1
|
Oliver-Cervelló L, López-Gómez P, Martin-Gómez H, Marion M, Ginebra MP, Mas-Moruno C. Functionalization of Alginate Hydrogels with a Multifunctional Peptide Supports Mesenchymal Stem Cell Adhesion and Reduces Bacterial Colonization. Chemistry 2024; 30:e202400855. [PMID: 39031737 DOI: 10.1002/chem.202400855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/07/2024] [Accepted: 06/19/2024] [Indexed: 07/22/2024]
Abstract
Hydrogels with cell adhesive moieties stand out as promising materials to enhance tissue healing and regeneration. Nonetheless, bacterial infections of the implants represent an unmet major concern. In the present work, we developed an alginate hydrogel modified with a multifunctional peptide containing the RGD cell adhesive motif in combination with an antibacterial peptide derived from the 1-11 region of lactoferrin (LF). The RGD-LF branched peptide was successfully anchored to the alginate backbone by carbodiimide chemistry, as demonstrated by 1H NMR and fluorescence measurements. The functionalized hydrogel presented desirable physicochemical properties (porosity, swelling and rheological behavior) to develop biomaterials for tissue engineering. The viability of mesenchymal stem cells (MSCs) on the peptide-functionalized hydrogels was excellent, with values higher than 85 % at day 1, and higher than 95 % after 14 days in culture. Moreover, the biological characterization demonstrated the ability of the hydrogels to significantly enhance ALP activity of MSCs as well as to decrease bacterial colonization of both Gram-positive and Gram-negative models. Such results prove the potential of the functionalized hydrogels as novel biomaterials for tissue engineering, simultaneously displaying cell adhesive activity and the capacity to prevent bacterial contamination, a dual bioactivity commonly not found for these types of hydrogels.
Collapse
Affiliation(s)
- Lluís Oliver-Cervelló
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
| | - Patricia López-Gómez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
| | - Helena Martin-Gómez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
| | - Mahalia Marion
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, 08028, Spain
| | - Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| |
Collapse
|
2
|
Babulic P, Cehlar O, Ondrovičová G, Moskalets T, Skrabana R, Leksa V. Lactoferrin Binds through Its N-Terminus to the Receptor-Binding Domain of the SARS-CoV-2 Spike Protein. Pharmaceuticals (Basel) 2024; 17:1021. [PMID: 39204126 PMCID: PMC11357225 DOI: 10.3390/ph17081021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Since Coronavirus disease 2019 (COVID-19) still presents a considerable threat, it is beneficial to provide therapeutic supplements against it. In this respect, glycoprotein lactoferrin (LF) and lactoferricin (LFC), a natural bioactive peptide yielded upon digestion from the N-terminus of LF, are of utmost interest, since both have been shown to reduce infections of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the virus responsible for COVID-19, in particular via blockade of the virus priming and binding. Here, we, by means of biochemical and biophysical methods, reveal that LF directly binds to the S-protein of SARS-CoV-2. We determined thermodynamic and kinetic characteristics of the complex formation and mapped the mutual binding sites involved in this interaction, namely the N-terminal region of LF and the receptor-binding domain of the S-protein (RBD). These results may not only explain many of the observed protective effects of LF and LFC in SARS-CoV-2 infection but may also be instrumental in proposing potent and cost-effective supplemental tools in the management of COVID-19.
Collapse
Affiliation(s)
- Patrik Babulic
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia; (P.B.); (G.O.); (T.M.)
- Department of Genetics, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| | - Ondrej Cehlar
- Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia;
| | - Gabriela Ondrovičová
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia; (P.B.); (G.O.); (T.M.)
| | - Tetiana Moskalets
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia; (P.B.); (G.O.); (T.M.)
| | - Rostislav Skrabana
- Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia;
| | - Vladimir Leksa
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia; (P.B.); (G.O.); (T.M.)
| |
Collapse
|
3
|
Biji CA, Balde A, Nazeer RA. Anti-inflammatory peptide therapeutics and the role of sulphur containing amino acids (cysteine and methionine) in inflammation suppression: A review. Inflamm Res 2024; 73:1203-1221. [PMID: 38769154 DOI: 10.1007/s00011-024-01893-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Inflammation serves as our body's immune response to combat infections, pathogens, viruses, and external stimuli. Inflammation can be classified into two types: acute inflammation and chronic inflammation. Non-steroidal anti-inflammatory medications (NSAIDs) are used to treat both acute and chronic inflammatory disorders. However, these treatments have various side effects such as reduced healing efficiency, peptic ulcers, gastrointestinal toxicities, etc. METHOD: This review assesses the potential of anti-inflammatory peptides (AIPs) derived from various natural sources, such as algae, fungi, plants, animals, and marine organisms. Focusing on peptides rich in cysteines and methionine, sulphur-containing amino acids known for their role in suppression of inflammation. RESULT Due to their varied biological activity, ability to penetrate cells, and low cytotoxicity, bioactive peptides have garnered interest as possible therapeutic agents. The utilisation of AIPs has shown great potential in the treatment of disorders associated with inflammation. AIPs can be obtained from diverse natural sources such as algae, fungi, plants, and animals. Cysteine and methionine are sulphur-containing amino acids that aid in the elimination of free radicals, hence assisting in the treatment of inflammatory diseases. CONCLUSION This review specifically examines several sources of AIPs including peptides that contain numerous cysteines and methionine. In addition, the biological characteristics of these amino acids and advancements in peptide delivery are also discussed.
Collapse
Affiliation(s)
- Catherin Ann Biji
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamilnadu, India
| | - Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamilnadu, India
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamilnadu, India.
| |
Collapse
|
4
|
Ramírez‐Sánchez DA, Canizalez‐Román A, León‐Sicairos N, Pérez Martínez G. The anticancer activity of bovine lactoferrin is reduced by deglycosylation and it follows a different pathway in cervix and colon cancer cells. Food Sci Nutr 2024; 12:3516-3528. [PMID: 38726451 PMCID: PMC11077203 DOI: 10.1002/fsn3.4020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/15/2024] [Accepted: 01/27/2024] [Indexed: 05/12/2024] Open
Abstract
Bovine lactoferrin (bLF) is a glycosylated protein with purported beneficial properties. The aim of this work was to determine the role of bLF glycosylation in the adhesion, internalization, and growth inhibition of cancer cells. The viability of cervix (HeLa) and colon (Caco-2) cancer cells (MTT assay and epifluorescence microscopy) was inhibited by bLF, while deglycosylated bLF (bLFdeg) had no effect. Adhesion to cell surfaces was quantified by immunofluorescence assay and showed that bLF was able to bind more efficiently to both cell lines than bLFdeg. Microscopic observations indicated that bLF glycosylation favored bLF binding to epithelial cells and that it was endocytosed through caveolin-1-mediated internalization. In addition, the mechanism of action of bLF on cancer cell proliferation was investigated by determining the amount of phosphorylated intermediates of signaling pathways such as epidermal growth factor receptor (EGFR), extracellular signal-regulated kinase (ERK) and protein kinase B (known as Akt). Chemoluminescence immunoassay of phosphorylated intermediates showed that bLF inhibited Akt phosphorylation, consistent with its growth inhibiting activity. This assay also indicated that the bLF receptor/signaling pathways may be different in the two cell lines, Caco-2 and HeLa. This work confirmed the effect of glycosylated bLF in inhibiting cancer cell growth and that glycosylation is required for optimal surface adhesion, internalization, and inhibition of the ERK/Akt pathway of cell proliferation through glycosylated cell surface receptors.
Collapse
Affiliation(s)
- Diana A. Ramírez‐Sánchez
- Programa Regional de Noroeste para el Doctorado en BiotecnologíaUniversidad Autónoma de Sinaloa Facultad de Ciencias Químico BiológicasCuliacanMexico
| | - Adrián Canizalez‐Román
- Unidad de Investigación, Facultad de MedicinaUniversidad Autónoma de SinaloaCuliacanMexico
- Servicios de Salud de SinaloaHospital de la MujerCuliacanMexico
| | - Nidia León‐Sicairos
- Unidad de Investigación, Facultad de MedicinaUniversidad Autónoma de SinaloaCuliacanMexico
- Servicios de Salud de Sinaloa, Departamento de Investigación del Hospital Pediátrico de SinaloaCuliacanMexico
| | - Gaspar Pérez Martínez
- Consejo Superior de Investigaciones CientificasInstituto de Agroquímica y Tecnología de AlimentosPaternaSpain
| |
Collapse
|
5
|
Vllahu M, Voli A, Licursi V, Zagami C, D'Amore A, Traulsen J, Woelffling S, Schmid M, Crickley R, Lisle R, Link A, Tosco A, Meyer TF, Boccellato F. Inflammation promotes stomach epithelial defense by stimulating the secretion of antimicrobial peptides in the mucus. Gut Microbes 2024; 16:2390680. [PMID: 39244776 PMCID: PMC11382725 DOI: 10.1080/19490976.2024.2390680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/10/2024] Open
Abstract
The mucus serves as a protective barrier in the gastrointestinal tract against microbial attacks. While its role extends beyond merely being a physical barrier, the extent of its active bactericidal properties remains unclear, and the mechanisms regulating these properties are not yet understood. We propose that inflammation induces epithelial cells to secrete antimicrobial peptides, transforming mucus into an active bactericidal agent. To investigate the properties of mucus, we previously developed mucosoid culture models that mimic the healthy human stomach epithelium. Similar to organoids, mucosoids are stem cell-driven cultures; however, the cells are cultivated on transwells at air-liquid interface. The epithelial cells of mucosoids form a polarized monolayer, allowing differentiation into all stomach lineages, including mucus-secreting cells. This setup facilitates the secretion and accumulation of mucus on the apical side of the mucosoids, enabling analysis of its bactericidal effects and protein composition, including antimicrobial peptides. Our findings show that TNFα, IL1β, and IFNγ induce the secretion of antimicrobials such as lactotransferrin, lipocalin2, complement component 3, and CXCL9 into the mucus. This antimicrobial-enriched mucus can partially eliminate Helicobacter pylori, a key stomach pathogen. The bactericidal activity depends on the concentration of each antimicrobial and their gene expression is higher in patients with inflammation and H.pylori-associated chronic gastritis. However, we also find that H. pylori infection can reduce the expression of antimicrobial encoding genes promoted by inflammation. These findings suggest that controlling antimicrobial secretion in the mucus is a critical component of epithelial immunity. However, pathogens like H. pylori can overcome these defenses and survive in the mucosa.
Collapse
Affiliation(s)
- Megi Vllahu
- Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Antonia Voli
- Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Valerio Licursi
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR) of Italy c/o Department of Biology and Biotechnology ''C. Darwin'', Sapienza University, Rome, Italy
| | - Claudia Zagami
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Antonella D'Amore
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Jan Traulsen
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Sara Woelffling
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Monika Schmid
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Robbie Crickley
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Richard Lisle
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Alessandra Tosco
- Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy
| | - Thomas F Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
- Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Christian Albrecht University of Kiel and University Hospital Schleswig-Holstein - Campus Kiel, Kiel, Germany
| | - Francesco Boccellato
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
6
|
Elizarova AY, Sokolov AV, Vasilyev VB. Ceruloplasmin Reduces the Lactoferrin/Oleic Acid Antitumor Complex-Mediated Release of Heme-Containing Proteins from Blood Cells. Int J Mol Sci 2023; 24:16711. [PMID: 38069040 PMCID: PMC10706732 DOI: 10.3390/ijms242316711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/24/2023] [Accepted: 10/29/2023] [Indexed: 12/18/2023] Open
Abstract
Our previous study showed that not only bovine lactoferrin (LF), the protein of milk and neutrophils, but also the human species forms complexes with oleic acid (OA) that inhibit tumor growth. Repeated injections of human LF in complex with OA (LF/8OA) to hepatoma-carrying mice decelerated tumor growth and increased animals' longevity. However, whether the effect of the LF/8OA complex is directed exclusively against malignant cells was not studied. Hence, its effect on normal blood cells was assayed, along with its possible modulation of ceruloplasmin (CP), the preferred partner of LF among plasma proteins. The complex LF/8OA (6 μM) caused hemolysis, unlike LF alone or BSA/8OA (250 μM). The activation of neutrophils with exocytosis of myeloperoxidase (MPO), a potent oxidant, was induced by 1 μM LF/8OA, whereas BSA/8OA had a similar effect at a concentration increased by an order. The egress of heme-containing proteins, i.e., MPO and hemoglobin, from blood cells affected by LF/8OA was followed by a pronounced oxidative/halogenating stress. CP, which is the natural inhibitor of MPO, added at a concentration of 2 mol per 1 mol of LF/8OA abrogated its cytotoxic effect. It seems likely that CP can be used effectively in regulating the LF/8OA complex's antitumor activity.
Collapse
Affiliation(s)
| | - Alexey V. Sokolov
- Institute of Experimental Medicine, 197376 Saint-Petersburg, Russia; (A.Y.E.); (V.B.V.)
| | | |
Collapse
|
7
|
Ostrówka M, Duda-Madej A, Pietluch F, Mackiewicz P, Gagat P. Testing Antimicrobial Properties of Human Lactoferrin-Derived Fragments. Int J Mol Sci 2023; 24:10529. [PMID: 37445717 DOI: 10.3390/ijms241310529] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Lactoferrin, an iron-binding glycoprotein, plays a significant role in the innate immune system, with antibacterial, antivirial, antifungal, anticancer, antioxidant and immunomodulatory functions reported. It is worth emphasizing that not only the whole protein but also its derived fragments possess antimicrobial peptide (AMP) activity. Using AmpGram, a top-performing AMP classifier, we generated three novel human lactoferrin (hLF) fragments: hLF 397-412, hLF 448-464 and hLF 668-683, predicted with high probability as AMPs. For comparative studies, we included hLF 1-11, previously confirmed to kill some bacteria. With the four peptides, we treated three Gram-negative and three Gram-positive bacterial strains. Our results indicate that none of the three new lactoferrin fragments have antimicrobial properties for the bacteria tested, but hLF 1-11 was lethal against Pseudomonas aeruginosa. The addition of serine protease inhibitors with the hLF fragments did not enhance their activity, except for hLF 1-11 against P. aeruginosa, which MIC dropped from 128 to 64 µg/mL. Furthermore, we investigated the impact of EDTA with/without serine protease inhibitors and the hLF peptides on selected bacteria. We stress the importance of reporting non-AMP sequences for the development of next-generation AMP prediction models, which suffer from the lack of experimentally validated negative dataset for training and benchmarking.
Collapse
Affiliation(s)
- Michał Ostrówka
- Faculty of Biotechnology, University of Wrocław, Fryderyka Joliot-Curie 14a, 50-137 Wrocław, Poland
| | - Anna Duda-Madej
- Department of Microbiology, Faculty of Medicine, Wrocław Medical University, Chałubińskiego 4, 50-368 Wrocław, Poland
| | - Filip Pietluch
- Faculty of Biotechnology, University of Wrocław, Fryderyka Joliot-Curie 14a, 50-137 Wrocław, Poland
| | - Paweł Mackiewicz
- Faculty of Biotechnology, University of Wrocław, Fryderyka Joliot-Curie 14a, 50-137 Wrocław, Poland
| | - Przemysław Gagat
- Faculty of Biotechnology, University of Wrocław, Fryderyka Joliot-Curie 14a, 50-137 Wrocław, Poland
| |
Collapse
|
8
|
Liu WJ, Chang YS, Tseng KC, Yu MH. Activity of bovine lactoferrin in resistance to white spot syndrome virus infection in shrimp. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 142:104651. [PMID: 36736936 DOI: 10.1016/j.dci.2023.104651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 06/18/2023]
Abstract
White spot syndrome virus (WSSV) is a notorious pathogen that has plagued shrimp farming worldwide for decades. To date, there are no known treatments that are effective against this virus. Lactoferrin (LF) is a protein with many bioactivities, including antiviral properties. In this study, the activities and mechanisms of bovine LF (bLF) against WSSV were analyzed. Our results showed that bLF treatment significantly reduced shrimp mortalities caused by WSSV infection. bLF was found to have the ability to bind to surfaces of both host cells and WSSV virions. These bindings may have been a result of bLF interactions with the host cellular chitin binding protein and F1 ATP synthase β subunit protein and the WSSV structural proteins VP28, VP110, VP150 and VP160B. bLF demonstrated potential for development as an anti-WSSV agent in shrimp culture. Furthermore, these reactionary proteins may play a role in WSSV infection.
Collapse
Affiliation(s)
- Wang-Jing Liu
- Department of Earth and Life Science, University of Taipei, Taipei, Taiwan.
| | - Yun-Shiang Chang
- Department of Biomedical Sciences, Da-Yeh University, Changhua, Taiwan
| | - Kou-Chun Tseng
- Department of Earth and Life Science, University of Taipei, Taipei, Taiwan
| | - Meng-Hua Yu
- Department of Earth and Life Science, University of Taipei, Taipei, Taiwan
| |
Collapse
|
9
|
Ohradanova-Repic A, Praženicová R, Gebetsberger L, Moskalets T, Skrabana R, Cehlar O, Tajti G, Stockinger H, Leksa V. Time to Kill and Time to Heal: The Multifaceted Role of Lactoferrin and Lactoferricin in Host Defense. Pharmaceutics 2023; 15:1056. [PMID: 37111542 PMCID: PMC10146187 DOI: 10.3390/pharmaceutics15041056] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Lactoferrin is an iron-binding glycoprotein present in most human exocrine fluids, particularly breast milk. Lactoferrin is also released from neutrophil granules, and its concentration increases rapidly at the site of inflammation. Immune cells of both the innate and the adaptive immune system express receptors for lactoferrin to modulate their functions in response to it. On the basis of these interactions, lactoferrin plays many roles in host defense, ranging from augmenting or calming inflammatory pathways to direct killing of pathogens. Complex biological activities of lactoferrin are determined by its ability to sequester iron and by its highly basic N-terminus, via which lactoferrin binds to a plethora of negatively charged surfaces of microorganisms and viruses, as well as to mammalian cells, both normal and cancerous. Proteolytic cleavage of lactoferrin in the digestive tract generates smaller peptides, such as N-terminally derived lactoferricin. Lactoferricin shares some of the properties of lactoferrin, but also exhibits unique characteristics and functions. In this review, we discuss the structure, functions, and potential therapeutic uses of lactoferrin, lactoferricin, and other lactoferrin-derived bioactive peptides in treating various infections and inflammatory conditions. Furthermore, we summarize clinical trials examining the effect of lactoferrin supplementation in disease treatment, with a special focus on its potential use in treating COVID-19.
Collapse
Affiliation(s)
- Anna Ohradanova-Repic
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Romana Praženicová
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| | - Laura Gebetsberger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Tetiana Moskalets
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| | - Rostislav Skrabana
- Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Ondrej Cehlar
- Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Gabor Tajti
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Hannes Stockinger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Vladimir Leksa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| |
Collapse
|
10
|
Naidu SAG, Clemens RA, Naidu AS. SARS-CoV-2 Infection Dysregulates Host Iron (Fe)-Redox Homeostasis (Fe-R-H): Role of Fe-Redox Regulators, Ferroptosis Inhibitors, Anticoagulants, and Iron-Chelators in COVID-19 Control. J Diet Suppl 2023; 20:312-371. [PMID: 35603834 DOI: 10.1080/19390211.2022.2075072] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Severe imbalance in iron metabolism among SARS-CoV-2 infected patients is prominent in every symptomatic (mild, moderate to severe) clinical phase of COVID-19. Phase-I - Hypoxia correlates with reduced O2 transport by erythrocytes, overexpression of HIF-1α, altered mitochondrial bioenergetics with host metabolic reprogramming (HMR). Phase-II - Hyperferritinemia results from an increased iron overload, which triggers a fulminant proinflammatory response - the acute cytokine release syndrome (CRS). Elevated cytokine levels (i.e. IL6, TNFα and CRP) strongly correlates with altered ferritin/TF ratios in COVID-19 patients. Phase-III - Thromboembolism is consequential to erythrocyte dysfunction with heme release, increased prothrombin time and elevated D-dimers, cumulatively linked to severe coagulopathies with life-threatening outcomes such as ARDS, and multi-organ failure. Taken together, Fe-R-H dysregulation is implicated in every symptomatic phase of COVID-19. Fe-R-H regulators such as lactoferrin (LF), hemoxygenase-1 (HO-1), erythropoietin (EPO) and hepcidin modulators are innate bio-replenishments that sequester iron, neutralize iron-mediated free radicals, reduce oxidative stress, and improve host defense by optimizing iron metabolism. Due to its pivotal role in 'cytokine storm', ferroptosis is a potential intervention target. Ferroptosis inhibitors such as ferrostatin-1, liproxstatin-1, quercetin, and melatonin could prevent mitochondrial lipid peroxidation, up-regulate antioxidant/GSH levels and abrogate iron overload-induced apoptosis through activation of Nrf2 and HO-1 signaling pathways. Iron chelators such as heparin, deferoxamine, caffeic acid, curcumin, α-lipoic acid, and phytic acid could protect against ferroptosis and restore mitochondrial function, iron-redox potential, and rebalance Fe-R-H status. Therefore, Fe-R-H restoration is a host biomarker-driven potential combat strategy for an effective clinical and post-recovery management of COVID-19.
Collapse
Affiliation(s)
| | - Roger A Clemens
- Department of International Regulatory Science, University of Southern California School of Pharmacy, Los Angeles, CA, USA
| | | |
Collapse
|
11
|
Tutanov O, Tamkovich S. The Influence of Proteins on Fate and Biological Role of Circulating DNA. Int J Mol Sci 2022; 23:7224. [PMID: 35806228 PMCID: PMC9266439 DOI: 10.3390/ijms23137224] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Circulating DNA has already proven itself as a valuable tool in translational medicine. However, one of the overlooked areas of circulating DNA research is its association with different proteins, despite considerable evidence that this association might impact DNA's fate in circulation and its biological role. In this review, we attempt to shed light on current ideas about circulating DNA origins and forms of circulation, known biological effects, and the clinical potential of circulating tumor deoxyribonucleoprotein complexes.
Collapse
Affiliation(s)
| | - Svetlana Tamkovich
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, 630090 Novosibirsk, Russia;
| |
Collapse
|
12
|
Excretion of Cell-Free and Cell-Associated Zika Virus into Breast Milk of Infected Dams and Identification of Antiviral Factors. Viruses 2022; 14:v14050851. [PMID: 35632593 PMCID: PMC9144681 DOI: 10.3390/v14050851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/15/2022] [Accepted: 04/17/2022] [Indexed: 02/04/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne RNA virus belonging to the Flavivirus genus of the Flaviviridae family. During the 60 years following its discovery in 1947, ZIKV caused little concern for public health as the associated infection was reported as mostly asymptomatic or inducing mild symptoms. However, since 2013, severe neurological symptoms have been associated with ZIKV infection, compelling the World Health Organization to declare a Public Health Emergency of International Concern. Among those symptoms, neurological birth defects may affect children born to mothers infected during pregnancy. Additionally, during the past 8 years, ZIKV transmission through breastfeeding has repeatedly been suggested in epidemiological studies and demonstrated on a mouse model by our team. To better understand the biological factors controlling ZIKV transmission through breastfeeding, we investigated the nature of the viral entities excreted in the breast milk of infected dams and evaluated viral transmission to breastfed pups. We show that both cell-free and cell-associated virus is excreted into breast milk and that ZIKV is efficiently transmitted to the breastfed pups. Additionally, we studied murine breast milk cell types, and identified a majority of mammary luminal cells. Finally, we investigated the effect on ZIKV infectivity of several breast milk components that are antiviral against different viruses such as lactoferrin (LF) and lactalbumin (LA), or free fatty acids (FFA). We showed no effect of LF and LA, whereas FFA inactivated the virus. These results bring new insight concerning the mechanisms of ZIKV transmission during breastfeeding and identify biological factors modulating it. These elements should be considered in risk assessment of ZIKV mother-to-child transmission.
Collapse
|
13
|
Fluid-Phase Endocytosis and Lysosomal Degradation of Bovine Lactoferrin in Lung Cells. Pharmaceutics 2022; 14:pharmaceutics14040855. [PMID: 35456688 PMCID: PMC9032238 DOI: 10.3390/pharmaceutics14040855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 12/10/2022] Open
Abstract
The iron-binding protein lactoferrin and the cell-penetrating peptides derived from its sequence utilise endocytosis to enter different cell types. The full-length protein has been extensively investigated as a potential therapeutic against a range of pathogenic bacteria, fungi, and viruses, including SARS-CoV-2. As a respiratory antiviral agent, several activity mechanisms have been demonstrated for lactoferrin, at the extracellular and plasma membrane levels, but as a protein that enters cells it may also have intracellular antiviral activity. Characterisation of lactoferrin’s binding, endocytic traffic to lysosomes, or recycling endosomes for exocytosis is lacking, especially in lung cell models. Here, we use confocal microscopy, flow cytometry, and degradation assays to evaluate binding, internalisation, endocytic trafficking, and the intracellular fate of bovine lactoferrin in human lung A549 cells. In comparative studies with endocytic probes transferrin and dextran, we show that lactoferrin binds to negative charges on the cell surface and actively enters cells via fluid-phase endocytosis, in a receptor-independent manner. Once inside the cell, we show that it is trafficked to lysosomes where it undergoes degradation within two hours. These findings provide opportunities for investigating both lactoferrin and derived cell-penetrating peptides activities of targeting intracellular pathogens.
Collapse
|
14
|
Nakamura M, Sato A. Glycan-binding Properties of Basic Whey Protein Lactoferrin and Its Application in Nerve Regenerative Medicine. TRENDS GLYCOSCI GLYC 2022. [DOI: 10.4052/tigg.1957.1e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Masao Nakamura
- Department of Peptidomics, Sasaki Institute, Sasaki Foundation
| | - Atsushi Sato
- School of Bioscience and Biotechnology, Tokyo University of Technology
| |
Collapse
|
15
|
Nakamura M, Sato A. Glycan-binding Properties of Basic Whey Protein Lactoferrin and Its Application in Nerve Regenerative Medicine. TRENDS GLYCOSCI GLYC 2022. [DOI: 10.4052/tigg.1957.1j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Masao Nakamura
- Department of Peptidomics, Sasaki Institute, Sasaki Foundation
| | - Atsushi Sato
- School of Bioscience and Biotechnology, Tokyo University of Technology
| |
Collapse
|
16
|
Rawson A, Saxena V, Gao H, Hooks J, Xuei X, McGuire P, Hato T, Hains DS, Anderson RM, Schwaderer AL. A Pilot Single Cell Analysis of the Zebrafish Embryo Cellular Responses to Uropathogenic Escherichia coli Infection. Pathog Immun 2022; 7:1-18. [PMID: 35178490 PMCID: PMC8843076 DOI: 10.20411/pai.v7i1.479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/06/2021] [Indexed: 11/23/2022] Open
Abstract
Background: Uropathogenic Escherichia coli (UPEC) infections are common and when they disseminate can be of high morbidity.
Methods: We studied the effects of UPEC infection using single cell RNA sequencing (scRNAseq) in zebrafish. Bulk RNA sequencing has historically been used to evaluate gene expression patterns, but scRNAseq allows gene expression to be evaluated at the single cell level and is optimal for evaluating heterogeneity within cell types and rare cell types. Zebrafish cohorts were injected with either saline or UPEC,and scRNAseq and canonical pathway analyses were performed.
Results: Canonical pathway analysis of scRNAseq data provided key information regarding innate immune pathways in the cells determined to be thymus cells, ionocytes, macrophages/monocytes, and pronephros cells. Pathways activated in thymus cells included interleukin 6 (IL-6) signaling and production of reactive oxygen species. Fc receptor-mediated phagocytosis was a leading canonical pathway in the pronephros and macrophages. Genes that were downregulated in UPEC vs saline exposed embryos involved the cellular response to the Gram-negative endotoxin lipopolysaccharide (LPS) and included Forkhead Box O1a (Foxo1a), Tribbles Pseudokinase 3 (Trib3), Arginase 2 (Arg2) and Polo Like Kinase 3 (Plk3).
Conclusions: Because 4-day post fertilization zebrafish embryos only have innate immune systems, the scRNAseq provides insights into pathways and genes that cell types utilize in the bacterial response. Based on our analysis, we have identified genes and pathways that might serve as genetic targets for treatment and further investigation in UPEC infections at the single cell level.
Collapse
Affiliation(s)
- Ashley Rawson
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology
| | - Vijay Saxena
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology
| | - Hongyu Gao
- Indiana University School of Medicine, Department of Medical & Molecular Genetics
| | - Jenaya Hooks
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology
| | - Xiaoling Xuei
- Indiana University School of Medicine, Department of Medical & Molecular Genetics
| | - Patrick McGuire
- Indiana University School of Medicine, Department of Medical & Molecular Genetics
| | - Takashi Hato
- Indiana University School of Medicine, Department of Medicine, Division of Nephrology
| | - David S. Hains
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology
| | - Ryan M. Anderson
- University of Chicago, Section of Endocrinology, Diabetes and Metabolism
- CORRESPONDING AUTHOR Andrew Schwaderer, Indiana University School of Medicine, Riley Hospital for Children, 699 Riley Hospital Dr., RR230, Indianapolis, IN 46202; Phone: 317-274-2527;
| | - Andrew L. Schwaderer
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology
- CORRESPONDING AUTHOR Ryan M Anderson, University of Chicago, Medicine-Endocrinology, Chicago, IL 60637;
| |
Collapse
|
17
|
Gruden Š, Poklar Ulrih N. Diverse Mechanisms of Antimicrobial Activities of Lactoferrins, Lactoferricins, and Other Lactoferrin-Derived Peptides. Int J Mol Sci 2021; 22:ijms222011264. [PMID: 34681923 PMCID: PMC8541349 DOI: 10.3390/ijms222011264] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 12/22/2022] Open
Abstract
Lactoferrins are an iron-binding glycoprotein that have important protective roles in the mammalian body through their numerous functions, which include antimicrobial, antitumor, anti-inflammatory, immunomodulatory, and antioxidant activities. Among these, their antimicrobial activity has been the most studied, although the mechanism behind antimicrobial activities remains to be elucidated. Thirty years ago, the first lactoferrin-derived peptide was isolated and showed higher antimicrobial activity than the native lactoferrin lactoferricin. Since then, numerous studies have investigated the antimicrobial potencies of lactoferrins, lactoferricins, and other lactoferrin-derived peptides to better understand their antimicrobial activities at the molecular level. This review defines the current antibacterial, antiviral, antifungal, and antiparasitic activities of lactoferrins, lactoferricins, and lactoferrin-derived peptides. The primary focus is on their different mechanisms of activity against bacteria, viruses, fungi, and parasites. The role of their structure, amino-acid composition, conformation, charge, hydrophobicity, and other factors that affect their mechanisms of antimicrobial activity are also reviewed.
Collapse
|
18
|
Desgraupes S, Hubert M, Gessain A, Ceccaldi PE, Vidy A. Mother-to-Child Transmission of Arboviruses during Breastfeeding: From Epidemiology to Cellular Mechanisms. Viruses 2021; 13:1312. [PMID: 34372518 PMCID: PMC8310101 DOI: 10.3390/v13071312] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/18/2021] [Accepted: 06/30/2021] [Indexed: 12/20/2022] Open
Abstract
Most viruses use several entry sites and modes of transmission to infect their host (parenteral, sexual, respiratory, oro-fecal, transplacental, transcutaneous, etc.). Some of them are known to be essentially transmitted via arthropod bites (mosquitoes, ticks, phlebotomes, sandflies, etc.), and are thus named arthropod-borne viruses, or arboviruses. During the last decades, several arboviruses have emerged or re-emerged in different countries in the form of notable outbreaks, resulting in a growing interest from scientific and medical communities as well as an increase in epidemiological studies. These studies have highlighted the existence of other modes of transmission. Among them, mother-to-child transmission (MTCT) during breastfeeding was highlighted for the vaccine strain of yellow fever virus (YFV) and Zika virus (ZIKV), and suggested for other arboviruses such as Chikungunya virus (CHIKV), dengue virus (DENV), and West Nile virus (WNV). In this review, we summarize all epidemiological and clinical clues that suggest the existence of breastfeeding as a neglected route for MTCT of arboviruses and we decipher some of the mechanisms that chronologically occur during MTCT via breastfeeding by focusing on ZIKV transmission process.
Collapse
Affiliation(s)
- Sophie Desgraupes
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Département Virologie, Institut Pasteur, 75015 Paris, France; (M.H.); (A.G.); (P.-E.C.)
- Université de Paris, 75013 Paris, France
- UMR Centre National de la Recherche Scientifique 3569, Institut Pasteur, 75015 Paris, France
| | - Mathieu Hubert
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Département Virologie, Institut Pasteur, 75015 Paris, France; (M.H.); (A.G.); (P.-E.C.)
- Université de Paris, 75013 Paris, France
- UMR Centre National de la Recherche Scientifique 3569, Institut Pasteur, 75015 Paris, France
| | - Antoine Gessain
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Département Virologie, Institut Pasteur, 75015 Paris, France; (M.H.); (A.G.); (P.-E.C.)
- Université de Paris, 75013 Paris, France
- UMR Centre National de la Recherche Scientifique 3569, Institut Pasteur, 75015 Paris, France
| | - Pierre-Emmanuel Ceccaldi
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Département Virologie, Institut Pasteur, 75015 Paris, France; (M.H.); (A.G.); (P.-E.C.)
- Université de Paris, 75013 Paris, France
- UMR Centre National de la Recherche Scientifique 3569, Institut Pasteur, 75015 Paris, France
| | - Aurore Vidy
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Département Virologie, Institut Pasteur, 75015 Paris, France; (M.H.); (A.G.); (P.-E.C.)
- Université de Paris, 75013 Paris, France
- UMR Centre National de la Recherche Scientifique 3569, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
19
|
Nayak A, Tiozzo C, Lin X, Mejia C, Gurzenda E, Kim M, Hanna N. Is Lactoferrin Supplementation Beneficial for All Preterm Infants? Am J Perinatol 2021; 40:680-687. [PMID: 34058763 DOI: 10.1055/s-0041-1730433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Human milk (HM) has antibacterial properties due to the presence of immune-modulators, including lactoferrin (LF). This study will determine effect(s) of HM maturation, fortification, and storage conditions on LF levels and its antibacterial properties. STUDY DESIGN HM samples (n = 30) were obtained from preterm and term mothers. The LF levels were analyzed by ELISA, and the antibacterial activity was measured after inoculation with Escherichia coli. RESULTS The highest level of LF in preterm HM was observed in the first week of lactation. However, storage of preterm HM at 4°C decreased LF levels significantly. Both LF levels and antibacterial activity in preterm HM was lower compared with term HM, but significantly higher than donor HM even after HM-based fortification. LF supplementation of donor HM improved its antibacterial activity. CONCLUSION Preterm infants fed donor HM, formula, or stored HM at 4°C may benefits from LF supplementation to improve HM antibacterial properties. KEY POINTS · Milk LF levels vary with storage and maturity.. · Donor milk is deficient in LF even after adding HM-based fortification.. · Donor HM and formula fed infants may benefit from LF..
Collapse
Affiliation(s)
- Amrita Nayak
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York
| | - Caterina Tiozzo
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York.,Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Xinhua Lin
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Claudia Mejia
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Ellen Gurzenda
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Maureen Kim
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York
| | - Nazeeh Hanna
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York.,Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| |
Collapse
|
20
|
Naidu SAG, Clemens RA, Pressman P, Zaigham M, Davies KJA, Naidu AS. COVID-19 during Pregnancy and Postpartum. J Diet Suppl 2020; 19:78-114. [PMID: 33164606 DOI: 10.1080/19390211.2020.1834047] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
As the COVID-19 pandemic intensified the global health crisis, the containment of SARS-CoV-2 infection in pregnancies, and the inherent risk of vertical transmission of virus from mother-to-fetus (or neonate) poses a major concern. Most COVID-19-Pregnancy patients showed mild to moderate COVID-19 pneumonia with no pregnancy loss and no congenital transmission of the virus; however, an increase in hypoxia-induced preterm deliveries was apparent. Also, the breastmilk of several mothers with COVID-19 tested negative for the virus. Taken together, the natural barrier function during pregnancy and postpartum seems to deter the SARS-CoV-2 transmission from mother-to-child. This clinical observation warrants to explore the maternal-fetal interface and identify the innate defense factors for prevention and control of COVID-19-Pregnancy. Lactoferrin (LF) is a potent antiviral iron-binding protein present in the maternal-fetal interface. In concert with immune co-factors, maternal-LF modulates chemokine release and lymphocyte migration and amplify host defense during pregnancy. LF levels during pregnancy may resolve hypertension via down-regulation of ACE2; consequently, may limit the membrane receptor access to SARS-CoV-2 for cellular entry. Furthermore, an LF-derived peptide (LRPVAA) has been shown to block ACE receptor activity in vitro. LF may also reduce viral docking and entry into host cells and limit the early phase of COVID-19 infection. An in-depth understanding of LF and other soluble mammalian milk-derived innate antiviral factors may provide insights to reduce co-morbidities and vertical transmission of SARS-CoV-2 infection and may lead to the development of effective nutraceutical supplements.
Collapse
Affiliation(s)
| | - Roger A Clemens
- School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | | | - Mehreen Zaigham
- Department of Obstetrics & Gynecology, Skåne University Hospital, Malmö, Sweden
| | - Kelvin J A Davies
- Division of Biogerontology, Leonard Davis School of Gerontology, The University of Southern California, Los Angeles, CA, USA.,Division of Molecular & Computational Biology, Dornsife College of Letters, Arts, and Sciences, The University of Southern California, Los Angeles, CA, USA.,Department Biochemistry & Molecular Medicine, Keck School of Medicine of USC, The University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
21
|
Nakamura M, Matsuzaki T, Iimori A, Sato A. Harnessing the chondroitin sulfate-binding characteristics of human lactoferrin to neutralize neurite outgrowth inhibition. Biochem Biophys Res Commun 2020; 534:1076-1082. [PMID: 33129446 DOI: 10.1016/j.bbrc.2020.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 10/06/2020] [Indexed: 11/30/2022]
Abstract
Human lactoferrin (hLF) is a glycosaminoglycan (GAG)-binding protein involved in various biological functions. It consists of two globular functional domains, referred to as the N- and C-lobes. Both heparin (HP) and heparan sulfate (HS) bind to the N-lobe domain of hLF. Although some biological functions of hLF such as neuroprotective effects and cancer growth inhibition are regulated by its binding to HS, the binding characteristics of hLF with other GAG subtypes, and their effects on biological activities are still poorly understood. Here, we report that hLF binds to chondroitin sulfate (CS)-E, a GAG subtype involved in various neurodegenerative diseases. The α-helical content of hLF, which is an indicator of changes in the secondary structure of hLF, increased in the presence of CS-C, CS-D, or CS-E, but not in the presence of HP, HS, CS-A, or CS-B. This structural change was also observed in the N-lobe, the N-terminal half region of the hLF. Additionally, the thermal stability of the N-lobe showed a dose-dependent improvement in the presence of CS-E, but not in the presence of HP. This indicates that the binding mode of hLF/N-lobe to CS-E may differ from that of HP. hLF was also found to neutralize CS-E-induced inhibition of neurite outgrowth and neuronal growth cone collapse, which are neurodegenerative responses to spinal cord injury, in cultured dorsal root ganglion neurons. Thus, hLF is a promising drug candidate for the treatment of CS-E-induced neurodegenerative diseases such as spinal cord injury.
Collapse
Affiliation(s)
- Masao Nakamura
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1 Katakura, Hachioji, Tokyo, 192-0982, Japan; Department of Peptidomics, Sasaki Institute, Sasaki Foundation, 2-2 Kandasurugadai, Chiyoda-ku, Tokyo, 101-0062, Japan.
| | - Takumi Matsuzaki
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1 Katakura, Hachioji, Tokyo, 192-0982, Japan
| | - Ami Iimori
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1 Katakura, Hachioji, Tokyo, 192-0982, Japan
| | - Atsushi Sato
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1 Katakura, Hachioji, Tokyo, 192-0982, Japan.
| |
Collapse
|
22
|
Li HY, Li P, Yang HG, Yao QQ, Huang SN, Wang JQ, Zheng N. Investigation and comparison of the protective activities of three functional proteins-lactoferrin, α-lactalbumin, and β-lactoglobulin-in cerebral ischemia reperfusion injury. J Dairy Sci 2020; 103:4895-4906. [PMID: 32229112 DOI: 10.3168/jds.2019-17725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/22/2020] [Indexed: 01/05/2023]
Abstract
The objective of this study was to evaluate the protection conferred by lactoferrin, α-lactalbumin, and β-lactoglobulin in cerebral ischemia reperfusion (I/R) injury. Rat pheochromocytoma (PC12) cells were used to construct an oxygen and glucose deprivation model in vitro, and ICR mice underwent carotid artery "ligation-relaxation" to construct a cerebral I/R injury model in vivo. The levels of toll-like receptor 4 (TLR4) and downstream factors including nuclear factor-κB, tumor necrosis factor-α, and IL-1β were measured. Metabonomics detection and data mining were conducted to identify the specific metabolic sponsor of the 3 proteins. The results showed that lactoferrin, α-lactalbumin, and β-lactoglobulin protected neurons from cerebral I/R injury by increasing the level of bopindolol and subsequently inhibiting the TLR4-related pathway to different degrees; β-lactoglobulin had the strongest activity of the 3 proteins. In summary, this study is the first to investigate and compare the protective effects of lactoferrin, α-lactalbumin, and β-lactoglobulin in a cerebral stroke model. The results implicate TLR4 as a novel target of the 3 bioactive proteins to prevent cerebral I/R injury.
Collapse
Affiliation(s)
- Hui-Ying Li
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Peng Li
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Key Laboratory of Quality and Safety Risk Assessment for Dairy Products, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Huai-Gu Yang
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Qian-Qian Yao
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Sheng-Nan Huang
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Jia-Qi Wang
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China.
| | - Nan Zheng
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China.
| |
Collapse
|
23
|
Basauri A, González-Fernández C, Fallanza M, Bringas E, Fernandez-Lopez R, Giner L, Moncalián G, de la Cruz F, Ortiz I. Biochemical interactions between LPS and LPS-binding molecules. Crit Rev Biotechnol 2020; 40:292-305. [DOI: 10.1080/07388551.2019.1709797] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Arantza Basauri
- Departamento de Ingenierías Química y Biomolecular, Universidad de Cantabria, Santander, Spain
| | | | - Marcos Fallanza
- Departamento de Ingenierías Química y Biomolecular, Universidad de Cantabria, Santander, Spain
| | - Eugenio Bringas
- Departamento de Ingenierías Química y Biomolecular, Universidad de Cantabria, Santander, Spain
| | - Raúl Fernandez-Lopez
- Instituto de Biomedicina y Biotecnologia (IBBTEC), CSIC, Universidad de Cantabria, Santander, Spain
| | - Laura Giner
- Instituto de Biomedicina y Biotecnologia (IBBTEC), CSIC, Universidad de Cantabria, Santander, Spain
| | - Gabriel Moncalián
- Instituto de Biomedicina y Biotecnologia (IBBTEC), CSIC, Universidad de Cantabria, Santander, Spain
| | - Fernando de la Cruz
- Instituto de Biomedicina y Biotecnologia (IBBTEC), CSIC, Universidad de Cantabria, Santander, Spain
| | - Inmaculada Ortiz
- Departamento de Ingenierías Química y Biomolecular, Universidad de Cantabria, Santander, Spain
| |
Collapse
|
24
|
Nakanishi J, Suzuki S, Yoshida K, Hirata-Tsuchiya S, Haruyama N, Yamada S, Shiba H. Dentin phosphoprotein inhibits lipopolysaccharide-induced macrophage activation independent of its serine/aspartic acid-rich repeats. Arch Oral Biol 2019; 110:104634. [PMID: 31855746 DOI: 10.1016/j.archoralbio.2019.104634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 11/10/2019] [Accepted: 12/11/2019] [Indexed: 10/25/2022]
Abstract
OBJECTIVE The objective of this study was to investigate the effects of dentin phosphoprotein (DPP) on lipopolysaccharide-induced inflammatory responses of macrophages in vitro. DESIGN Wildtype and mutant recombinant dentin phosphoprotein (rDPP) proteins were generated using a mammalian expression system. Macrophages, phorbol 12-myristate 13-acetate-differentiated THP-1 cells, were stimulated with lipopolysaccharide in the absence or presence of rDPP proteins. After the 24-hr incubation, the inflammatory gene expression levels were examined by quantitative reverse-transcription polymerase chain reaction and the amount of secreted TNF-α protein was evaluated by enzyme-linked immunosorbent assay. Furthermore, the subcellular localization of exogenously added rDPP was examined by immunocytochemistry, and the direct binding of rDPP to lipopolysaccharide was quantified by solid-phase binding assay. RESULTS rDPP dose-dependently reduced the expression of lipopolysaccharide-induced inflammatory genes, such as TNFα, IL-1β, and IL-8, and TNF-α protein secretion from the macrophages. Furthermore, mutant rDPP having a shortened serine/aspartic acid-rich repeats (SDrr) was also able to inhibit lipopolysaccharide-induced inflammatory responses of macrophages. rDPP was localized adjacent to the cellular membrane rather than in the cytoplasm, and rDPP was able to bind to lipopolysaccharide. These results suggested that rDPP inhibited lipopolysaccharide-induced inflammatory responses by binding to lipopolysaccharide. CONCLUSIONS In addition to the well-known functions of DPP for dentin mineralization that depend on the SDrr, we demonstrated that DPP possesses anti-inflammatory effects on lipopolysaccharide-stimulated macrophages that are independent of the SDrr.
Collapse
Affiliation(s)
- Jun Nakanishi
- Department of Biological Endodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Shigeki Suzuki
- Department of Biological Endodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan; Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan.
| | - Kazuma Yoshida
- Department of Biological Endodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Shizu Hirata-Tsuchiya
- Department of Biological Endodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Naoto Haruyama
- Section of Orthodontics and Dentofacial Orthopedics, Faculty of Dental Science, Kyushu University, Fukuoka, 812-5852, Japan
| | - Satoru Yamada
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan
| | - Hideki Shiba
- Department of Biological Endodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| |
Collapse
|
25
|
Kühnle A, Galuska CE, Zlatina K, Galuska SP. The Bovine Antimicrobial Peptide Lactoferricin Interacts with Polysialic Acid without Loss of Its Antimicrobial Activity against Escherichia coli. Animals (Basel) 2019; 10:E1. [PMID: 31861263 PMCID: PMC7022438 DOI: 10.3390/ani10010001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/09/2019] [Accepted: 12/15/2019] [Indexed: 01/28/2023] Open
Abstract
The lactoferrin-derived peptide lactoferricin (LFcin) belongs to the family of antimicrobial peptides, and its bovine form has already been successfully applied to counteract enterohemorrhagic Escherichia coli (EHEC) infection. Recently, it was described that LFcin interacts with the sugar polymer polysialic acid (polySia) and that the binding of lactoferrin to polySia is mediated by LFcin, included in the N-terminal domain of lactoferrin. For this reason, the impact of polySia on the antimicrobial activity of bovine LFcin was investigated. Initially, the interaction of LFcin was characterized in more detail by native agarose gel electrophoresis, demonstrating that a chain length of 10 sialic acid residues was necessary to bind LFcin, whereas approximately twice-as-long chains were needed to detect binding of lactoferrin. Remarkably, the binding of polySia showed, independently of the chain length, no impact on the antimicrobial effects of LFcin. Thus, LFcin binds polySia without loss of its protective activity as an antimicrobial peptide.
Collapse
Affiliation(s)
- Andrea Kühnle
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany; (A.K.); (C.E.G.); (K.Z.)
- Institute of Biochemistry, Faculty of Medicine, Justus-Liebig-University, Friedrichstr. 24, 35392 Giessen, Germany
| | - Christina E. Galuska
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany; (A.K.); (C.E.G.); (K.Z.)
- Institute of Biochemistry, Faculty of Medicine, Justus-Liebig-University, Friedrichstr. 24, 35392 Giessen, Germany
| | - Kristina Zlatina
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany; (A.K.); (C.E.G.); (K.Z.)
| | - Sebastian P. Galuska
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany; (A.K.); (C.E.G.); (K.Z.)
- Institute of Biochemistry, Faculty of Medicine, Justus-Liebig-University, Friedrichstr. 24, 35392 Giessen, Germany
| |
Collapse
|
26
|
Chen M, Wen F, Zhang Y, Li P, Zheng N, Wang J. Determination of native lactoferrin in milk by HPLC on HiTrapTM Heparin HP column. FOOD ANAL METHOD 2019. [DOI: 10.1007/s12161-019-01572-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
27
|
Tamkovich S, Laktionov P. Cell-surface-bound circulating DNA in the blood: Biology and clinical application. IUBMB Life 2019; 71:1201-1210. [PMID: 31091350 DOI: 10.1002/iub.2070] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/17/2019] [Indexed: 01/04/2023]
Abstract
Cell-surface-bound extracellular DNA (csbDNA) is present on the outer membrane of blood cells, including both red blood cells and leukocytes. Although less well characterized than cell-free DNA (cfDNA) in plasma and serum, leukocyte and red blood cell csbDNA form a considerable fraction of the blood extracellular nucleic acids pool, with typically at least comparable amount of DNA occurring bound to the outer surface of cells as compared with circulating free DNA in plasma. The cellular origin of csbDNA is not clear; however, as with cfDNA, in patients with cancer a proportion is derived from the tumor, thus making it potentially a useful source of DNA for cancer diagnosis, prognosis, and monitoring. © 2019 IUBMB Life, 71(9):1201-1210, 2019.
Collapse
Affiliation(s)
- Svetlana Tamkovich
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia.,Novosibirsk National Research State University, Novosibirsk, Russia
| | - Pavel Laktionov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia.,Meshalkin Novosibirsk Research Institute of Circulation Pathology, Novosibirsk, Russia
| |
Collapse
|
28
|
Polysialic Acid Modulates the Binding of External Lactoferrin in Neutrophil Extracellular Traps. BIOLOGY 2019; 8:biology8020020. [PMID: 30925725 PMCID: PMC6627751 DOI: 10.3390/biology8020020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/28/2019] [Accepted: 03/21/2019] [Indexed: 02/07/2023]
Abstract
Neutrophil extracellular traps (NETs) are formed by neutrophils during inflammation. Among other things, these DNA constructs consist of antimicrobial proteins such as lactoferrin and histones. With these properties, NETs capture and destroy invading microorganisms. The carbohydrate polysialic acid (polySia) interacts with both lactoferrin and histones. Previous experiments demonstrated that, in humans, lactoferrin inhibits the release of NET and that this effect is supported by polySia. In this study, we examined the interplay of lactoferrin and polySia in already-formed NETs from bovine neutrophils. The binding of polySia was considered to occur at the lactoferricin (LFcin)-containing domain of lactoferrin. The interaction with the peptide LFcin was studied in more detail using groups of defined polySia chain lengths, which suggested a chain-length-dependent interaction mechanism with LFcin. The LFcin domain of lactoferrin was found to interact with DNA. Therefore, the possibility that polySia influences the integration of lactoferrin into the DNA-structures of NETs was tested by isolating bovine neutrophils and inducing NETosis. Experiments with NET fibers saturated with lactoferrin demonstrated that polySia initiates the incorporation of external lactoferrin in already-loaded NETs. Thus, polySia may modulate the constituents of NET.
Collapse
|
29
|
Wang B, Timilsena YP, Blanch E, Adhikari B. Lactoferrin: Structure, function, denaturation and digestion. Crit Rev Food Sci Nutr 2017; 59:580-596. [DOI: 10.1080/10408398.2017.1381583] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Bo Wang
- Food Research and Innovation Centre, School of Science, RMIT University, Melbourne, VIC, Australia
| | - Yakindra Prasad Timilsena
- Food Research and Innovation Centre, School of Science, RMIT University, Melbourne, VIC, Australia
- Materials Science and Engineering, CSIRO Manufacturing Flagship, Clayton South, VIC, Australia
| | - Ewan Blanch
- Food Research and Innovation Centre, School of Science, RMIT University, Melbourne, VIC, Australia
| | - Benu Adhikari
- Food Research and Innovation Centre, School of Science, RMIT University, Melbourne, VIC, Australia
- Materials Science and Engineering, CSIRO Manufacturing Flagship, Clayton South, VIC, Australia
| |
Collapse
|
30
|
El-Hawiet A. A Simple, Sensitive, and Label-Free Platform for the Quantification of Lactoferrin in Camel and Goat Milk Based on Thin-Layer Chromatography. Chromatographia 2017. [DOI: 10.1007/s10337-017-3414-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
31
|
Hu L, Hu X, Long K, Gao C, Dong HL, Zhong Q, Gao XM, Gong FY. Extraordinarily potent proinflammatory properties of lactoferrin-containing immunocomplexes against human monocytes and macrophages. Sci Rep 2017; 7:4230. [PMID: 28652573 PMCID: PMC5484712 DOI: 10.1038/s41598-017-04275-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 05/14/2017] [Indexed: 12/25/2022] Open
Abstract
Lactoferrin (LTF), an important first line defense molecule against infection, is a common target for humoral autoimmune reactions in humans. Since LTF is a multifunctional protein capable of activating innate immune cells via various surface receptors, we hypothesized that LTF-containing immune complexes (ICs) (LTF-ICs), likely formed in patients with high titer anti-LTF autoantibodies, could possess unique monocyte/macrophage-activating properties compared with other ICs. ELISA analysis on serum samples from rheumatoid arthritis (RA) patients (n = 80) and healthy controls (n = 35) for anti-LTF autoantibodies confirmed a positive correlation between circulating LTF-specific IgG and RA. ICs between human LTF and LTF-specific IgG purified from patient sera or immunized rabbits and mice, but not control ICs, LTF or Abs alone, elicited strong production of TNF-α and IL-1β by freshly fractionated human peripheral blood monocytes and monocytes-derived macrophages. Furthermore, LTF-ICs utilized both membrane-anchored CD14 and CD32a (FcγRIIa) to trigger monocyte activation in an internalization-, Toll-like receptor (TLR)4- and TLR9-dependent manner, and also that LTF-IC-induced cytokine production was blocked by specific inhibitors of caspase-1, NF-κB and MAPK. These results uncover a possible pathway for LTF-ICs perpetuating local inflammation and contributing to the pathogenesis of autoimmune diseases by triggering activation of infiltrating monocytes or tissue macrophages in vivo.
Collapse
Affiliation(s)
- Lulu Hu
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xiaomin Hu
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Kai Long
- Department of Immunology, Peking University Health Science Center, Beijing, China.,Department of Physiology, Jiujiang College, Jiangxi Province, China
| | - Chenhui Gao
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Hong-Liang Dong
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Qiao Zhong
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xiao-Ming Gao
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China.
| | - Fang-Yuan Gong
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
32
|
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; 12:251. [PMID: 27829421 PMCID: PMC5103330 DOI: 10.1186/s12917-016-0878-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/02/2016] [Indexed: 12/12/2022] Open
Abstract
Background Lactoferrin (LF) is an 80 kDa glycoprotein which is known for its effects against bacteria, viruses and other pathogens. It also has a high potential in nutrition therapy and welfare of people and a variety of animals, including piglets. The ability to bind lipopolysaccharide (LPS) is one of the described anti-inflammatory mechanisms of LF. Previous studies suggested that cells can be stimulated even by LPS-free LF. Therefore, the aim of our study was to bring additional information about this possibility. Porcine monocyte derived macrophages (MDMF) and human embryonic kidney (HEK) cells were stimulated with unpurified LF in complex with LPS and with purified LF without bound LPS. Results Both cell types were stimulated with unpurified as well as purified LF. On the other hand, neither HEK0 cells not expressing any TLR nor HEK4a cells transfected with TLR4 produced any pro-inflammatory cytokine transcripts after stimulation with purified LF. This suggests that purified LF without LPS stimulates cells via another receptor than TLR4. An alternative, TLR4-independent, pathway was further confirmed by analyses of the NF-kappa-B-inducing kinase (NIK) activation. Western blot analyses showed NIK which activates different NFκB subunits compared to LF-LPS signaling via TLR4. Though, this confirmed an alternative pathway which is used by the purified LF free of LPS. This stimulation of MDMF led to low, but significant amounts of pro-inflammatory cytokines, which can be considered as a positive stimulation of the immune system. Conclusion Our results suggest that LF’s ability is not only to bind LPS, but LF itself may be a stimulant of pro-inflammatory pathways.
Collapse
Affiliation(s)
- Nada Zemankova
- Veterinary Research Institute, Hudcova 296/70, Brno, 621 00, Czech Republic.,Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Katarina Chlebova
- Veterinary Research Institute, Hudcova 296/70, Brno, 621 00, Czech Republic.,Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Jan Matiasovic
- Veterinary Research Institute, Hudcova 296/70, Brno, 621 00, Czech Republic
| | - Jana Prodelalova
- Veterinary Research Institute, Hudcova 296/70, Brno, 621 00, Czech Republic
| | - Jan Gebauer
- Veterinary Research Institute, Hudcova 296/70, Brno, 621 00, Czech Republic.,Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Martin Faldyna
- Veterinary Research Institute, Hudcova 296/70, Brno, 621 00, Czech Republic.
| |
Collapse
|
33
|
Kim WS, Ohashi M, Shimazaki KI. Inhibitory Effects of Synthetic Peptides Containing Bovine Lactoferrin C-lobe Sequence on Bacterial Growth. Korean J Food Sci Anim Resour 2016; 36:452-7. [PMID: 27621684 PMCID: PMC5018503 DOI: 10.5851/kosfa.2016.36.4.452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/15/2016] [Accepted: 06/15/2016] [Indexed: 11/12/2022] Open
Abstract
Lactoferrin is a glycoprotein with various biological effects, with antibacterial activity being one of the first effects reported. This glycoprotein suppresses bacterial growth through bacteriostatic or bactericidal action. It also stimulates the growth of certain kinds of bacteria such as lactic acid bacteria and bifidobacteria. In this study, Asn-Leu-Asn-Arg was selected and chemically synthesized based on the partial sequences of bovine lactoferrin tryptic fragments. Synthetic Asn-Leu-Asn-Arg suppressed the growth of Pseudomonas fluorescens, P. syringae and Escherichia coli. P. fluorescens is a major psychrotrophic bacteria found in raw and pasteurized milk, which decreases milk quality. P. syringae is a harmful infectious bacterium that damages plants. However, synthetic Asn-Leu-Asn-Arg did not inhibit the growth of Lactobacillus acidophilus. It is expected that this synthetic peptide would be the first peptide sequence from the bovine lactoferrin C-lobe that shows antibacterial activity.
Collapse
Affiliation(s)
- Woan-Sub Kim
- Department of Animal Life and Environmental Science, Hankyong National University, Anseong 17579, Korea
| | - Midori Ohashi
- Research Faculty of Agriculture, Hokkaido University, Sapporo 060-8589, Japan
| | - Kei-Ichi Shimazaki
- Research Faculty of Agriculture, Hokkaido University, Sapporo 060-8589, Japan
| |
Collapse
|
34
|
Donovan SM. The Role of Lactoferrin in Gastrointestinal and Immune Development and Function: A Preclinical Perspective. J Pediatr 2016; 173 Suppl:S16-28. [PMID: 27234407 DOI: 10.1016/j.jpeds.2016.02.072] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The early postnatal period is a critical time for gastrointestinal (GI) and immune development. Neonates fed mother's milk have more rapid GI and immune development than fed-formula infants. In addition, clinical and epidemiologic data provide strong evidence that breastfeeding reduces the incidence and/or severity of infectious diseases. Lactoferrin is a 77 kDa, iron-binding glycoprotein that is present at high concentration in human milk compared with bovine milk and infant formula. It is a multifunctional protein that mediates many of the physiological processes in which breastfed infants have advantages over their formula-fed peers, including promoting GI and immune development, protection from infections, and improved cognitive development. Feeding bovine lactoferrin or recombinant human lactoferrin was well tolerated and stimulated intestinal cell proliferation and increased villus length and crypt depth in piglets. Lactoferrin also influenced both systemic and GI immune development by stimulating a balanced T-helper-1/T-helper-2 cytokine immune response. Further, there was a tendency for immune cells to secrete more anti-inflammatory cytokines in an unstimulated state, while being primed for a robust pro-inflammatory response when presented with a bacterial trigger in piglets fed lactoferrin. These findings support clinical studies demonstrating benefits of dietary lactoferrin in the prevention of infections, late onset sepsis, and necrotizing enterocolitis.
Collapse
Affiliation(s)
- Sharon M Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL.
| |
Collapse
|
35
|
Zumoffen CM, Massa E, Caille AM, Munuce MJ, Ghersevich SA. Effects of lactoferrin, a protein present in the female reproductive tract, on parameters of human sperm capacitation and gamete interaction. Andrology 2015; 3:1068-75. [PMID: 26445132 DOI: 10.1111/andr.12093] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 07/10/2015] [Accepted: 07/20/2015] [Indexed: 11/28/2022]
Abstract
In a recent study, lactoferrin (LF) was detected in human oviductal secretion. The protein was able to bind to oocytes and sperm, and modulated gamete interaction. The aim of the present study was to investigate the effect of LF on parameters related to human sperm capacitation and sperm-zona pellucida interaction. Semen samples were obtained from healthy normozoospermic donors (n = 7). Human follicular fluids and oocytes were collected from patients undergoing in vitro fertilization. Motile sperm obtained by swim-up were incubated for 6 or 22 h under capacitating conditions with LF (0-100 μg/mL). After incubations, viability, motility, presence of α-d-mannose receptors (using a fluorescent probe on mannose coupled to bovine serum albumin), spontaneous and induced acrosome reaction (assessed with Pisum sativum agglutinin conjugated to fluorescein isothiocyanate), and tyrosine phosphorylation of sperm proteins were evaluated. Sperm-zona pellucida interaction in the presence of LF was investigated using the hemizone assay. The presence of LF did not affect sperm viability or motility, but caused a dose-dependent significant decrease in sperm α-d-mannose-binding sites, and the effect was already significant with the lowest concentration of the protein used after 22 h incubation. Dose-dependent significant increases in both induced acrosome reaction and tyrosine phosphorylation of sperm proteins were observed in the presence of LF. The present data indicate that LF modulates parameters of sperm function. The inhibition of gamete interaction by LF could be partially explained by the decrease in sperm d-mannose-binding sites. The presence of the LF promoted sperm capacitation in vitro.
Collapse
Affiliation(s)
- C M Zumoffen
- Area of Clinical Biochemistry, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - E Massa
- Area of Clinical Biochemistry, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - A M Caille
- Area of Clinical Biochemistry, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - M J Munuce
- Area of Clinical Biochemistry, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - S A Ghersevich
- Area of Clinical Biochemistry, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| |
Collapse
|
36
|
Hu L, Gao CH, Hong C, Zhong Q, Dong HL, Gao XM. Expression, purification, and breast cancer cell inhibiting effect of recombinant human lactoferrin C-lobe. Biosci Biotechnol Biochem 2015; 80:257-63. [PMID: 26405758 DOI: 10.1080/09168451.2015.1088376] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Lactoferrin (LTF), a multifunctional glycoprotein of the transferrin family mainly found in exotic secretions in mammals, is an important defense molecule against not only microbial invasion but also tumors. It folds into two globular domains (N- and C-lobes) each containing an iron-binding site. The cationic antimicrobial peptide in N-lobe is known to exert anti-tumor effect via a non-receptor-mediated pathway. However, whether LTF C-lobe also contributes to its anti-tumor activity remains to be investigated. In this study, a human LTF fragment (amino acid residues 343-682) covering the C-lobe was expressed with a histidine tag in E. coli and the purified polypeptide refolded through a series of buffer changing procedure. The resultant recombinant protein caused significant growth arrest of breast carcinoma cells MDA-MB-231 in a dose- and time-dependent manner, evidently via induction of apoptosis of the cell. Our data suggest a positive role for the C-lobe of human LTF in controlling tumors in vitro.
Collapse
Affiliation(s)
- Lulu Hu
- a Institute of Biology and Medical Sciences, School of Biology and Basic Medical Science, Soochow University , Suzhou , China
| | - Chen-Hui Gao
- a Institute of Biology and Medical Sciences, School of Biology and Basic Medical Science, Soochow University , Suzhou , China
| | - Chao Hong
- a Institute of Biology and Medical Sciences, School of Biology and Basic Medical Science, Soochow University , Suzhou , China
| | - Qiao Zhong
- a Institute of Biology and Medical Sciences, School of Biology and Basic Medical Science, Soochow University , Suzhou , China
| | - Hong-Liang Dong
- a Institute of Biology and Medical Sciences, School of Biology and Basic Medical Science, Soochow University , Suzhou , China
| | - Xiao-Ming Gao
- a Institute of Biology and Medical Sciences, School of Biology and Basic Medical Science, Soochow University , Suzhou , China
| |
Collapse
|
37
|
Lecaille F, Lalmanach G, Andrault PM. Antimicrobial proteins and peptides in human lung diseases: A friend and foe partnership with host proteases. Biochimie 2015; 122:151-68. [PMID: 26341472 DOI: 10.1016/j.biochi.2015.08.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/31/2015] [Indexed: 12/20/2022]
Abstract
Lung antimicrobial proteins and peptides (AMPs) are major sentinels of innate immunity by preventing microbial colonization and infection. Nevertheless bactericidal activity of AMPs against Gram-positive and Gram-negative bacteria is compromised in patients with chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF) and asthma. Evidence is accumulating that expression of harmful human serine proteases, matrix metalloproteases and cysteine cathepsins is markedely increased in these chronic lung diseases. The local imbalance between proteases and protease inhibitors compromises lung tissue integrity and function, by not only degrading extracellular matrix components, but also non-matrix proteins. Despite the fact that AMPs are somewhat resistant to proteolytic degradation, some human proteases cleave them efficiently and impair their antimicrobial potency. By contrast, certain AMPs may be effective as antiproteases. Host proteases participate in concert with bacterial proteases in the degradation of key innate immunity peptides/proteins and thus may play immunomodulatory activities during chronic lung diseases. In this context, the present review highlights the current knowledge and recent discoveries on the ability of host enzymes to interact with AMPs, providing a better understanding of the role of human proteases in innate host defense.
Collapse
Affiliation(s)
- Fabien Lecaille
- INSERM, UMR 1100, Pathologies Respiratoires: Protéolyse et Aérosolthérapie, Centre d'Etude des Pathologies Respiratoires, Equipe 2: « Mécanismes Protéolytiques dans l'Inflammation », Université François Rabelais, F-37032 Tours cedex, France.
| | - Gilles Lalmanach
- INSERM, UMR 1100, Pathologies Respiratoires: Protéolyse et Aérosolthérapie, Centre d'Etude des Pathologies Respiratoires, Equipe 2: « Mécanismes Protéolytiques dans l'Inflammation », Université François Rabelais, F-37032 Tours cedex, France
| | - Pierre-Marie Andrault
- INSERM, UMR 1100, Pathologies Respiratoires: Protéolyse et Aérosolthérapie, Centre d'Etude des Pathologies Respiratoires, Equipe 2: « Mécanismes Protéolytiques dans l'Inflammation », Université François Rabelais, F-37032 Tours cedex, France
| |
Collapse
|
38
|
Barrientos L, Bignon A, Gueguen C, de Chaisemartin L, Gorges R, Sandré C, Mascarell L, Balabanian K, Kerdine-Römer S, Pallardy M, Marin-Esteban V, Chollet-Martin S. Neutrophil extracellular traps downregulate lipopolysaccharide-induced activation of monocyte-derived dendritic cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:5689-98. [PMID: 25339673 DOI: 10.4049/jimmunol.1400586] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Polymorphonuclear neutrophils (PMN) play a central role in inflammation and participate in its control, notably by modulating dendritic cell (DC) functions via soluble mediators or cell-cell contacts. Neutrophil extracellular traps (NETs) released by PMN could play a role in this context. To evaluate NET effects on DC maturation, we developed a model based on monocyte-derived DC (moDC) and calibrated NETs isolated from fresh human PMN. We found that isolated NETs alone had no discernable effect on moDC. In contrast, they downregulated LPS-induced moDC maturation, as shown by decreased surface expression of HLA-DR, CD80, CD83, and CD86, and by downregulated cytokine production (TNF-α, IL-6, IL-12, IL-23), with no increase in the expression of tolerogenic DC genes. Moreover, the presence of NETs during moDC maturation diminished the capacity of these moDC to induce T lymphocyte proliferation in both autologous and allogeneic conditions, and modulated CD4(+) T lymphocyte polarization by promoting the production of Th2 cytokines (IL-5 and IL-13) and reducing that of Th1 and Th17 cytokines (IFN-γ and IL-17). Interestingly, the expression and activities of the lymphoid chemokine receptors CCR7 and CXCR4 on moDC were not altered when moDC matured in the presence of NETs. Together, these findings reveal a new role for NETs in adaptive immune responses, modulating some moDC functions and thereby participating in the control of inflammation.
Collapse
Affiliation(s)
- Lorena Barrientos
- INSERM, Unité Mixte de Recherche-S 996, "Cytokines, chimiokines et immunopathologie," Université Paris-Sud, 92296 Châtenay-Malabry and Clamart, France; Université Paris-Sud, Faculté de Pharmacie, 92296 Châtenay-Malabry, France
| | - Alexandre Bignon
- INSERM, Unité Mixte de Recherche-S 996, "Cytokines, chimiokines et immunopathologie," Université Paris-Sud, 92296 Châtenay-Malabry and Clamart, France; Laboratory of Excellence in Research on Medication and Innovative Therapeutics, 92296 Clamart, France
| | | | - Luc de Chaisemartin
- INSERM, Unité Mixte de Recherche-S 996, "Cytokines, chimiokines et immunopathologie," Université Paris-Sud, 92296 Châtenay-Malabry and Clamart, France; Université Paris-Sud, Faculté de Pharmacie, 92296 Châtenay-Malabry, France; Assistance Publique Hôpitaux de Paris, Groupe Hospitalier Paris Nord Val de Seine, Hôpital Bichat, Unité d'Immunologie (Auto-immunité et Hypersensibilités), 75018 Paris, France
| | - Roseline Gorges
- INSERM, Unité Mixte de Recherche-S 996, "Cytokines, chimiokines et immunopathologie," Université Paris-Sud, 92296 Châtenay-Malabry and Clamart, France; Université Paris-Sud, Faculté de Pharmacie, 92296 Châtenay-Malabry, France
| | - Catherine Sandré
- INSERM, Unité Mixte de Recherche-S 996, "Cytokines, chimiokines et immunopathologie," Université Paris-Sud, 92296 Châtenay-Malabry and Clamart, France; Université Paris-Sud, Faculté de Pharmacie, 92296 Châtenay-Malabry, France
| | | | - Karl Balabanian
- INSERM, Unité Mixte de Recherche-S 996, "Cytokines, chimiokines et immunopathologie," Université Paris-Sud, 92296 Châtenay-Malabry and Clamart, France; Laboratory of Excellence in Research on Medication and Innovative Therapeutics, 92296 Clamart, France
| | - Saadia Kerdine-Römer
- INSERM, Unité Mixte de Recherche-S 996, "Cytokines, chimiokines et immunopathologie," Université Paris-Sud, 92296 Châtenay-Malabry and Clamart, France; Université Paris-Sud, Faculté de Pharmacie, 92296 Châtenay-Malabry, France
| | - Marc Pallardy
- INSERM, Unité Mixte de Recherche-S 996, "Cytokines, chimiokines et immunopathologie," Université Paris-Sud, 92296 Châtenay-Malabry and Clamart, France; Université Paris-Sud, Faculté de Pharmacie, 92296 Châtenay-Malabry, France
| | - Viviana Marin-Esteban
- INSERM, Unité Mixte de Recherche-S 996, "Cytokines, chimiokines et immunopathologie," Université Paris-Sud, 92296 Châtenay-Malabry and Clamart, France; Université Paris-Sud, Faculté de Pharmacie, 92296 Châtenay-Malabry, France
| | - Sylvie Chollet-Martin
- INSERM, Unité Mixte de Recherche-S 996, "Cytokines, chimiokines et immunopathologie," Université Paris-Sud, 92296 Châtenay-Malabry and Clamart, France; Université Paris-Sud, Faculté de Pharmacie, 92296 Châtenay-Malabry, France; Assistance Publique Hôpitaux de Paris, Groupe Hospitalier Paris Nord Val de Seine, Hôpital Bichat, Unité d'Immunologie (Auto-immunité et Hypersensibilités), 75018 Paris, France
| |
Collapse
|
39
|
Sohrabi SM, Niazi A, Chahardoli M, Hortamani A, Setoodeh P. In silico investigation of lactoferrin protein characterizations for the prediction of anti-microbial properties. MOLECULAR BIOLOGY RESEARCH COMMUNICATIONS 2014; 3:85-100. [PMID: 27843978 PMCID: PMC5047140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Lactoferrin (Lf) is an iron-binding multi-functional glycoprotein which has numerous physiological functions such as iron transportation, anti-microbial activity and immune response. In this study, different in silico approaches were exploited to investigate Lf protein properties in a number of mammalian species. Results showed that the iron-binding site, DNA and RNA-binding sites, signal peptides and transferrin motifs in the Lf structure were highly conserved. Examined sequences showed three conserved motifs which were repeated twice in the Lf structure, demonstrating ancient duplication events in its gene. Also, results suggest that the functional domains in mammalian Lf proteins are Zinc finger, Tubulin/FtsZ, GTPase, α/β hydrolase and Zinc knuckle. The potential site for nucleic acid binding and the major DNA and RNA- binding sites in this protein were found in the lactoferricin (Lfc) fragment. Due to its high positive charge, Lf is able to bind a large number of compounds. Our analysis also revealed that the interactions between Lf and ITLN1, LYZ, CSN2, and CD14 proteins played an important role in the protective activities of Lf. Analysis for the prediction of secondary structures indicated that high amounts of α-helix, β-strand and β-sheet were present in Lf. The high degree of conservation among mammalian Lf proteins indicates that there is a close relationship between these proteins, reflecting their important role.
Collapse
Affiliation(s)
| | - Ali Niazi
- Institute of Biotechnology, Shiraz University, Shiraz, Iran,Address for correspondence Institute of Biotechnology, University of Shiraz, Shiraz, IR Iran, Tel: +98-7112272805, E-mail:
| | | | - Ali Hortamani
- Institute of Biotechnology, Shiraz University, Shiraz, Iran
| | - Payam Setoodeh
- Institute of Biotechnology, Shiraz University, Shiraz, Iran,School of Petroleum and Chemical Engineering, Shiraz University, Shiraz, Iran
| |
Collapse
|
40
|
Majka G, Śpiewak K, Kurpiewska K, Heczko P, Stochel G, Strus M, Brindell M. A high-throughput method for the quantification of iron saturation in lactoferrin preparations. Anal Bioanal Chem 2013; 405:5191-200. [PMID: 23604471 PMCID: PMC3656221 DOI: 10.1007/s00216-013-6943-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 03/14/2013] [Accepted: 03/25/2013] [Indexed: 11/29/2022]
Abstract
Lactoferrin is considered as a part of the innate immune system that plays a crucial role in preventing bacterial growth, mostly via an iron sequestration mechanism. Recent data show that bovine lactoferrin prevents late-onset sepsis in preterm very low birth weight neonates by serving as an iron chelator for some bacterial strains; thus, it is very important to control the iron saturation level during diet supplementation. An accurate estimation of lactoferrin iron saturation is essential not only because of its clinical applications but also for a wide range of biochemical experiments. A comprehensive method for the quantification of iron saturation in lactoferrin preparations was developed to obtain a calibration curve enabling the determination of iron saturation levels relying exclusively on the defined ratio of absorbances at 280 and 466 nm (A280/466). To achieve this goal, selected techniques such as spectrophotometry, ELISA, and ICP-MS were combined. The ability to obtain samples of lactoferrin with determination of its iron content in a simple and fast way has been proven to be very useful. Furthermore, a similar approach could easily be implemented to facilitate the determination of iron saturation level for other metalloproteins in which metal binding results in the appearance of a distinct band in the visible part of the spectrum.
Collapse
Affiliation(s)
- Grzegorz Majka
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University, Ingardena 3, 30-060 Krakow, Poland
| | | | | | | | | | | | | |
Collapse
|
41
|
Zumoffen CM, Gil R, Caille AM, Morente C, Munuce MJ, Ghersevich SA. A protein isolated from human oviductal tissue in vitro secretion, identified as human lactoferrin, interacts with spermatozoa and oocytes and modulates gamete interaction. Hum Reprod 2013; 28:1297-308. [DOI: 10.1093/humrep/det016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
42
|
Antimicrobial lactoferrin peptides: the hidden players in the protective function of a multifunctional protein. INTERNATIONAL JOURNAL OF PEPTIDES 2013; 2013:390230. [PMID: 23554820 PMCID: PMC3608178 DOI: 10.1155/2013/390230] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 01/22/2013] [Indexed: 11/17/2022]
Abstract
Lactoferrin is a multifunctional, iron-binding glycoprotein which displays a wide array of modes of action to execute its primary antimicrobial function. It contains various antimicrobial peptides which are released upon its hydrolysis by proteases. These peptides display a similarity with the antimicrobial cationic peptides found in nature. In the current scenario of increasing resistance to antibiotics, there is a need for the discovery of novel antimicrobial drugs. In this context, the structural and functional perspectives on some of the antimicrobial peptides found in N-lobe of lactoferrin have been reviewed. This paper provides the comparison of lactoferrin peptides with other antimicrobial peptides found in nature as well as interspecies comparison of the structural properties of these peptides within the native lactoferrin.
Collapse
|
43
|
Zimecki M, Artym J, Kocięba M, Kaleta-Kuratewicz K, Kruzel ML. Lactoferrin restrains allergen-induced pleurisy in mice. Inflamm Res 2012; 61:1247-55. [PMID: 22810368 PMCID: PMC3472062 DOI: 10.1007/s00011-012-0522-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Revised: 06/06/2012] [Accepted: 06/27/2012] [Indexed: 11/28/2022] Open
Abstract
Objectives The aim of this study was to assess the utility of lactoferrin (LF), a natural immunomodulator, to restrain allergen-induced pleurisy in mice. Material and subjects BALB/c female mice, 8- to 10-week old, weighing 24 g on average, were used. Treatment Mice were immunized intraperitoneally with 50 μg of ovalbumin (OVA) and the pleurisy was elicited 14 days later by intrapleural injection of 12.5 μg of OVA. LF was given 24 and 3 h before elicitation of the allergic reaction. Methods The cytokine levels in the pleural exudates were measured by immunoassays. The blood and pleural exudates smears were stained with Giemsa and May-Grünwald reagents and reviewed histologically. Lung sections were stained with eosin and hematoxylin for histological evaluation. Results Lactoferrin significantly decreased manifestation of pleurisy induced by OVA in a sensitized mouse model. In particular, the percentages of eosinophils in blood and pleural exudates were strongly diminished. The histological analysis of lungs revealed that LF diminished the development of pathological lesions, such as pulmonary edema, diffuse alveolar hemorrhage and hemosiderosis, which were found in the lungs after injection of the eliciting dose of OVA. LF also decreased the level of IL-5 secreted into the pleural fluid. Conclusions This is a first demonstration that LF significantly decreases antigen-specific pleurisy in a sensitized mouse model.
Collapse
Affiliation(s)
- Michał Zimecki
- Institute of Immunology and Experimental Therapy, Wrocław, Poland
| | | | | | | | | |
Collapse
|
44
|
Curran CS, Bertics PJ. Lactoferrin regulates an axis involving CD11b and CD49d integrins and the chemokines MIP-1α and MCP-1 in GM-CSF-treated human primary eosinophils. J Interferon Cytokine Res 2012; 32:450-61. [PMID: 22731992 DOI: 10.1089/jir.2011.0111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Eosinophils are multifunctional immune cells that contribute to innate and adaptive immune/repair responses. Lactoferrin (LF) is an iron-binding protein indicated to alter cell adhesion and immune function by receptor-mediated interactions or by participating in redox mechanisms. The eosinophil adhesion molecules, αMβ2 and α4β1, are differentially expressed following exposure to the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) and various redox agents. We hypothesized that LF can alter the function and production of proteins involved in adhesion/migration. Utilizing eosinophil peroxidase activity or fluorescent labeling adhesion assays, LF reduced GM-CSF-induced eosinophil adhesion in the presence of fibronectin or vascular adhesion molecule-1 compared with GM-CSF treatment alone. Flow cytometric analysis of eosinophil αM (CD11b) and α4 (CD49d) integrins revealed that cotreatments (24 h) with LF plus GM-CSF induced a significant increase in CD11b compared with control and GM-CSF treatments but a significant decrease in CD49d compared with control and GM-CSF treatments. These changes in CD11b and CD49d levels were significantly correlated with the increased production of chemokines (macrophage inflammatory Protein-1α, monocyte chemotactic protein-1) and an identified increase in S100A9 production. Thus, LF release at sites of inflammation may alter eosinophil recruitment/activation and possibly the progression of diseases such as cancer and asthma where significant eosinophil influx has been described.
Collapse
Affiliation(s)
- Colleen S Curran
- Department of Molecular Biology, University of Wisconsin, Madison, Wisconsin 53706, USA.
| | | |
Collapse
|
45
|
Mariller C, Hardivillé S, Hoedt E, Huvent I, Pina-Canseco S, Pierce A. Delta-lactoferrin, an intracellular lactoferrin isoform that acts as a transcription factor1This article is part of a Special Issue entitled Lactoferrin and has undergone the Journal's usual peer review process. Biochem Cell Biol 2012; 90:307-19. [DOI: 10.1139/o11-070] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Delta-lactoferrin (ΔLf) is a transcription factor of which the expression is downregulated in cancer. It is a healthy tissue marker and a high expression level of its transcripts was correlated with a good prognosis in breast cancer. ΔLf results from alternative promoter usage of the hLf gene leading to the production of 2 isoforms with alternative N-termini: lactoferrin, which is secreted, and ΔLf, its nucleocytoplasmic counterpart. ΔLf possesses antiproliferative properties and induces cell cycle arrest. It is an efficient transcription factor interacting in vivo via a ΔLf response element found in the Skp1, Bax, DcpS, and SelH promoters. Since ΔLf possesses different target genes, modifications in its activity or concentration may have crucial effects on cell homeostasis. Posttranslational modifications modulate ΔLf transcription factor activity. Our earlier investigations showed that O-GlcNAcylation negatively regulates ΔLf transcriptional activity, whilst inhibiting its ubiquitination and increasing its half-life. On the other hand, phosphorylation potentiates ΔLf transcriptional activity. Recently, we showed that ΔLf is also modified by SUMOylation. Therefore, cooperation and (or) competition among SUMOylation, ubiquitination, phosphorylation, and O-GlcNAcylation may contribute to the establishment of a fine regulation of ΔLf transcriptional activity depending on the type of target gene and cellular homeostasis.
Collapse
Affiliation(s)
- Christophe Mariller
- Unité de Glycobiologie Structurale et Fonctionnelle, UMR 8576 CNRS-Université des Sciences et Technologies de Lille, IFR 148, 59655 Villeneuve d’Ascq, France
| | - Stephan Hardivillé
- Unité de Glycobiologie Structurale et Fonctionnelle, UMR 8576 CNRS-Université des Sciences et Technologies de Lille, IFR 148, 59655 Villeneuve d’Ascq, France
| | - Esthelle Hoedt
- Unité de Glycobiologie Structurale et Fonctionnelle, UMR 8576 CNRS-Université des Sciences et Technologies de Lille, IFR 148, 59655 Villeneuve d’Ascq, France
| | - Isabelle Huvent
- Unité de Glycobiologie Structurale et Fonctionnelle, UMR 8576 CNRS-Université des Sciences et Technologies de Lille, IFR 148, 59655 Villeneuve d’Ascq, France
| | - Socorro Pina-Canseco
- Unité de Glycobiologie Structurale et Fonctionnelle, UMR 8576 CNRS-Université des Sciences et Technologies de Lille, IFR 148, 59655 Villeneuve d’Ascq, France
| | - Annick Pierce
- Unité de Glycobiologie Structurale et Fonctionnelle, UMR 8576 CNRS-Université des Sciences et Technologies de Lille, IFR 148, 59655 Villeneuve d’Ascq, France
| |
Collapse
|
46
|
Baker HM, Baker EN. A structural perspective on lactoferrin function1This article is part of a Special Issue entitled Lactoferrin and has undergone the Journal's usual peer review process. Biochem Cell Biol 2012; 90:320-8. [PMID: 22292559 DOI: 10.1139/o11-071] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The 3-D structure of human lactoferrin was first solved in atomic detail in 1987. Since that time, a variety of proven and postulated activities have been added to the original annotation of lactoferrin as an iron-binding protein. Structural studies have also expanded to include iron-bound and iron-free (apo) forms, mutants, and the lactoferrins of different species. In this review, we take the current information on both structure and function and show that the 3-D structure provides a useful framework for understanding some activities and also points to productive research directions that could help elucidate other reported functions. Some functions relate to iron binding where the role of lactoferrin is to scavenge and retain iron across a wide pH range. We specifically focus on functions that depend on the surface structure of the molecule, identifying features that may determine the many other protective properties of this multifunctional protein.
Collapse
Affiliation(s)
- Heather M. Baker
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Edward N. Baker
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| |
Collapse
|
47
|
Videm V, Dahl H, Wålberg LE, Wiseth R. Functional polymorphisms in the LTF gene and risk of coronary artery stenosis. Hum Immunol 2012; 73:554-9. [PMID: 22406253 DOI: 10.1016/j.humimm.2012.02.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 02/21/2012] [Accepted: 02/22/2012] [Indexed: 10/28/2022]
Abstract
Plasma lactoferrin concentrations are increased in patients with coronary artery stenosis. We investigated the effects of LTF gene polymorphisms in 305 healthy blood donors and their associations with coronary artery stenosis in 236 patients admitted for coronary angiography. Lactoferrin concentrations were determined by enzyme immunoassay. Genotyping was performed by polymerase chain reaction and DNA sequencing of LTF exons 2 and 4. In the blood donors, the deletion variant of rs10662431 and the G allele of rs1126478 were associated with higher plasma lactoferrin concentrations. The G allele of rs1126478 was more frequent in patients with significant coronary artery stenosis (p = 0.018, p value limit for significance by permutation = 0.030). The association remained significant in logistic regression with adjustment for clinical risk factors (odds ratio 2.485 [95% confidence interval 1.116-5.536], p = 0.026), but was weakened upon the inclusion of plasma lactoferrin (odds ratio 2.295 [0.949-5.550], p = 0.064). Current evidence indicates that rs1126478 affects the antibacterial effect of lactoferrin and that lactoferrin is involved in lipid metabolism. The relationships among lactoferrin genotypes, lactoferrin concentrations, and clinical factors on the risk for atherosclerosis are not fully understood, but the G allele of rs1126478 seems to have a detrimental effect in a European population.
Collapse
Affiliation(s)
- Vibeke Videm
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway.
| | | | | | | |
Collapse
|
48
|
Legrand D. Lactoferrin, a key molecule in immune and inflammatory processes. Biochem Cell Biol 2011; 90:252-68. [PMID: 22136726 DOI: 10.1139/o11-056] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lactoferrin (Lf) belongs to the family of antimicrobial molecules that constitute the principal defense line of nonvertebrate organisms. In human immunity, their roles are considerably extended, and actually exceed mere direct antimicrobial properties. As a result, Lf is involved in both innate and adaptive immunities where its modulating effects not only help the host fight against microbes but also protect the host against harmful effects of inflammation. Such beneficial effects have been noticed in studies using dietary Lf, without the experimenters always explaining the exact modes of action of Lf. Effects on mucosal and systemic immunities are indeed often observed, which make the roles of Lf tricky to decipher. It is now known that the immunomodulatory properties of Lf are due to its ability to interact with numerous cellular and molecular targets. At the cellular level, Lf modulates the migration, maturation, and functions of immune cells. At the molecular level, in addition to iron binding, interactions of Lf with a plethora of compounds, either soluble or cell-surface molecules, account for its modulatory properties. This paper reviews our current understanding of the mechanisms that explain the regulatory properties of Lf in immune and inflammatory processes.
Collapse
Affiliation(s)
- Dominique Legrand
- UMR 8576 CNRS / Université des Sciences et Technologies de Lille, Unité de Glycobiologie Structurale et Fonctionnelle, IFR 147, F-59650 Villeneuve d'Ascq, France.
| |
Collapse
|
49
|
Lactoferrin-lipopolysaccharide (LPS) binding as key to antibacterial and antiendotoxic effects. Int Immunopharmacol 2011; 12:1-9. [PMID: 22101278 DOI: 10.1016/j.intimp.2011.11.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2011] [Revised: 09/01/2011] [Accepted: 11/03/2011] [Indexed: 01/03/2023]
Abstract
Lactoferrin (Lf), a multifunctional protein of the innate immune response, seems to act as a permeabilizing agent of Gram negative bacteria, apparently due to its interaction with enterobacterial lipopolysaccharide (LPS) on the bacterial surface. In both human and bovine Lf, a six residue sequence lying in an 18-loop region of the lactoferricin domain is key to Lf-LPS binding. There is much evidence that, by its action on LPS, Lf destabilizes the bacterial membrane and therefore increases bacterial permeability. By itself, Lf is not an effective antibacterial agent, but it permits the penetration of the bacterial membrane by some antibacterial substances whose hydrophobicity otherwise limits their efficacy. Additionally, Lf neutralizes free LPS by keeping the latter from forming complexes that activate TLR-4 signaling pathways. Such pathways, when over-activated, lead to the abundant production of pro-inflammatory mediators such as tumor necrosis factor (TNF) with fatal consequences to the host. The effect of Lf in reducing inflammation and destabilizing Gram negative bacteria has clinical implications in the control of sepsis, multiple organ dysfunction and bacterial invasion.
Collapse
|
50
|
Jiang R, Lönnerdal B. Apo- and holo-lactoferrin stimulate proliferation of mouse crypt cells but through different cellular signaling pathways. Int J Biochem Cell Biol 2011; 44:91-100. [PMID: 22009034 DOI: 10.1016/j.biocel.2011.10.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 10/03/2011] [Accepted: 10/04/2011] [Indexed: 11/26/2022]
Abstract
Lactoferrin (Lf), an iron-binding glycoprotein in human milk, plays an important role in intestinal epithelial cell proliferation, but the underlying molecular mechanisms remain unknown. Mitogen-activated protein kinase (MAPK)/extracellular signal-regulated protein kinases (ERK) and the phosphoinositide 3-kinase (PI3K) cascades are chief pathways controlling cell proliferation. We hypothesized that Lf stimulates proliferation of intestinal epithelial cells by activating ERK and PI3K signaling cascades and that lactoferrin receptor (LfR) is involved in these processes. Confocal microscopy showed that LfR is expressed and localized at the plasma membrane of mouse crypt cells isolated from 7 to 10 days old pups, specifically binding both iron-free Lf (apo-Lf) and iron-saturated Lf (holo-Lf). Using the BrdU assay, we observed that apo-Lf had a stronger stimulatory effect than holo-Lf on proliferation of crypt cells. Both function-blocking antibody against LfR and MEK inhibitor (U0126) resulted in inhibitory effects on apo-Lf-induced cell proliferation, whereas PI3K inhibitor (LY294002) significantly decreased both apo- and holo-Lf-induced proliferation. Although both apo- and holo-Lf up-regulated transcription of cyclin D1, an effector of ERK1/2 and PI3K/Akt signaling cascades, only apo-Lf initiated ERK1/2 signaling and both apo- and holo-Lf were capable of activating the PI3K/Akt signaling pathway. Function-blocking antibody against LfR and LfR siRNA inhibited apo-Lf induced activation of the ERK1/2 signaling pathway, suggesting that LfR is involved in apo-Lf induced activation of the ERK1/2, but not the PI3K signaling pathway. In conclusion, apo-Lf and holo-Lf stimulate intestinal proliferation but through different signaling pathways.
Collapse
Affiliation(s)
- Rulan Jiang
- Department of Nutrition, University of California, Davis, CA 95616, United States
| | | |
Collapse
|