carrying out scratch tests on HaCat cells.


Subject(s)
Bandages , Polyurethanes , Wound Healing , Humans , Wound Healing/drug effects , Polyurethanes/chemistry , Cell Line , Lactoferrin/chemistry , Lactoferrin/administration & dosage , Human Umbilical Vein Endothelial Cells , Drug Liberation , Fibroblasts/drug effects , Polyethylene Glycols/chemistry , Hydrophobic and Hydrophilic Interactions , Membranes, Artificial , HaCaT Cells , Cell Survival/drug effects
19.
Int J Biol Macromol ; 272(Pt 1): 132812, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38825275

ABSTRACT

Complexes of polysaccharides and proteins have superior physicochemical and functional properties compared to single proteins or polysaccharides. In this study, lactoferrin-hyaluronic acid (LF-HA) complexes were prepared by both ultrasonic and thermal treatment. Appropriate preparation conditions, including ultrasonic and thermal treatment conditions, have been established. The complexes formed by different methods were structurally characterized by sodium dodecyl sulfate polyacrylamide gel electrophoresis, fourier transform infrared spectroscopy, and circular dichroism spectroscopy. Ultrasound formed non-covalent binding, while thermal treatment generated covalent bonding, altering the structure of LF. The LF-HA complexes showed improved heat stability, foaming stability, emulsifying activity and antioxidant capacity, but deceased foaming ability. Iron binding ability could only be improved by HA through thermal treatment. Moreover, the in vitro digestibility of LF-HA complexes decreased to below 80 % compared to LF.


Subject(s)
Hyaluronic Acid , Lactoferrin , Lactoferrin/chemistry , Hyaluronic Acid/chemistry , Antioxidants/chemistry , Hot Temperature , Ultrasonic Waves , Iron/chemistry , Spectroscopy, Fourier Transform Infrared
20.
Biomater Adv ; 162: 213924, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38875802

ABSTRACT

Chronic myeloid leukemia is a hematological cancer, where disease relapse and drug resistance are caused by bone-hosted-residual leukemia cells. An innovative resolution is bone-homing and selective-active targeting of anticancer loaded-nanovectors. Herein, ivermectin (IVM) and methyl dihydrojasmonate (MDJ)-loaded nanostructured lipid carriers (IVM-NLC) were formulated then dually decorated by lactoferrin (Lf) and alendronate (Aln) to optimize (Aln/Lf/IVM-NLC) for active-targeting and bone-homing potential, respectively. Aln/Lf/IVM-NLC (1 mg) revealed nano-size (73.67 ± 0.06 nm), low-PDI (0.43 ± 0.06), sustained-release of IVM (62.75 % at 140-h) and MDJ (78.7 % at 48-h). Aln/Lf/IVM-NLC afforded substantial antileukemic-cytotoxicity on K562-cells (4.29-fold lower IC50), higher cellular uptake and nuclear fragmentation than IVM-NLC with acceptable cytocompatibility on oral-epithelial-cells (as normal cells). Aln/Lf/IVM-NLC effectively upregulated caspase-3 and BAX (4.53 and 15.9-fold higher than IVM-NLC, respectively). Bone homing studies verified higher hydroxyapatite affinity of Aln/Lf/IVM-NLC (1 mg; 22.88 ± 0.01 % at 3-h) and higher metaphyseal-binding (1.5-fold increase) than untargeted-NLC. Moreover, Aln/Lf/IVM-NLC-1 mg secured 1.35-fold higher in vivo bone localization than untargeted-NLC, with lower off-target distribution. Ex-vivo hemocompatibility and in-vivo biocompatibility of Aln/Lf/IVM-NLC (1 mg/mL) were established, with pronounced amelioration of hepatic and renal toxicity compared to higher Aln doses. The innovative Aln/Lf/IVM-NLC could serve as a promising nanovector for bone-homing, active-targeted leukemia therapy.


Subject(s)
Alendronate , Drug Carriers , Ivermectin , Lactoferrin , Humans , Animals , Drug Carriers/chemistry , Lactoferrin/chemistry , Lactoferrin/pharmacology , Lactoferrin/administration & dosage , Alendronate/chemistry , Alendronate/pharmacology , Alendronate/administration & dosage , Ivermectin/chemistry , Ivermectin/analogs & derivatives , Ivermectin/pharmacology , Ivermectin/administration & dosage , Ivermectin/pharmacokinetics , K562 Cells , Nanoparticles/chemistry , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Bone and Bones/drug effects , Bone and Bones/metabolism , Lipids/chemistry , Apoptosis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL