1
|
Guzmán-Mejía F, Molotla-Torres DE, Godínez-Victoria M, Valdes-Hilarios X, Sánchez-Miranda E, Oros-Pantoja R, Drago-Serrano ME. Looking Inside of the Intestinal Permeability Regulation by Protein-Derivatives from Bovine Milk. Mol Nutr Food Res 2024:e2400384. [PMID: 39530631 DOI: 10.1002/mnfr.202400384] [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: 05/21/2024] [Revised: 09/19/2024] [Indexed: 11/16/2024]
Abstract
The prime function of the epithelium is to regulate the intestinal permeability; the latter is a quantitative parameter that refers to the measurement of the rate of passage of solutes through the epithelial monolayer. Function of epithelial monolayer depends on the expression of protein complexes known as tight junction proteins; whose function and expression can be disrupted under conditions of inflammation including irritable bowel disease (IBD), intestinal infections, and high-fat diets, among others. This manuscript is focused to outline the effects of bovine milk protein derivatives on the intestinal permeability addressed mostly in animal models in which the intestinal barrier is disrupted. At present, the properties of bovine milk protein derivatives on intestinal permeability have been scarcely documented in humans, but evidence raised from clinical trials provides promising findings of potential application of colostrum to control of the intestinal permeability in critically ill patients, users of non-steroid anti-inflammatory drugs, like athletes and militia members.
Collapse
Affiliation(s)
- Fabiola Guzmán-Mejía
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Calzada del Hueso No. 1100, Ciudad de México CP, 04960, México
| | - Daniel Efrain Molotla-Torres
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Calzada del Hueso No. 1100, Ciudad de México CP, 04960, México
| | - Marycarmen Godínez-Victoria
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Ciudad de México CP, 11340, México
| | - Ximena Valdes-Hilarios
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Calzada del Hueso No. 1100, Ciudad de México CP, 04960, México
| | - Elizabeth Sánchez-Miranda
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Calzada del Hueso No. 1100, Ciudad de México CP, 04960, México
| | - Rigoberto Oros-Pantoja
- Laboratorio de Neuroinmunoendocrinología, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, 50180, Mexico
| | - Maria Elisa Drago-Serrano
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Calzada del Hueso No. 1100, Ciudad de México CP, 04960, México
| |
Collapse
|
2
|
Häussler S, Ghaffari MH, Seibt K, Sadri H, Alaedin M, Huber K, Frahm J, Dänicke S, Sauerwein H. Blood and liver telomere length, mitochondrial DNA copy number, and hepatic gene expression of mitochondrial dynamics in mid-lactation cows supplemented with l-carnitine under systemic inflammation. J Dairy Sci 2023; 106:9822-9842. [PMID: 37641324 DOI: 10.3168/jds.2023-23556] [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: 03/31/2023] [Accepted: 06/21/2023] [Indexed: 08/31/2023]
Abstract
The current study was conducted to examine the effect of l-carnitine (LC) supplementation on telomere length and mitochondrial DNA copy number (mtDNAcn) per cell in mid-lactation cows challenged by lipopolysaccharide (LPS) in blood and liver. The mRNA abundance of 31 genes related to inflammation, oxidative stress, and the corresponding stress response mechanisms, the mitochondrial quality control and the protein import system, as well as the phosphatidylinositol 3-kinase/protein kinase B pathway, were assessed using microfluidics integrated fluidic circuit chips (96.96 dynamic arrays). In addition to comparing the responses in cows with or without LC, our objectives were to characterize the oxidative and inflammatory status by assessing the circulating concentration of lactoferrin (Lf), haptoglobin (Hp), fibrinogen, derivates of reactive oxygen metabolites (dROM), and arylesterase activity (AEA), and to extend the measurement of Lf and Hp to milk. Pluriparous Holstein cows were assigned to either a control group (CON, n = 26) or an LC-supplemented group (CAR; 25 g LC/cow per day; d 42 ante partum to d 126 postpartum (PP), n = 27). On d 111 PP, each cow was injected intravenously with LPS (Escherichia coli O111:B4, 0.5 µg/kg). The mRNA abundance was examined in liver biopsies of d -11 and +1 relative to LPS administration. Plasma and milk samples were frequently collected before and after the challenge. After LPS administration, circulating plasma fibrinogen and serum dROM concentrations increased, whereas AEA decreased. Moreover, serum P4 initially increased by 3 h after LPS administration and declined thereafter irrespective of grouping. The Lf concentrations increased in both groups after LPS administration, with the CAR group showing greater concentrations in serum and milk than the CON group. After LPS administration, telomere length in blood increased, whereas mtDNAcn per cell decreased; however, both remained unaffected in liver. For mitochondrial protein import genes, the hepatic mRNA abundance of the translocase of the mitochondrial inner membrane (TIM)-17B was increased in CAR cows. Moreover, TIM23 increased in both groups after LPS administration. Regarding the mRNA abundance of genes related to stress response mechanisms, 7 out of 14 genes showed group × time interactions, indicating a (local) protective effect due to the dietary LC supplementation against oxidative stress in mid-lactating dairy cows. For mtDNAcn and telomere length, the effects of the LPS-induced inflammation were more pronounced than the dietary supplementation of LC. Dietary LC supplementation affected the response to LPS primarily by altering mitochondrial dynamics. Regarding mRNA abundance of genes related to the mitochondrial protein import system, the inner mitochondrial membrane translocase (TIM complex) seemed to be more sensitive to dietary LC than the outer mitochondrial membrane translocase (TOM complex).
Collapse
Affiliation(s)
- S Häussler
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - M H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany.
| | - K Seibt
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - H Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, 516616471 Tabriz, Iran
| | - M Alaedin
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - K Huber
- Institute of Animal Science, Functional Anatomy of Livestock, University of Hohenheim, 70599 Stuttgart, Germany
| | - J Frahm
- Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, 38116 Braunschweig, Germany
| | - S Dänicke
- Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, 38116 Braunschweig, Germany
| | - H Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
3
|
Fux AC, Casonato Melo C, Michelini S, Swartzwelter BJ, Neusch A, Italiani P, Himly M. Heterogeneity of Lipopolysaccharide as Source of Variability in Bioassays and LPS-Binding Proteins as Remedy. Int J Mol Sci 2023; 24:ijms24098395. [PMID: 37176105 PMCID: PMC10179214 DOI: 10.3390/ijms24098395] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Lipopolysaccharide (LPS), also referred to as endotoxin, is the major component of Gram-negative bacteria's outer cell wall. It is one of the main types of pathogen-associated molecular patterns (PAMPs) that are known to elicit severe immune reactions in the event of a pathogen trespassing the epithelial barrier and reaching the bloodstream. Associated symptoms include fever and septic shock, which in severe cases, might even lead to death. Thus, the detection of LPS in medical devices and injectable pharmaceuticals is of utmost importance. However, the term LPS does not describe one single molecule but a diverse class of molecules sharing one common feature: their characteristic chemical structure. Each bacterial species has its own pool of LPS molecules varying in their chemical composition and enabling the aggregation into different supramolecular structures upon release from the bacterial cell wall. As this heterogeneity has consequences for bioassays, we aim to examine the great variability of LPS molecules and their potential to form various supramolecular structures. Furthermore, we describe current LPS quantification methods and the LPS-dependent inflammatory pathway and show how LPS heterogeneity can affect them. With the intent of overcoming these challenges and moving towards a universal approach for targeting LPS, we review current studies concerning LPS-specific binders. Finally, we give perspectives for LPS research and the use of LPS-binding molecules.
Collapse
Affiliation(s)
- Alexandra C Fux
- Division of Allergy & Immunology, Department of Biosciences & Medical Biology, Paris Lodron University of Salzburg (PLUS), Hellbrunnerstraße 34, 5020 Salzburg, Austria
- Chemical Biology Department, R&D Reagents, Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
| | - Cristiane Casonato Melo
- Division of Allergy & Immunology, Department of Biosciences & Medical Biology, Paris Lodron University of Salzburg (PLUS), Hellbrunnerstraße 34, 5020 Salzburg, Austria
- Chemical Biology Department, R&D Reagents, Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Straße 68, 51429 Bergisch Gladbach, Germany
| | - Sara Michelini
- Biotechnical Faculty, Department of Biology, University of Ljubljana, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Benjamin J Swartzwelter
- Department of Microbiology, Immunology, and Pathology, 1601 Campus Delivery, Colorado State University, Fort Collins, CO 80523, USA
| | - Andreas Neusch
- Experimental Medical Physics, Heinrich-Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Paola Italiani
- Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche (CNR), Via P. Castellino 111, 80131 Naples, Italy
- Stazione Zoologica Anton Dohrn (SZN), Villa Comunale, 80121 Naples, Italy
| | - Martin Himly
- Division of Allergy & Immunology, Department of Biosciences & Medical Biology, Paris Lodron University of Salzburg (PLUS), Hellbrunnerstraße 34, 5020 Salzburg, Austria
| |
Collapse
|
4
|
Stater EP, Morcos G, Isaac E, Ogirala A, Hsu HT, Longo VA, Grimm J. Translatable Drug-Loaded Iron Oxide Nanophore Sensitizes Murine Melanoma Tumors to Monoclonal Antibody Immunotherapy. ACS NANO 2023; 17:6178-6192. [PMID: 36971591 PMCID: PMC10324163 DOI: 10.1021/acsnano.2c05800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Macrophages comprise a significant portion of the immune cell compartment within tumors and are known contributors to tumor pathology; however, cancer immunotherapies targeting these cells are not clinically available. The iron oxide nanoparticle, ferumoxytol (FH), may be utilized as a nanophore for drug delivery to tumor-associated macrophages. We have demonstrated that a vaccine adjuvant, monophosphoryl lipid A (MPLA), can be stably captured within the carbohydrate shell of ferumoxytol without chemical modification of either the drug or the nanophore. This drug-nanoparticle combination (FH-MPLA) activated macrophages to an antitumorigenic phenotype at clinically relevant concentrations. In the immunotherapy-resistant B16-F10 model of murine melanoma, FH-MPLA treatment induced tumor necrosis and regression in combination with agonistic α-CD40 monoclonal antibody therapy. FH-MPLA, composed of clinically approved nanoparticle and drug payload, represents a potential cancer immunotherapy with translational relevance. FH-MPLA may be useful as an adjunctive therapy to existing antibody-based cancer immunotherapies which target only lymphocytic cells, reshaping the tumor immune environment.
Collapse
Affiliation(s)
- Evan P. Stater
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York NY, 10065
- Department of Pharmacology, Weill Cornell Medical College, 1300 York Ave, New York NY, 10065
| | | | - Elizabeth Isaac
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York NY, 10065
- Department of Pharmacology, Weill Cornell Medical College, 1300 York Ave, New York NY, 10065
| | - Anuja Ogirala
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York NY, 10065
| | - Hsiao-Ting Hsu
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York NY, 10065
| | - Valerie A. Longo
- Small Animal Imaging Core Facility, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York NY, 10065
| | - Jan Grimm
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York NY, 10065
- Department of Pharmacology, Weill Cornell Medical College, 1300 York Ave, New York NY, 10065
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York NY, 10065
| |
Collapse
|
5
|
Cheng Y, Chen J, Zou S, Huang L, Li G. The mechanism underlying the remodeling effect of lactoferrin on midpalatal sutures during maxillary expansion and relapse in rats. Am J Orthod Dentofacial Orthop 2023; 163:e137-e151. [PMID: 37012109 DOI: 10.1016/j.ajodo.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 03/01/2023] [Accepted: 03/01/2023] [Indexed: 04/03/2023]
Abstract
INTRODUCTION The remodeling effects of intragastric administration and intramaxillary injection of lactoferrin (LF) on midpalatal sutures (MPS) during maxillary expansion and relapse in rats were studied to explore the underlying bone remodeling mechanism. METHODS Using a rat model of maxillary expansion and relapse, rats were treated with LF by intragastric administration (1 g·kg-1·d-1) or intramaxillary injection (5 mg·25 μl-1·d-1). The effects of LF on the osteogenic and osteoclast activities of MPS were observed by microcomputed tomography, histologic staining, and immunohistochemical staining, and the expressions of key factors in the extracellular regulated protein kinase 1/2 (ERK1/2) pathway and osteoprotegerin (OPG)-receptor activator of nuclear factor-KB ligand (RANKL)-receptor activator of nuclear factor-KB (RANK) axis were detected. RESULTS Compared with the group with maxillary expansion alone, osteogenic activity was relatively enhanced, whereas osteoclast activity was relatively weakened in the groups administered LF, and the phosphorylated-ERK1/2: ERK1/2 and OPG: RANKL expression ratios increased significantly. The difference was more significant in the group administered LF intramaxillary. CONCLUSIONS Administration of LF promoted osteogenic activity at MPS and inhibited osteoclast activity during maxillary expansion and relapse in rats, which may have occurred through regulation of the ERK1/2 pathway and the OPG-RANKL-RANK axis. The efficiency of intramaxillary LF injection was greater than that of intragastric LF administration.
Collapse
|
6
|
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
|
7
|
Levy E, Marcil V, Tagharist Ép Baumel S, Dahan N, Delvin E, Spahis S. Lactoferrin, Osteopontin and Lactoferrin–Osteopontin Complex: A Critical Look on Their Role in Perinatal Period and Cardiometabolic Disorders. Nutrients 2023; 15:nu15061394. [PMID: 36986124 PMCID: PMC10052990 DOI: 10.3390/nu15061394] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/02/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
Abstract
Milk-derived bioactive proteins have increasingly gained attention and consideration throughout the world due to their high-quality amino acids and multiple health-promoting attributes. Apparently, being at the forefront of functional foods, these bioactive proteins are also suggested as potential alternatives for the management of various complex diseases. In this review, we will focus on lactoferrin (LF) and osteopontin (OPN), two multifunctional dairy proteins, as well as to their naturally occurring bioactive LF–OPN complex. While describing their wide variety of physiological, biochemical, and nutritional functionalities, we will emphasize their specific roles in the perinatal period. Afterwards, we will evaluate their ability to control oxidative stress, inflammation, gut mucosal barrier, and intestinal microbiota in link with cardiometabolic disorders (CMD) (obesity, insulin resistance, dyslipidemia, and hypertension) and associated complications (diabetes and atherosclerosis). This review will not only attempt to highlight the mechanisms of action, but it will critically discuss the potential therapeutic applications of the underlined bioactive proteins in CMD.
Collapse
Affiliation(s)
- Emile Levy
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Department of Nutrition, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
| | - Valérie Marcil
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Department of Nutrition, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
| | - Sarah Tagharist Ép Baumel
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Department of Nutrition, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
| | - Noam Dahan
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
| | - Edgard Delvin
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Biochemistry &Molecular Medicine, Faculty of Medicine, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
| | - Schohraya Spahis
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Biochemistry &Molecular Medicine, Faculty of Medicine, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
- Correspondence: ; Tel.: +1-(514)-345-4832
| |
Collapse
|
8
|
Almehdar HA, Abd El-Baky N, Mattar EH, Albiheyri R, Bamagoos A, Aljaddawi A, Uversky VN, Redwan EM. Exploring the mechanisms by which camel lactoferrin can kill Salmonella enterica serovar typhimurium and Shigella sonnei. PeerJ 2023; 11:e14809. [PMID: 36743956 PMCID: PMC9893911 DOI: 10.7717/peerj.14809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/05/2023] [Indexed: 01/31/2023] Open
Abstract
There is a continuously increasing pressure associated with the appearance of Salmonella enterica Serovar typhimurium (S. typhimurium) and Shigella sonnei (S. sonnei) that have developed pathogenic multiple antibiotic resistance and the cost of cure and control of these enterobacteriaceae infections increases annually. The current report for first time demonstrated the distinguished antimicrobial action of camel lactoferrin (cLf) obtained from the milk of different clans of camel in Saudi Arabia against S. typhimurium and S. sonnei. These cLf subtypes showed comparable antimicrobial potential when tested against the two bacterial strains but were superior to either bovine (bLf) or human lactoferrin (hLf). The synergism between lactoferrins and antibiotics concerning their antibacterial efficacies against the two bacterial strains was evident. Exploring mechanisms by which camel lactoferrin can kill S. typhimurium and S. sonnei revealed that cLf affects bacterial protein profile. Besides, it interacts with bacterial lipopolysaccharides (LPS) and numerous membrane proteins of S. typhimurium and S. sonnei, with each bacterial strain possessing distinctive binding membrane proteins for lactoferrin. Furthermore, as evidenced by electron microscopy analysis, cLf induces extracellular and intracellular morphological changes in the test bacterial strains when used alone or in combination treatment with antibiotics. Lactoferrin and antibiotics combination strongly disrupts the integrity of the bacterial cells and their membranes. Therefore, cLf can kill S. typhimurium and S. sonnei by four different mechanisms, such as iron chelation, affecting some bacterial proteins, binding to bacterial LPS and membrane proteins, and impairing the integrity of the bacterial cells and their membranes.
Collapse
Affiliation(s)
- Hussein A. Almehdar
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nawal Abd El-Baky
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, Alexandria, Egypt
| | - Ehab H. Mattar
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Raed Albiheyri
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Atif Bamagoos
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdullah Aljaddawi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Vladimir N. Uversky
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States of America
| | - Elrashdy M. Redwan
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia,Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, Alexandria, Egypt
| |
Collapse
|
9
|
Einerhand AWC, van Loo-Bouwman CA, Weiss GA, Wang C, Ba G, Fan Q, He B, Smit G. Can Lactoferrin, a Natural Mammalian Milk Protein, Assist in the Battle against COVID-19? Nutrients 2022; 14:nu14245274. [PMID: 36558432 PMCID: PMC9782828 DOI: 10.3390/nu14245274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Notwithstanding mass vaccination against specific SARS-CoV-2 variants, there is still a demand for complementary nutritional intervention strategies to fight COVID-19. The bovine milk protein lactoferrin (LF) has attracted interest of nutraceutical, food and dairy industries for its numerous properties-ranging from anti-viral and anti-microbial to immunological-making it a potential functional ingredient in a wide variety of food applications to maintain health. Importantly, bovine LF was found to exert anti-viral activities against several types of viruses, including certain SARS-CoV-2 variants. LF's potential effect on COVID-19 patients has seen a rapid increase of in vitro and in vivo studies published, resulting in a model on how LF might play a role during different phases of SARS-CoV-2 infection. Aim of this narrative review is two-fold: (1) to highlight the most relevant findings concerning LF's anti-viral, anti-microbial, iron-binding, immunomodulatory, microbiota-modulatory and intestinal barrier properties that support health of the two most affected organs in COVID-19 patients (lungs and gut), and (2) to explore the possible underlying mechanisms governing its mode of action. Thanks to its potential effects on health, bovine LF can be considered a good candidate for nutritional interventions counteracting SARS-CoV-2 infection and related COVID-19 pathogenesis.
Collapse
Affiliation(s)
| | | | | | - Caiyun Wang
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot 010110, China
| | - Genna Ba
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010110, China
| | - Qicheng Fan
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010110, China
| | - Baoping He
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010110, China
| | - Gerrit Smit
- Yili Innovation Center Europe, 6708 WH Wageningen, The Netherlands
| |
Collapse
|
10
|
Casciaro B, Loffredo MR, Cappiello F, O’Sullivan N, Tortora C, Manzer R, Karmakar S, Haskell A, Hasan SK, Mangoni ML. KDEON WK-11: A short antipseudomonal peptide with promising potential. Front Chem 2022; 10:1000765. [PMID: 36465859 PMCID: PMC9713011 DOI: 10.3389/fchem.2022.1000765] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/01/2022] [Indexed: 08/27/2023] Open
Abstract
The plight of antimicrobial resistance continues to limit the availability of antibiotic treatment effective in combating resistant bacterial infections. Despite efforts made to rectify this issue and minimise its effects on both patients and the wider community, progress in this area remains minimal. Here, we de-novo designed a peptide named KDEON WK-11, building on previous work establishing effective residues and structures active in distinguished antimicrobial peptides such as lactoferrin. We assessed its antimicrobial activity against an array of bacterial strains and identified its most potent effect, against Pseudomonas aeruginosa with an MIC value of 3.12 μM, lower than its counterparts developed with similar residues and chain lengths. We then determined its anti-biofilm properties, potential mechanism of action and in vitro cytotoxicity. We identified that KDEON WK-11 had a broad range of antimicrobial activity and specific capabilities to fight Pseudomonas aeruginosa with low in vitro cytotoxicity and promising potential to express anti-lipopolysaccharide qualities, which could be exploited to expand its properties into an anti-sepsis agent.
Collapse
Affiliation(s)
- Bruno Casciaro
- Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Maria Rosa Loffredo
- Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Floriana Cappiello
- Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Niamh O’Sullivan
- Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Carola Tortora
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018–2022”, Sapienza University of Rome, Rome, Italy
| | - Rizwan Manzer
- Iuventis Technologies Inc. (DBA Immunotrex Biologics), Lowell, MA, United States
| | - Sougata Karmakar
- Iuventis Technologies Inc. (DBA Immunotrex Biologics), Lowell, MA, United States
| | - Alan Haskell
- Iuventis Technologies Inc. (DBA Immunotrex Biologics), Lowell, MA, United States
| | - Syed K. Hasan
- Iuventis Technologies Inc. (DBA Immunotrex Biologics), Lowell, MA, United States
| | - Maria Luisa Mangoni
- Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
11
|
Xu Y, Wang Y, He J, Zhu W. Antibacterial properties of lactoferrin: A bibliometric analysis from 2000 to early 2022. Front Microbiol 2022; 13:947102. [PMID: 36060777 PMCID: PMC9428516 DOI: 10.3389/fmicb.2022.947102] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundHere, a bibliometric and knowledge map analysis are used to analyze the research hot spots and development trends regarding the antibacterial effect of lactoferrin (LF). By looking for research hot spots and new topics, we provide new clues and research directions for future research.MethodsArticles and reviews regarding the antibacterial effect of LF were retrieved and from the Web of Science Core Collection (WoSCC) on 25 June 2022. CiteSpace and VOSviewer were used to conduct the bibliometric and knowledge map analysis.ResultsIn total, 8,292 authors at 2,151 institutions from 86 countries published 1,923 articles in 770 academic journals. The United States was the leader regarding research on the antibacterial effects of LF, while the Netherlands was a pioneer in conducting research in this field. The University of California system contributed the most publications. Bolscher JGM published most articles, while Wayne Bellamy had most cocitations. However, there was insufficient cooperation among the various institutions and authors. BioMetals published most LF-antibacterial activity-related articles, whereas Infection and Immunity was most commonly cocited journal. The most influential research hot spots about the antibacterial effect of LF focused on antimicrobial peptides, casein, human milk, expression, and Escherichia coli-related research. The latest hot spots and research frontier included COVID-19, antibiofilm activity, and immune defense.ConclusionsLF is a multifunctional protein with a broad spectrum of antimicrobial activities. The related field of antibacterial properties of LF will remain a research hot spot in future.
Collapse
Affiliation(s)
- Yunling Xu
- Department of Basic Medical, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Yuji Wang
- Department of Basic Medical, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Jiaolong He
- Department of Intensive Care, First Affiliated Hospital of Jishou University, Jishou, China
- *Correspondence: Jiaolong He
| | - Wanping Zhu
- Department of Basic Medical, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
- Wanping Zhu
| |
Collapse
|
12
|
Effect of bovine lactoferrin on recurrent urinary tract infections: in vitro and in vivo evidences. Biometals 2022; 36:491-507. [PMID: 35768747 DOI: 10.1007/s10534-022-00409-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/09/2022] [Indexed: 11/02/2022]
Abstract
Uropathogenic Escherichia coli (UPEC) strains are the primary cause of urinary tract infections (UTIs). UPEC strains are able to invade, multiply and persisting in host cells. Therefore, UPEC strains are associated to recurrent UTIs requiring long-term antibiotic therapy. However, this therapy is suboptimal due to the increase of multidrug-resistant UPEC. The use of non-antibiotic treatments for managing UTIs is required. Among these, bovine lactoferrin (bLf), a multifunctional cationic glycoprotein, could be a promising tool because inhibits the entry into the host cells of several intracellular bacteria. Here, we demonstrate that 100 μg/ml bLf hinders the invasion of 2.0 ± 0.5 × 104 CFU/ml E. coli CFT073, prototype of UPEC, infecting 2.0 ± 0.5 × 105 cells/ml urinary bladder T24 epithelial cells. The highest protection (100%) is due to the bLf binding with host surface components even if an additional binding to bacterial surface components cannot be excluded. Of note, in the absence of bLf, UPEC survives and multiplies, while bLf significantly decreases bacterial intracellular survival. After these encouraging results, an observational survey on thirty-three patients affected by recurrent cystitis was performed. The treatment consisted in the oral administration of bLf alone or in combination with antibiotics and/or probiotics. After the observation period, a marked reduction of cystitis episodes was observed (p < 0.001) in all patients compared to the episodes occurred during the 6 months preceding the bLf-treatment. Twenty-nine patients did not report cystitis episodes (87.9%) whereas the remaining four (12.1%) experienced only one episode, indicating that bLf could be a worthwhile and safe treatment in counteracting recurrent cystitis.
Collapse
|
13
|
Tajbakhsh A, Yousefi F, Abedi SM, Rezaee M, Savardashtaki A, Teng Y, Sahebkar A. The cross-talk between soluble "Find me" and "Keep out" signals as an initial step in regulating efferocytosis. J Cell Physiol 2022; 237:3113-3126. [PMID: 35578547 DOI: 10.1002/jcp.30770] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/16/2022] [Accepted: 04/21/2022] [Indexed: 12/20/2022]
Abstract
The rapid clearance of apoptotic cells (ACs), known as efferocytosis, prompts the inhibition of inflammatory responses and autoimmunity and maintains homeostatic cell turnover by controlling the release of intracellular contents. The fast clearance of ACs requires professional and nonprofessional phagocytic cells that can accurately and promptly recognize ACs and migrate towards them. Cells undergoing apoptosis alarm their presence by releasing special soluble chemotactic factors, such as lactoferrin, that act as "Find me," "Keep out," or "Stay away" signals to recruit phagocytic cells, such as macrophages or prevent granulocyte migration. Efferocytosis effectively serves to prevent damage-associated molecular pattern release and secondary necrosis and inhibit inflammation/autoimmunity at the very first step. Since less attention has been given to the cross-talk and balance of "Find me" and "Keep out" signals released from ACs in efferocytosis, we set out to investigate the current knowledge of the roles of "Find me" and "Keep out" signals in the efferocytosis process.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Yousefi
- Department of Biological Sciences, Faculty of Genetics, Tarbiat Modares University, Tehran, Iran
| | - Seyedeh M Abedi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Rezaee
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Amir Savardashtaki
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Western Australia, Australia.,Depatment of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
14
|
Tsugami Y, Harada R, Nii T, Suzuki N, Isobe N. Effects of frequent teat stimulation on antimicrobial component production in mammary glands of lactating goats. Vet Immunol Immunopathol 2022; 249:110431. [DOI: 10.1016/j.vetimm.2022.110431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
|
15
|
Varela-Fernández R, García-Otero X, Díaz-Tomé V, Regueiro U, López-López M, González-Barcia M, Isabel Lema M, Otero-Espinar FJ. Mucoadhesive PLGA Nanospheres and Nanocapsules for Lactoferrin Controlled Ocular Delivery. Pharmaceutics 2022; 14:pharmaceutics14040799. [PMID: 35456633 PMCID: PMC9029159 DOI: 10.3390/pharmaceutics14040799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/25/2022] [Accepted: 04/01/2022] [Indexed: 12/13/2022] Open
Abstract
Background: the present work describes the preparation, characterization and optimization of eight types of PLGA-based nanosystems (nanospheres and nanocapsules) as innovative mucoadhesive drug delivery systems of lactoferrin, in order to achieve a preclinical consistent base as an alternative pharmacological treatment to different ocular syndromes and diseases. Methods: All different nanoparticles were prepared via two modified nanoprecipitation techniques, using a three-component mixture of drug/polymer/surfactant (Lf/PLGA/Poloxamer), as a way to overcome the inherent limitations of conventional PLGA NPs. These modified polymeric nanocarriers, intended for topical ophthalmic administration, were subjected to in vitro characterization, surface modification and in vitro and in vivo assessments. Results: An appropriate size range, uniform size distribution and negative ζ potential values were obtained for all types of formulations. Lactoferrin could be effectively included into all types of nanoparticles with appropriate encapsulation efficiency and loading capacity values. A greater, extended, and controlled delivery of Lf from the polymeric matrix was observed through the in vitro release studies. No instability or cytotoxicity was proved for all the formulations by means of organotypic models. Additionally, mucoadhesive in vitro and in vivo experiments show a significant increase in the residence time of the nanoparticles in the eye surface. Conclusions: all types of prepared PLGA nanoparticles might be a potential alternative for the topical ophthalmic administration of lactoferrin.
Collapse
Affiliation(s)
- Rubén Varela-Fernández
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela (USC), Campus Vida, 15782 Santiago de Compostela, Spain; (R.V.-F.); (X.G.-O.); (V.D.-T.)
- Clinical Neurosciences Group, University Clinical Hospital, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (U.R.); (M.L.-L.)
| | - Xurxo García-Otero
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela (USC), Campus Vida, 15782 Santiago de Compostela, Spain; (R.V.-F.); (X.G.-O.); (V.D.-T.)
- Molecular Imaging Group, University Clinical Hospital, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Victoria Díaz-Tomé
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela (USC), Campus Vida, 15782 Santiago de Compostela, Spain; (R.V.-F.); (X.G.-O.); (V.D.-T.)
| | - Uxía Regueiro
- Clinical Neurosciences Group, University Clinical Hospital, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (U.R.); (M.L.-L.)
| | - Maite López-López
- Clinical Neurosciences Group, University Clinical Hospital, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (U.R.); (M.L.-L.)
| | - Miguel González-Barcia
- Clinical Pharmacology Group, University Clinical Hospital, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain;
| | - María Isabel Lema
- Department of Surgery and Medical-Surgical Specialties, Ophthalmology Area, University of Santiago de Compostela (USC), Campus Vida, 15706 Santiago de Compostela, Spain
- Correspondence: (M.I.L.); (F.J.O.-E.)
| | - Francisco Javier Otero-Espinar
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela (USC), Campus Vida, 15782 Santiago de Compostela, Spain; (R.V.-F.); (X.G.-O.); (V.D.-T.)
- Institute of Materials Imatus, University of Santiago de Compostela (USC), Campus Vida, 15782 Santiago de Compostela, Spain
- Paraquasil Group, University Clinical Hospital, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Correspondence: (M.I.L.); (F.J.O.-E.)
| |
Collapse
|
16
|
Varela-Fernández R, García-Otero X, Díaz-Tomé V, Regueiro U, López-López M, González-Barcia M, Isabel Lema M, Javier Otero-Espinar F. Lactoferrin-loaded nanostructured lipid carriers (NLCs) as a new formulation for optimized ocular drug delivery. Eur J Pharm Biopharm 2022; 172:144-156. [DOI: 10.1016/j.ejpb.2022.02.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/02/2022] [Accepted: 02/14/2022] [Indexed: 11/17/2022]
|
17
|
Zeinali LI, Giuliano S, Lakshminrusimha S, Underwood MA. Intestinal Dysbiosis in the Infant and the Future of Lacto-Engineering to Shape the Developing Intestinal Microbiome. Clin Ther 2021; 44:193-214.e1. [PMID: 34922744 DOI: 10.1016/j.clinthera.2021.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 11/06/2021] [Accepted: 11/12/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE The goal of this study was to review the role of human milk in shaping the infant intestinal microbiota and the potential of human milk bioactive molecules to reverse trends of increasing intestinal dysbiosis and dysbiosis-associated diseases. METHODS This narrative review was based on recent and historic literature. FINDINGS Human milk immunoglobulins, oligosaccharides, lactoferrin, lysozyme, milk fat globule membranes, and bile salt-stimulating lipase are complex multifunctional bioactive molecules that, among other important functions, shape the composition of the infant intestinal microbiota. IMPLICATIONS The co-evolution of human milk components and human milk-consuming commensal anaerobes many thousands of years ago resulted in a stable low-diversity infant microbiota. Over the past century, the introduction of antibiotics and modern hygiene practices plus changes in the care of newborns have led to significant alterations in the intestinal microbiota, with associated increases in risk of dysbiosis-associated disease. A better understanding of mechanisms by which human milk shapes the intestinal microbiota of the infant during a vulnerable period of development of the immune system is needed to alter the current trajectory and decrease intestinal dysbiosis and associated diseases.
Collapse
Affiliation(s)
- Lida I Zeinali
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA
| | | | | | - Mark A Underwood
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
18
|
Ramírez-Rico G, Martinez-Castillo M, Avalos-Gómez C, de la Garza M. Bovine apo-lactoferrin affects the secretion of proteases in Mannheimia haemolytica A2. Access Microbiol 2021; 3:000269. [PMID: 34816089 PMCID: PMC8604176 DOI: 10.1099/acmi.0.000269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 08/23/2021] [Indexed: 02/05/2023] Open
Abstract
Mannheimia haemolytica serotype A2 is the main bacterial causative agent of ovine mannheimiosis, a disease that leads to substantial economic losses for livestock farmers. Several virulence factors allow M. haemolytica to colonize the lungs and establish infection. Virulence factors can be directly secreted into the environment by bacteria but are also released through outer membrane vesicles (OMVs). In addition, due to the abuse of antibiotics in the treatment of this disease, multidrug-resistant bacterial strains of M. haemolytica have emerged. One therapeutic alternative to antibiotics or an adjuvant to be used in combination with antibiotics could be lactoferrin (Lf), a multifunctional cationic glycoprotein of the mammalian innate immune system to which no bacterial resistance has been reported. The aim of this work was to determine the effect of bovine iron-free Lf (apo-BLf) on the production and secretion of proteases into culture supernatant (CS) and on their release in OMVs. Zymography assays showed that addition of sub-MIC concentrations of apo-BLf to M. haemolytica cultures inhibited protease secretion without affecting culture growth. Biochemical characterization revealed that these proteases were mainly cysteine- and metalloproteases. The secretion of a 100 kDa metalloprotease was inhibited by sub-MIC concentrations of apo-BLf since this protease was present in the cytoplasm and OMVs but not in CS proteins, as corroborated by Western blotting. On the other hand, proteases produced by M. haemolytica caused cleavage of apo-BLf. However, when Lf is cleaved, peptides known as lactoferricins, which are more bactericidal than natural Lf, can be produced. M. haemolytica A2 protease-mediated degradation of host tissue proteins could be an important virulence factor during the infectious process of pneumonia in ovines. The mechanism of M. haemolytica protease secretion could be inhibited by treatment with apo-BLf in animals.
Collapse
Affiliation(s)
- Gerardo Ramírez-Rico
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, CdMx 07360, Mexico.,Present address: Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Km 2.5 Carretera Cuautitlán-Teoloyucan, Cuautitlán Izcalli, 54714, Mexico
| | - Moises Martinez-Castillo
- Laboratorio de Hígado, Páncreas y Motilidad (HIPAM). Unidad de Investigación en Medicina Experimental, Facultad de Medicina, UNAM. Hospital General de México, Col Doctores, CdMx 06726, Mexico
| | - Christian Avalos-Gómez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, CdMx 07360, Mexico
| | - Mireya de la Garza
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, CdMx 07360, Mexico
| |
Collapse
|
19
|
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
|
20
|
Pan S, Weng H, Hu G, Wang S, Zhao T, Yao X, Liao L, Zhu X, Ge Y. Lactoferrin may inhibit the development of cancer via its immunostimulatory and immunomodulatory activities (Review). Int J Oncol 2021; 59:85. [PMID: 34533200 DOI: 10.3892/ijo.2021.5265] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/11/2021] [Indexed: 11/05/2022] Open
Abstract
Lactoferrin (Lf) is secreted by ectodermal tissue and has a structure similar to that of transferrin. Although Lf seems to be multifunctional, its main function is related to the natural defense system of mammals. The present review aims to highlight the major actions of Lf, including the regulation of cell growth, the inhibition of toxic compound formation, the removal of harmful free radicals and its important role in immune response regulation. Moreover, Lf has antibacterial, antiviral, antioxidant, anticancer and anti‑inflammatory activities. In addition, the use of Lf for functionalization of drug nanocarriers, with emphasis on tumor‑targeted drug delivery, is illustrated. Such effects serve as an important theoretical basis for its future development and application. In neurodegenerative diseases and the brains of elderly people, Lf expression is markedly upregulated. Lf may exert an anti‑inflammatory effect by inhibiting the formation of hydroxyl free radicals. Through its antioxidant properties, Lf can prevent DNA damage, thereby preventing tumor formation in the central nervous system. In addition, Lf specifically activates the p53 tumor suppressor gene.
Collapse
Affiliation(s)
- Sian Pan
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| | - Huiting Weng
- Department of Clinical Nursing, The Second Xiangya Hospital of Central South University, Changsha, Hunan 430011, P.R. China
| | - Guohong Hu
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| | - Shiwen Wang
- Department of Histology and Embryology, School of Basic Medicine Sciences, Xinjiang Medical University, Urumqi, Xinjiang 830017, P.R. China
| | - Tian Zhao
- Department of Histology and Embryology, School of Basic Medicine Sciences, Xinjiang Medical University, Urumqi, Xinjiang 830017, P.R. China
| | - Xueping Yao
- Department of Histology and Embryology, School of Basic Medicine Sciences, Xinjiang Medical University, Urumqi, Xinjiang 830017, P.R. China
| | - Libin Liao
- Department of Histology and Embryology, School of Basic Medicine Sciences, Xinjiang Medical University, Urumqi, Xinjiang 830017, P.R. China
| | - Xiaopeng Zhu
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| | - Yanshan Ge
- The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, The Third Affiliated Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
21
|
Lu J, Haley KP, Francis JD, Guevara MA, Doster RS, Craft KM, Moore RE, Chambers SA, Delgado AG, Piazuelo MB, Damo SM, Townsend SD, Gaddy JA. The Innate Immune Glycoprotein Lactoferrin Represses the Helicobacter pylori cag Type IV Secretion System. Chembiochem 2021; 22:2783-2790. [PMID: 34169626 PMCID: PMC8560179 DOI: 10.1002/cbic.202100249] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/23/2021] [Indexed: 01/06/2023]
Abstract
Chronic infection with Helicobacter pylori increases risk of gastric diseases including gastric cancer. Despite development of a robust immune response, H. pylori persists in the gastric niche. Progression of gastric inflammation to serious disease outcomes is associated with infection with H. pylori strains which encode the cag Type IV Secretion System (cag T4SS). The cag T4SS is responsible for translocating the oncogenic protein CagA into host cells and inducing pro-inflammatory and carcinogenic signaling cascades. Our previous work demonstrated that nutrient iron modulates the activity of the T4SS and biogenesis of T4SS pili. In response to H. pylori infection, the host produces a variety of antimicrobial molecules, including the iron-binding glycoprotein, lactoferrin. Our work shows that apo-lactoferrin exerts antimicrobial activity against H. pylori under iron-limited conditions, while holo-lactoferrin enhances bacterial growth. Culturing H. pylori in the presence of holo-lactoferrin prior to co-culture with gastric epithelial cells, results in repression of the cag T4SS activity. Concomitantly, a decrease in biogenesis of cag T4SS pili at the host-pathogen interface was observed under these culture conditions by high-resolution electron microscopy analyses. Taken together, these results indicate that acquisition of alternate sources of nutrient iron plays a role in regulating the pro-inflammatory activity of a bacterial secretion system and present novel therapeutic targets for the treatment of H. pylori-related disease.
Collapse
Affiliation(s)
- Jacky Lu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, U.S.A
| | - Kathryn P. Haley
- Department of Biology, Grand Valley State University, Allendale, Michigan, 49401, U.S.A
| | - Jamisha D. Francis
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, U.S.A
| | - Miriam A. Guevara
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, U.S.A
| | - Ryan S. Doster
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, U.S.A
| | - Kelly M. Craft
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, 37235, U.S.A
| | - Rebecca E. Moore
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, 37235, U.S.A
| | - Schuyler A. Chambers
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, 37235, U.S.A
| | - Alberto G. Delgado
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, U.S.A
| | - Maria Blanca Piazuelo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, U.S.A
| | - Steven M. Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, Tennessee, 37208, U.S.A
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, 37232, U.S.A
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, 37232, U.S.A
| | - Steven D. Townsend
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, 37235, U.S.A
| | - Jennifer A. Gaddy
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, U.S.A
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, U.S.A
- Tennessee Valley Healthcare Systems, Department of Veterans Affairs, Nashville, Tennessee, 37212, U.S.A
| |
Collapse
|
22
|
Cao Y, Zhang Y, Qiu F. Low endotoxin recovery and its impact on endotoxin detection. Biopolymers 2021; 112:e23470. [PMID: 34407207 DOI: 10.1002/bip.23470] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 01/05/2023]
Abstract
Endotoxin exists on the outer membrane of Gram-negative bacteria and poses risks to human health by triggering a series of immune responses. Therefore, its accurate detection is essential. The Limulus amoebocyte lysate (LAL) test is the most pharmacopeia-recognized and popular technique for endotoxin detection. Despite its wide industry adoption, the low endotoxin recovery (LER) phenomenon can compromise the LAL test's reliability. This review summarizes the possible reasons attributing to the LER phenomenon from three different perspectives: the endotoxin standards used in hold time study, protein active pharmaceutical ingredients, and excipients. Potential mechanisms and strategies to mitigate the LER phenomenon are also discussed as presented by different research groups.
Collapse
Affiliation(s)
- Yuan Cao
- Department of Process Development, BeiGene Guangzhou Biologics Manufacturing Co., Ltd., Guangzhou, China
| | - Yujie Zhang
- Department of Process Development, BeiGene Guangzhou Biologics Manufacturing Co., Ltd., Guangzhou, China
| | - Frank Qiu
- Department of Process Development, BeiGene Guangzhou Biologics Manufacturing Co., Ltd., Guangzhou, China
| |
Collapse
|
23
|
Singh J, Vijayan V, Ahmedi S, Pant P, Manzoor N, Singh TP, Sharma P, Sharma S. Lactosmart: A Novel Therapeutic Molecule for Antimicrobial Defense. Front Microbiol 2021; 12:672589. [PMID: 34220755 PMCID: PMC8250155 DOI: 10.3389/fmicb.2021.672589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/30/2021] [Indexed: 11/13/2022] Open
Abstract
The problem of antibiotic resistance has prompted researchers around the globe to search for new antimicrobial agents. Antimicrobial proteins and peptides are naturally secreted by almost all the living organisms to fight infections and can be safer alternatives to chemical antibiotics. Lactoferrin (LF) is a known antimicrobial protein present in all body secretions. In this study, LF was digested by trypsin, and the resulting hydrolysates were studied with respect to their antimicrobial properties. Among the hydrolysates, a 21-kDa basic fragment of LF (termed lactosmart) showed promise as a new potent antimicrobial agent. The antimicrobial studies were performed on various microorganisms including Shigella flexneri, Pseudomonas aeruginosa, Staphylococcus aureus, and Escherichia coli as well as fungal pathogens such as Candida albicans, Candida tropicalis, and Candida glabrata. In addition, the lipopolysaccharide (LPS)-binding properties of lactosmart were studied using surface plasmon resonance technique in vitro, along with docking of LPS and molecular dynamics (MD) simulation studies. The results showed that lactosmart had better inhibitory effects against pathogenic microorganisms compared to LF. The results of docking and MD simulation studies further validated the tighter binding of LPS to lactosmart compared to LF. The two LPS-binding sites have been characterized structurally in detail. Through these studies, it has been demonstrated that in native LF, only one LPS-binding site remains exposed due to its location being on the surface of the molecule. However, due to the generation of the lactosmart molecule, the second LPS-binding site gets exposed too. Since LPS is an essential and conserved part of the bacterial cell wall, the pro-inflammatory response in the human body caused by LPS can be targeted using the newly identified lactosmart. These findings highlight the immense potential of lactosmart in comparison to native LF in antimicrobial defense. We propose that lactosmart can be further developed as an antibacterial, antifungal, and antibiofilm agent.
Collapse
Affiliation(s)
- Jiya Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Viswanathan Vijayan
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Saiema Ahmedi
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Pradeep Pant
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi, India
| | - Nikhat Manzoor
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Tej P. Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Sujata Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
24
|
Goulding DA, Vidal K, Bovetto L, O'Regan J, O'Brien NM, O'Mahony JA. The impact of thermal processing on the simulated infant gastrointestinal digestion, bactericidal and anti-inflammatory activity of bovine lactoferrin - An in vitro study. Food Chem 2021; 362:130142. [PMID: 34087706 DOI: 10.1016/j.foodchem.2021.130142] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/13/2021] [Accepted: 05/16/2021] [Indexed: 02/07/2023]
Abstract
Lactoferrin (LF) is a multifunctional glycoprotein which, when thermally processed, undergoes significant physicochemical changes. The link between such changes and the bioactivity of LF is not well characterised and requires much research. In this work, bovine LF solutions (1%, w/v, protein, pH 7) were thermally processed using high temperature short time conditions (72, 80, 85 or 95 °C with 15 s holding times). Following this, it was shown that LF and heat induced LF aggregates were largely resistant to simulated infant gastric, but not intestinal, digestion. Also, the efficacy of LF bactericidal activity, and inhibition of lipopolysaccharide-induced NF-κB activation were negatively impacted by thermal processing. This study confirmed that the efficacy of LF bio-functionalities was affected by the extent of heat-induced changes in protein structure whereby processing conditions of least severity (i.e. pasteurisation) had the least impact on bioactivity.
Collapse
Affiliation(s)
- David A Goulding
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | - Karine Vidal
- Nestlé Research, Nestlé Institute of Health Sciences, Vers-chez-les-Blanc, CH-1000 Lausanne 26, Switzerland
| | - Lionel Bovetto
- Nestlé Research, Nestlé Institute of Material Sciences, Vers-chez-les-Blanc, CH-1000 Lausanne 26, Switzerland
| | - Jonathan O'Regan
- Nestlé Development Centre Nutrition, Askeaton, Co. Limerick, Ireland
| | - Nora M O'Brien
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | - James A O'Mahony
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland.
| |
Collapse
|
25
|
Yan M, Rodgers M, Harbhajanka A, Gilmore H. Lactotransferrin-Related Breast Amyloidosis: Report of a First Case. Int J Surg Pathol 2021; 30:50-54. [PMID: 33939557 DOI: 10.1177/10668969211016053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Breast amyloidosis is a rare condition which is mostly associated with hematological disorders or hereditary genetic disorders. Imaging findings of breast amyloidosis can mimic malignancy, which often leads to biopsy or excision of the lesion. Here, we presented a case of localized lactotransferrin-related breast amyloidosis in an elderly female patient. Histologic examination revealed extensive involvement of breast lobules by amorphous amyloid materials, with attenuation of lobular structures and prominent calcifications. Positive immunostains for myoepithelial cells helped to exclude the possibility of invasive carcinoma. The patient had no hematologic malignancy besides immunoglobulin G lambda monoclonal gammopathy of undetermined significance. Mass spectrometry of the breast amyloid identified lactotransferrin and no immunoglobulin or its light chain. On follow-up, the patient showed no recurrence of the breast lesion after local excision nor showed other systematic comorbidities, indicating the benign nature of the lesion. This first report of lactotransferrin-related amyloidosis may represent a special type of localized breast amyloidosis that has no correlation with systematic disorders.
Collapse
Affiliation(s)
- Mingfei Yan
- 24575University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Mark Rodgers
- 24575University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | | | - Hannah Gilmore
- 24575University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| |
Collapse
|
26
|
Sánchez C, Franco L, Regal P, Lamas A, Cepeda A, Fente C. Breast Milk: A Source of Functional Compounds with Potential Application in Nutrition and Therapy. Nutrients 2021; 13:1026. [PMID: 33810073 PMCID: PMC8005182 DOI: 10.3390/nu13031026] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/09/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022] Open
Abstract
Breast milk is an unbeatable food that covers all the nutritional requirements of an infant in its different stages of growth up to six months after birth. In addition, breastfeeding benefits both maternal and child health. Increasing knowledge has been acquired regarding the composition of breast milk. Epidemiological studies and epigenetics allow us to understand the possible lifelong effects of breastfeeding. In this review we have compiled some of the components with clear functional activity that are present in human milk and the processes through which they promote infant development and maturation as well as modulate immunity. Milk fat globule membrane, proteins, oligosaccharides, growth factors, milk exosomes, or microorganisms are functional components to use in infant formulas, any other food products, nutritional supplements, nutraceuticals, or even for the development of new clinical therapies. The clinical evaluation of these compounds and their commercial exploitation are limited by the difficulty of isolating and producing them on an adequate scale. In this work we focus on the compounds produced using milk components from other species such as bovine, transgenic cattle capable of expressing components of human breast milk or microbial culture engineering.
Collapse
Affiliation(s)
- Cristina Sánchez
- Pharmacy Faculty, San Pablo-CEU University, 28003 Madrid, Spain;
| | - Luis Franco
- Medicine Faculty, Santiago de Compostela University, 15782 Santiago de Compostela, Spain;
| | - Patricia Regal
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Alexandre Lamas
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Alberto Cepeda
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| | - Cristina Fente
- Department of Analytical Chemistry, Nutrition and Bromatology, Santiago de Compostela University, 27002 Lugo, Spain; (P.R.); (A.L.); (A.C.)
| |
Collapse
|
27
|
Gill K, Horsley H, Swamy S, Khasriya R, Malone-Lee J. A prospective observational study of urinary cytokines and inflammatory response in patients with Overactive Bladder Syndrome. BMC Urol 2021; 21:39. [PMID: 33740940 PMCID: PMC7980577 DOI: 10.1186/s12894-021-00809-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/03/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Contemporary studies have discredited the methods used to exclude urinary tract infection (UTI) when treating overactive bladder (OAB). Thus we must revisit the OAB phenotype to check that UTI has not been overlooked. AIMS To examine the differences in urinary cytokines IL6 and lactoferrin in OAB patients compared to controls, with references to microscopy of urine and enhanced quantitative urine culture. METHODS A blinded, prospective cohort study with normal controls using six repeated measures, achieved two-monthly, over 12 months. RESULTS The differences between patients and controls in urine IL6 (F = 49.0, p < .001) and lactoferrin (F = 228.5, p < .001) were significant and of a magnitude to have clinical implications. These differences were for lactoferrin correlated to symptoms (9.3, p = .003); for both to pyuria (IL6 F = 66.2, p < .001, Lactoferrin F = 73.9, p < .001); and for IL6 microbial abundance (F = 5.1, p = .024). The pathological markers had been missed by urinary dipsticks and routine MSU culture. CONCLUSION The OAB phenotype may encompass patients with UTI that is being overlooked because of the failure of standard screening methods.
Collapse
Affiliation(s)
- Kiren Gill
- Women’s Health, Whittington Health NHS Trust, Magdala Avenue, London, N19 5FN UK
| | - Harry Horsley
- Bladder Infection and Immunity Group (BIIG), Department of Renal Medicine, Division of Medicine, University College London, Royal Free Hospital Campus, Rowland Hill Street, London, NW3 2PF UK
| | - Sheela Swamy
- Women’s Health, St Thomas’ Hospital, Westminster Bridge Road, London, SE1 7EH UK
| | - Rajvinder Khasriya
- Women’s Health, Whittington Health NHS Trust, Magdala Avenue, London, N19 5FN UK
| | - James Malone-Lee
- Bladder Infection and Immunity Group (BIIG), Department of Renal Medicine, Division of Medicine, University College London, 10 Harley Street, London, W1G 9PF UK
| |
Collapse
|
28
|
Cutone A, Ianiro G, Lepanto MS, Rosa L, Valenti P, Bonaccorsi di Patti MC, Musci G. Lactoferrin in the Prevention and Treatment of Intestinal Inflammatory Pathologies Associated with Colorectal Cancer Development. Cancers (Basel) 2020; 12:E3806. [PMID: 33348646 PMCID: PMC7766217 DOI: 10.3390/cancers12123806] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022] Open
Abstract
The connection between inflammation and cancer is well-established and supported by genetic, pharmacological and epidemiological data. The inflammatory bowel diseases (IBDs), including Crohn's disease and ulcerative colitis, have been described as important promoters for colorectal cancer development. Risk factors include environmental and food-borne mutagens, dysbalance of intestinal microbiome composition and chronic intestinal inflammation, with loss of intestinal epithelial barrier and enhanced cell proliferation rate. Therapies aimed at shutting down mucosal inflammatory response represent the foundation for IBDs treatment. However, when applied for long periods, they can alter the immune system and promote microbiome dysbiosis and carcinogenesis. Therefore, it is imperative to find new safe substances acting as both potent anti-inflammatory and anti-pathogen agents. Lactoferrin (Lf), an iron-binding glycoprotein essential in innate immunity, is generally recognized as safe and used as food supplement due to its multifunctionality. Lf possesses a wide range of immunomodulatory and anti-inflammatory properties against different aseptic and septic inflammatory pathologies, including IBDs. Moreover, Lf exerts anti-adhesive, anti-invasive and anti-survival activities against several microbial pathogens that colonize intestinal mucosa of IBDs patients. This review focuses on those activities of Lf potentially useful for the prevention/treatment of intestinal inflammatory pathologies associated with colorectal cancer development.
Collapse
Affiliation(s)
- Antimo Cutone
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (A.C.); (G.I.)
| | - Giusi Ianiro
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (A.C.); (G.I.)
| | - Maria Stefania Lepanto
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (P.V.)
| | - Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (P.V.)
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (P.V.)
| | | | - Giovanni Musci
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (A.C.); (G.I.)
| |
Collapse
|
29
|
Zarzosa-Moreno D, Avalos-Gómez C, Ramírez-Texcalco LS, Torres-López E, Ramírez-Mondragón R, Hernández-Ramírez JO, Serrano-Luna J, de la Garza M. Lactoferrin and Its Derived Peptides: An Alternative for Combating Virulence Mechanisms Developed by Pathogens. Molecules 2020; 25:E5763. [PMID: 33302377 PMCID: PMC7762604 DOI: 10.3390/molecules25245763] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 12/16/2022] Open
Abstract
Due to the emergence of multidrug-resistant pathogens, it is necessary to develop options to fight infections caused by these agents. Lactoferrin (Lf) is a cationic nonheme multifunctional glycoprotein of the innate immune system of mammals that provides numerous benefits. Lf is bacteriostatic and/or bactericidal, can stimulate cell proliferation and differentiation, facilitate iron absorption, improve neural development and cognition, promote bone growth, prevent cancer and exert anti-inflammatory and immunoregulatory effects. Lactoferrin is present in colostrum and milk and is also produced by the secondary granules of polymorphonuclear leukocytes, which store this glycoprotein and release it at sites of infection. Lf is also present in many fluids and exocrine secretions, on the surfaces of the digestive, respiratory and reproductive systems that are commonly exposed to pathogens. Apo-Lf (an iron-free molecule) can be microbiostatic due to its ability to capture ferric iron, blocking the availability of host iron to pathogens. However, apo-Lf is mostly microbicidal via its interaction with the microbial surface, causing membrane damage and altering its permeability function. Lf can inhibit viral entry by binding to cell receptors or viral particles. Lf is also able to counter different important mechanisms evolved by microbial pathogens to infect and invade the host, such as adherence, colonization, invasion, production of biofilms and production of virulence factors such as proteases and toxins. Lf can also cause mitochondrial and caspase-dependent regulated cell death and apoptosis-like in pathogenic yeasts. All of these mechanisms are important targets for treatment with Lf. Holo-Lf (the iron-saturated molecule) can contain up to two ferric ions and can also be microbicidal against some pathogens. On the other hand, lactoferricins (Lfcins) are peptides derived from the N-terminus of Lf that are produced by proteolysis with pepsin under acidic conditions, and they cause similar effects on pathogens to those caused by the parental Lf. Synthetic analog peptides comprising the N-terminus Lf region similarly exhibit potent antimicrobial properties. Importantly, there are no reported pathogens that are resistant to Lf and Lfcins; in addition, Lf and Lfcins have shown a synergistic effect with antimicrobial and antiviral drugs. Due to the Lf properties being microbiostatic, microbicidal, anti-inflammatory and an immune modulator, it represents an excellent natural alternative either alone or as adjuvant in the combat to antibiotic multidrug-resistant bacteria and other pathogens. This review aimed to evaluate the data that appeared in the literature about the effects of Lf and its derived peptides on pathogenic bacteria, protozoa, fungi and viruses and how Lf and Lfcins inhibit the mechanisms developed by these pathogens to cause disease.
Collapse
Affiliation(s)
- Daniela Zarzosa-Moreno
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Zacatenco 07360, CdMx, Mexico; (D.Z.-M.); (C.A.-G.); (J.S.-L.)
| | - Christian Avalos-Gómez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Zacatenco 07360, CdMx, Mexico; (D.Z.-M.); (C.A.-G.); (J.S.-L.)
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Coyoacán 04510, CdMx, Mexico
| | - Luisa Sofía Ramírez-Texcalco
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán Izcalli 54714, Estado de México, Mexico; (L.S.R.-T.); (E.T.-L.); (R.R.-M.); (J.O.H.-R.)
| | - Erick Torres-López
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán Izcalli 54714, Estado de México, Mexico; (L.S.R.-T.); (E.T.-L.); (R.R.-M.); (J.O.H.-R.)
| | - Ricardo Ramírez-Mondragón
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán Izcalli 54714, Estado de México, Mexico; (L.S.R.-T.); (E.T.-L.); (R.R.-M.); (J.O.H.-R.)
| | - Juan Omar Hernández-Ramírez
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán Izcalli 54714, Estado de México, Mexico; (L.S.R.-T.); (E.T.-L.); (R.R.-M.); (J.O.H.-R.)
| | - Jesús Serrano-Luna
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Zacatenco 07360, CdMx, Mexico; (D.Z.-M.); (C.A.-G.); (J.S.-L.)
| | - Mireya de la Garza
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Zacatenco 07360, CdMx, Mexico; (D.Z.-M.); (C.A.-G.); (J.S.-L.)
| |
Collapse
|
30
|
Papademas P, Kamilari E, Aspri M, Anagnostopoulos DA, Mousikos P, Kamilaris A, Tsaltas D. Investigation of donkey milk bacterial diversity by 16S rDNA high-throughput sequencing on a Cyprus donkey farm. J Dairy Sci 2020; 104:167-178. [PMID: 33162091 DOI: 10.3168/jds.2020-19242] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022]
Abstract
The interest in milk originating from donkeys is growing worldwide due to its claimed functional and nutritional properties, especially for sensitive population groups, such as infants with cow milk protein allergy. The current study aimed to assess the microbiological quality of donkey milk produced in a donkey farm in Cyprus using culture-based and high-throughput sequencing techniques. The culture-based microbiological analysis showed very low microbial counts, whereas important food-borne pathogens were not detected in any sample. In addition, high-throughput sequencing was applied to characterize the bacterial communities of donkey milk samples. Donkey milk mostly composed of gram-negative Proteobacteria, including Sphingomonas, Pseudomonas, Mesorhizobium, and Acinetobacter; lactic acid bacteria, including Lactobacillus and Streptococcus; the endospores forming Clostridium; and the environmental genera Flavobacterium and Ralstonia, detected in lower relative abundances. The results of the study support existing findings that donkey milk contains mostly gram-negative bacteria. Moreover, it raises questions regarding the contribution of (1) antimicrobial agents (i.e., lysozyme, peptides) in shaping the microbial communities and (2) bacterial microbiota to the functional value of donkey milk.
Collapse
Affiliation(s)
- P Papademas
- Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University of Technology, Limassol 3036, Cyprus.
| | - E Kamilari
- Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University of Technology, Limassol 3036, Cyprus
| | - M Aspri
- Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University of Technology, Limassol 3036, Cyprus
| | - D A Anagnostopoulos
- Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University of Technology, Limassol 3036, Cyprus
| | - P Mousikos
- Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University of Technology, Limassol 3036, Cyprus
| | - A Kamilaris
- Faculty of Electrical Engineering, Mathematics and Computer Science (EEMCS), University of Twente, Enschede, 7522 NB, the Netherlands; Research Centre on Interactive Media, Smart Systems and Emerging Technologies-RISE, Nicosia 1066, Cyprus
| | - D Tsaltas
- Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University of Technology, Limassol 3036, Cyprus.
| |
Collapse
|
31
|
Ueda K, Shimizu M, Ohashi A, Murata D, Suzuki T, Kobayashi N, Baba J, Takeuchi T, Shiga Y, Nakamura M, Kagaya S, Sato A. Albumin fusion at the N-terminus or C-terminus of human lactoferrin leads to improved pharmacokinetics and anti-proliferative effects on cancer cell lines. Eur J Pharm Sci 2020; 155:105551. [PMID: 32946958 DOI: 10.1016/j.ejps.2020.105551] [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] [Received: 05/25/2020] [Revised: 08/23/2020] [Accepted: 09/12/2020] [Indexed: 12/18/2022]
Abstract
Human lactoferrin (hLF), a soluble factor of the innate immune system, exhibits various biological functions and therefore has potential as a therapeutic protein. However, the clinical applications of hLF are limited by its low stability in blood. We therefore attempted to resolve this by producing recombinant hLF fused to human serum albumin (HSA). Two HSA-fused hLFs with different fusion orientations (hLF-HSA and HSA-hLF) were produced in Chinese hamster ovary (CHO) DG44 cells. hLF-HSA revealed higher thermal stability, resistance to peptic degradation, and stability during the process of cellular uptake and release in an intestinal enterocyte model (Caco-2 cells) than HSA-hLF. The lower stability of HSA-hLF is presumably due to the steric hindrance imposed by HSA fusion to the N-terminus of hLF. Both HSA fusion proteins, especially HSA-hLF, displayed improved pharmacokinetic properties despite the lower protein stability of HSA-hLF. hLF-HSA and HSA-hLF exhibited approximately 3.3- and 20.7-fold longer half-lives (64.0 and 403.6 min), respectively, than holo-rhLF (19.5 min). Both HSA fusion proteins were found to exert enhanced growth inhibition effects on cancer cells in vitro, but not normal cells. Their enhanced growth inhibitory activities were considered to be due to the synergetic effects of hLF and HSA because hLF alone or HSA alone failed to exert such an effect. Altogether, Fusion of HSA to hLF yielded superior pharmacokinetics and anti-proliferative activities against cancer cells. HSA-fused hLF is a novel candidate for further application of hLF as biopharmaceuticals for intravenous administration.
Collapse
Affiliation(s)
- Keisuke Ueda
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1, Katakura, Hachioji, Tokyo, 192-0982, Japan
| | - Maya Shimizu
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1, Katakura, Hachioji, Tokyo, 192-0982, Japan
| | - Aimi Ohashi
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1, Katakura, Hachioji, Tokyo, 192-0982, Japan
| | - Daisuke Murata
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1, Katakura, Hachioji, Tokyo, 192-0982, Japan
| | - Takuo Suzuki
- Division of Biological Chemistry and Biologicals, National Institute of Health, Sciences, Kawasaki, Kanagawa, 210-9501, Japan
| | - Natsuki Kobayashi
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1, Katakura, Hachioji, Tokyo, 192-0982, Japan
| | - Junpei Baba
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1, Katakura, Hachioji, Tokyo, 192-0982, Japan
| | - Takashi Takeuchi
- Department of Veterinary Medicine, Tottori University, Koyama-Minami, Tottori, 680-8553, Japan
| | - Yuki Shiga
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1, Katakura, Hachioji, Tokyo, 192-0982, Japan
| | - Masao Nakamura
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1, Katakura, Hachioji, Tokyo, 192-0982, Japan
| | - Shinji Kagaya
- NRL Pharma, Inc., Kawasaki, Kanagawa, 213-0012, Japan
| | - Atsushi Sato
- School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1, Katakura, Hachioji, Tokyo, 192-0982, Japan.
| |
Collapse
|
32
|
Sasaki H, Pham Thi Ngoc D, Nishikawa M, Kanauchi M. Lipopolysaccharide neutralizing protein in Miso, Japanese fermented soybean paste. J Food Sci 2020; 85:2498-2505. [PMID: 32632955 DOI: 10.1111/1750-3841.15315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/08/2020] [Accepted: 05/14/2020] [Indexed: 11/29/2022]
Abstract
Miso, a fermented paste made from soybeans, is used traditionally for seasoning of food. It has been a protein and nitrogen source since ancient times in Japan because of its high nutritional value. Furthermore, it has important health functions such as the estrogen-like activity of isoflavones, anti-oxidation, and angiotensin-converting-enzyme inhibition activity. Moreover, it has activity for neutralization of lipopolysaccharide (LPS) from Escherichia coli. Nevertheless, the mechanisms of that activity remain unclear. For this study, we purified and identified the proteins responsible for LPS-neutralization. After proteins were isolated from a miso extract using Blue native polyacrylamide gel electrophoresis, a protein found at 10 to 30 kDa on the polyacrylamide gel was identified using nano LC-MS/MS as 2S albumin in soybean (Glycine max). The protein had two LPS binding motifs: SKWQHQ (22 amino acid residues) and EKQKKKMEKE (131 amino acid residues). The protein in miso was found to have LPS neutralization activity, as assayed by prostaglandin D2 (PGD2 ) production from macrophage cells. The PGD2 production by macrophage cells was inhibited by LPS-neutralizing protein (LNP) from miso. Particularly, 50 mg/mL of LNP solution and LPS (10 µg/mL) inhibited production of PGD2 from the cells. The data were inferred as significantly different (P < 0.05) from statistical analyses by analysis of variance testing and Tukey tests. The 2S albumin in soybean is LNP, an LPS-neutralizing protein, produced in miso. PRACTICAL APPLICATION: A protein from miso fermented soy paste neutralizes an Escherichia coli intestinal bacterial product, lipopolysaccharide (LPS), which causes intestinal inflammation. Miso and its protein are anticipated for use as a probiotic agent to prevent intestinal inflammation in humans and domestic animals. Miso is useful not only as a seasoning for food, but also as a health-functional food because it is an LPS-neutralizing agent.
Collapse
Affiliation(s)
- Hitomi Sasaki
- School of Food, Agricultural and Environmental Sciences, Miyagi University, 2-2-1 Hatatate, Taihaku-ku, Sendai, Miyagi, Japan
| | - Diep Pham Thi Ngoc
- School of Food, Agricultural and Environmental Sciences, Miyagi University, 2-2-1 Hatatate, Taihaku-ku, Sendai, Miyagi, Japan
| | - Masazumi Nishikawa
- School of Food, Agricultural and Environmental Sciences, Miyagi University, 2-2-1 Hatatate, Taihaku-ku, Sendai, Miyagi, Japan
| | - Makoto Kanauchi
- School of Food, Agricultural and Environmental Sciences, Miyagi University, 2-2-1 Hatatate, Taihaku-ku, Sendai, Miyagi, Japan
| |
Collapse
|
33
|
Wright SW, Lovelace-Macon L, Ducken D, Tandhavanant S, Teparrukkul P, Hantrakun V, Limmathurotsakul D, Chantratita N, West TE. Lactoferrin is a dynamic protein in human melioidosis and is a TLR4-dependent driver of TNF-α release in Burkholderia thailandensis infection in vitro. PLoS Negl Trop Dis 2020; 14:e0008495. [PMID: 32764765 PMCID: PMC7439809 DOI: 10.1371/journal.pntd.0008495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 08/19/2020] [Accepted: 06/18/2020] [Indexed: 11/19/2022] Open
Abstract
Melioidosis is an often-severe tropical infection caused by Burkholderia pseudomallei (Bp) with high associated morbidity and mortality. Burkholderia thailandensis (Bt) is a closely related surrogate that does not require BSL-3 conditions for study. Lactoferrin is an iron-binding glycoprotein that can modulate the innate inflammatory response. Here we investigated the impact of lactoferrin on the host immune response in melioidosis. Lactoferrin concentrations were measured in plasma from patients with melioidosis and following ex vivo stimulation of blood from healthy individuals. Bt growth was quantified in liquid media in the presence of purified and recombinant human lactoferrin. Differentiated THP-1 cells and human blood monocytes were infected with Bt in the presence of purified and recombinant human lactoferrin, and bacterial intracellular replication and cytokine responses (tumor necrosis factor-α (TNF-α), interleukin-1β and interferon-γ) were measured. In a cohort of 49 melioidosis patients, non-survivors to 28 days had significantly higher plasma lactoferrin concentrations compared to survivors (median (interquartile range (IQR)): 326 ng/ml (230-748) vs 144 ng/ml (99-277), p<0.001). In blood stimulated with heat-killed Bp, plasma lactoferrin concentration significantly increased compared to unstimulated blood (median (IQR): 424 ng/ml (349-479) vs 130 ng/ml (91-214), respectively; p<0.001). Neither purified nor recombinant human lactoferrin impaired growth of Bt in media. Lactoferrin significantly increased TNF-α production by differentiated THP-1 cells and blood monocytes after Bt infection. This phenotype was largely abrogated when Toll-like receptor 4 (TLR4) was blocked with a monoclonal antibody. In sum, lactoferrin is produced by blood cells after exposure to Bp and lactoferrin concentrations are higher in 28-day survivors in melioidosis. Lactoferrin induces proinflammatory cytokine production after Bt infection that may be TLR4 dependent.
Collapse
Affiliation(s)
- Shelton W. Wright
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Lara Lovelace-Macon
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Deirdre Ducken
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Sarunporn Tandhavanant
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Prapit Teparrukkul
- Department of Internal Medicine, Sunpasitthiprasong Hospital, Ubon Ratchathani, Thailand
| | - Viriya Hantrakun
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Direk Limmathurotsakul
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Narisara Chantratita
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - T. Eoin West
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
34
|
Lu J, Francis J, Doster RS, Haley KP, Craft KM, Moore RE, Chambers SA, Aronoff DM, Osteen K, Damo SM, Manning S, Townsend SD, Gaddy JA. Lactoferrin: A Critical Mediator of Both Host Immune Response and Antimicrobial Activity in Response to Streptococcal Infections. ACS Infect Dis 2020; 6:1615-1623. [PMID: 32329605 DOI: 10.1021/acsinfecdis.0c00050] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Streptococcal species are Gram-positive bacteria responsible for a variety of disease outcomes including pneumonia, meningitis, endocarditis, erysipelas, necrotizing fasciitis, periodontitis, skin and soft tissue infections, chorioamnionitis, premature rupture of membranes, preterm birth, and neonatal sepsis. In response to streptococcal infections, the host innate immune system deploys a repertoire of antimicrobial and immune modulating molecules. One important molecule that is produced in response to streptococcal infections is lactoferrin. Lactoferrin has antimicrobial properties including the ability to bind iron with high affinity and sequester this important nutrient from an invading pathogen. Additionally, lactoferrin has the capacity to alter the host inflammatory response and contribute to disease outcome. This Review presents the most recent published work that studies the interaction between the host innate immune protein lactoferrin and the invading pathogen, Streptococcus.
Collapse
Affiliation(s)
- Jacky Lu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Jamisha Francis
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Ryan S. Doster
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Kathryn P. Haley
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan 49401, United States
| | - Kelly M. Craft
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Rebecca E. Moore
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Schuyler A. Chambers
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - David M. Aronoff
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Kevin Osteen
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Veterans Affairs, Tennessee Valley Healthcare Systems, Nashville, Tennessee 37212, United States
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Steven M. Damo
- Department of Chemistry, Fisk University, Nashville, Tennessee 37208, United States
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Shannon Manning
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Steven D. Townsend
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Jennifer A. Gaddy
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Veterans Affairs, Tennessee Valley Healthcare Systems, Nashville, Tennessee 37212, United States
| |
Collapse
|
35
|
Rosa L, Lepanto MS, Cutone A, Ianiro G, Pernarella S, Sangermano R, Musci G, Ottolenghi L, Valenti P. Lactoferrin and oral pathologies: a therapeutic treatment. Biochem Cell Biol 2020; 99:81-90. [PMID: 32213143 DOI: 10.1139/bcb-2020-0052] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The oral cavity is a non-uniform, extraordinary environment characterized by mucosal, epithelial, abiotic surfaces and secretions as saliva. Aerobic and anaerobic commensal and pathogenic microorganisms colonize the tongue, teeth, jowl, gingiva, and periodontium. Commensals exert an important role in host defenses, while pathogenic microorganisms can nullify this protective function causing oral and systemic diseases. Every day, 750-1000 mL of saliva, containing several host defense constituents including lactoferrin (Lf), are secreted and swallowed. Lf is a multifunctional iron-chelating cationic glycoprotein of innate immunity. Depending on, or regardless of its iron-binding ability, Lf exerts bacteriostatic, bactericidal, antibiofilm, antioxidant, antiadhesive, anti-invasive, and anti-inflammatory activities. Here, we report the protective role of Lf in different oral pathologies, such as xerostomia, halitosis, alveolar or maxillary bone damage, gingivitis, periodontitis, and black stain. Unlike antibiotic therapy, which is ineffective against bacteria that are within a biofilm, adherent, or intracellular, the topical administration of Lf, through its simultaneous activity against microbial replication, biofilms, adhesion, and invasiveness, as well as inflammation, has been proven to be efficient in the treatment of all known oral pathologies without any adverse effects.
Collapse
Affiliation(s)
- Luigi Rosa
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Italy
| | | | - Antimo Cutone
- Department of Biosciences and Territory, University of Molise, Pesche, Italy
| | - Giusi Ianiro
- Department of Biosciences and Territory, University of Molise, Pesche, Italy
| | - Stefania Pernarella
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, Italy
| | - Riccardo Sangermano
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Italy
| | - Giovanni Musci
- Department of Biosciences and Territory, University of Molise, Pesche, Italy
| | - Livia Ottolenghi
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, Italy
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Italy
| |
Collapse
|
36
|
Gopalakrishna KP, Hand TW. Influence of Maternal Milk on the Neonatal Intestinal Microbiome. Nutrients 2020; 12:E823. [PMID: 32244880 PMCID: PMC7146310 DOI: 10.3390/nu12030823] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/13/2020] [Accepted: 03/18/2020] [Indexed: 02/07/2023] Open
Abstract
The intestinal microbiome plays an important role in maintaining health throughout life. The microbiota develops progressively after birth and is influenced by many factors, including the mode of delivery, antibiotics, and diet. Maternal milk is critically important to the development of the neonatal intestinal microbiota. Different bioactive components of milk, such as human milk oligosaccharides, lactoferrin, and secretory immunoglobulins, modify the composition of the neonatal microbiota. In this article, we review the role of each of these maternal milk-derived bioactive factors on the microbiota and how this modulation of intestinal bacteria shapes health, and disease.
Collapse
Affiliation(s)
| | - Timothy W. Hand
- R. K. Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15217, USA;
| |
Collapse
|
37
|
Avalos-Gómez C, Reyes-López M, Ramírez-Rico G, Díaz-Aparicio E, Zenteno E, González-Ruiz C, de la Garza M. Effect of apo-lactoferrin on leukotoxin and outer membrane vesicles of Mannheimia haemolytica A2. Vet Res 2020; 51:36. [PMID: 32138772 PMCID: PMC7059318 DOI: 10.1186/s13567-020-00759-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/17/2020] [Indexed: 01/17/2023] Open
Abstract
Mannheimia haemolytica serotype A2 is the principal cause of pneumonic mannheimiosis in ovine and caprine livestock; this disease is a consequence of immune suppression caused by stress and associated viruses and is responsible for significant economic losses in farm production worldwide. Gram-negative bacteria such as M. haemolytica produce outer membrane (OM)-derived spherical structures named outer membrane vesicles (OMVs) that contain leukotoxin and other biologically active virulence factors. In the present study, the relationship between M. haemolytica A2 and bovine lactoferrin (BLf) was studied. BLf is an 80 kDa glycoprotein that possesses bacteriostatic and bactericidal properties and is part of the mammalian innate immune system. Apo-BLf (iron-free) showed a bactericidal effect against M. haemolytica A2, with an observed minimal inhibitory concentration (MIC) of 16 µM. Sublethal doses (2–8 µM) of apo-BLf increased the release of OMVs, which were quantified by flow cytometry. Apo-BLf modified the normal structure of the OM and OMVs, as observed through transmission electron microscopy. Apo-BLf also induced lipopolysaccharide (LPS) release from bacteria, disrupting OM permeability and functionality, as measured by silver staining and SDS and polymyxin B cell permeability assays. Western blot results showed that apo-BLf increased the secretion of leukotoxin in M. haemolytica A2 culture supernatants, possibly through its iron-chelating activity. In contrast, holo-BLf (with iron) did not have this effect, possibly due to differences in the tertiary structure between these proteins. In summary, apo-BLf affected the levels of several M. haemolytica virulence factors and could be evaluated for use in animals as an adjuvant in the treatment of ovine mannheimiosis.
Collapse
Affiliation(s)
- Christian Avalos-Gómez
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), 04510, Coyoacán, CdMx, Mexico.,Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ave. Instituto Politécnico Nacional 2508, Zacatenco, 07360, CdMx, Mexico
| | - Magda Reyes-López
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ave. Instituto Politécnico Nacional 2508, Zacatenco, 07360, CdMx, Mexico
| | - Gerardo Ramírez-Rico
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), 54714, Cuautitlán Izcalli, Estado de México, Mexico
| | - Efrén Díaz-Aparicio
- Centro Nacional de Investigación Disciplinaria en Salud animal e inocuidad, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias (INIFAP), 05110, Cuajimalpa, CdMx, Mexico
| | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), 04510, Coyoacán, CdMx, Mexico
| | - Cynthia González-Ruiz
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), 54714, Cuautitlán Izcalli, Estado de México, Mexico
| | - Mireya de la Garza
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ave. Instituto Politécnico Nacional 2508, Zacatenco, 07360, CdMx, Mexico.
| |
Collapse
|
38
|
Magacz M, Kędziora K, Sapa J, Krzyściak W. The Significance of Lactoperoxidase System in Oral Health: Application and Efficacy in Oral Hygiene Products. Int J Mol Sci 2019; 20:ijms20061443. [PMID: 30901933 PMCID: PMC6472183 DOI: 10.3390/ijms20061443] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/13/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022] Open
Abstract
Lactoperoxidase (LPO) present in saliva are an important element of the nonspecific immune response involved in maintaining oral health. The main role of this enzyme is to oxidize salivary thiocyanate ions (SCN-) in the presence of hydrogen peroxide (H₂O₂) to products that exhibit antimicrobial activity. LPO derived from bovine milk has found an application in food, cosmetics, and medical industries due to its structural and functional similarity to the human enzyme. Oral hygiene products enriched with the LPO system constitute an alternative to the classic fluoride caries prophylaxis. This review describes the physiological role of human salivary lactoperoxidase and compares the results of clinical trials and in vitro studies of LPO alone and complex dentifrices enriched with bovine LPO. The role of reactivators and inhibitors of LPO is discussed together with the possibility of using nanoparticles to increase the stabilization and activity of this enzyme.
Collapse
Affiliation(s)
- Marcin Magacz
- Department of Medical Diagnostics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland.
| | - Karolina Kędziora
- Department of Medical Diagnostics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland.
| | - Jacek Sapa
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland.
| | - Wirginia Krzyściak
- Department of Medical Diagnostics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland.
| |
Collapse
|
39
|
Koshi R, Kotani K, Ohtsu M, Yoshinuma N, Sugano N. Application of Lactoferrin and α1-Antitrypsin in Gingival Retention Fluid to Diagnosis of Periodontal Disease. DISEASE MARKERS 2018; 2018:4308291. [PMID: 30524520 PMCID: PMC6247577 DOI: 10.1155/2018/4308291] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/26/2018] [Accepted: 10/08/2018] [Indexed: 12/03/2022]
Abstract
OBJECTIVES Periodontal disease is prevalent and has an inflammation associated with not only oral but also systemic pathologies. The diagnosis by biomarkers is required for clinical practice on periodontal disease. The lactoferrin and α1-antitrypsin were both inflammation-related molecules. The present study investigated the relationship between the periodontal status and the two biomarkers in gingival retention fluid (GRF). PATIENTS AND METHODS In 63 subjects with periodontitis, the GRF was sampled from maxillary anterior gingiva using a microbrush for 30 seconds. The lactoferrin and α1-antitrypsin levels in GRF were measured by an enzyme-link solvent immunoassay. Periodontal status was evaluated by probing pocket depth (PD) and bleeding on probing (BOP). RESULTS There was a higher level of these biomarkers in saliva (median (ng/mL), lactoferrin: 3611.9, α1-antitrypsin: 4573.3) than in GRF (lactoferrin: 61.0, α1-antitrypsin: 54.7). There was a mild-to-moderate but significantly positive correlation in lactoferrin or α1-antitrypsin between GRF and saliva. There was a positively mild-to-moderate accuracy (area under the curve: 0.60-0.81) of lactoferrin or α1-antitrypsin in GRF or in saliva to distinguish the severity of periodontal status. The cutoff level (ng/mL) of lactoferrin in GRF for detecting ≥30% of PD ≥ 4 mm (moderate periodontitis) was 68.6 and for detecting ≥20% of BOP (clinically active periodontitis) was 61.2. The cutoff level (ng/mL) of α1-antitrypsin in GRF for detecting ≥30% of PD ≥ 4 mm was 54.5 and for detecting ≥20% of BOP was 35.3. CONCLUSIONS The data can promote an application of the measurements of lactoferrin and α1-antitrypsin in GRF to clinical practice on periodontal disease.
Collapse
Affiliation(s)
- Ryosuke Koshi
- Department of Periodontology, Nihon University School of Dentistry, Tokyo, Japan
| | - Kazuhiko Kotani
- Division of Community and Family Medicine, Jichi Medical University, Tochigi, Japan
| | - Mariko Ohtsu
- Department of Pathology, Nihon University School of Dentistry, Tokyo, Japan
| | - Naoto Yoshinuma
- Department of Periodontology, Nihon University School of Dentistry, Tokyo, Japan
| | - Naoyuki Sugano
- Department of Periodontology, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
40
|
Marth CD, Firestone SM, Hanlon D, Glenton LY, Browning GF, Young ND, Krekeler N. Innate immune genes in persistent mating-induced endometritis in horses. Reprod Fertil Dev 2018; 30:533-545. [PMID: 28834688 DOI: 10.1071/rd17157] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/29/2017] [Indexed: 11/23/2022] Open
Abstract
Persistent mating-induced endometritis (PMIE) severely decreases fertility in horses. The aim of the present study was to evaluate differences between horses susceptible to PMIE and a control group in terms of the expression of selected immune response and effector genes, and the effects of oestrous cycle stage on this expression. Endometrial biopsies from 18 uterine samples of mares in the control group (eight in dioestrus, 10 in oestrus) and 16 PMIE-susceptible mares (four in dioestrus, 12 in oestrus) were analysed by quantitative real-time reverse transcription-polymerase chain reaction. Genes for pathogen recognition receptors Toll-like receptor 2 (TLR2) and NLR family CARD domain containing 5 (NLRC5), as well as tissue-specific inhibitor of metalloproteinase 1 (TIMP1), C-X-C motif chemokine ligand (CXCL) 9, CXCL10 and CXCL11 and uteroferrin were expressed at similar levels in the control group and in susceptible mares. Genes for C-C motif chemokine ligand 2 (CCL2) and the antimicrobial peptides secreted phospholipase A2 (sPLA2), lipocalin 2 and lactoferrin were all expressed at higher levels in susceptible compared with control mares. The expression of genes for the antimicrobial peptides equine β-defensin 1 (EBD1), lysozyme (LYZ) and secretory leukoprotease inhibitor (SLPI) was also higher in susceptible than control mares. The diagnostic sensitivity of assays for EBD1, LYZ and SLP1 gene expression to detect susceptibility to PMIE was estimated to be 100%, 94% and 100% respectively, with specificities of 83%, 78% and 78% respectively. When all three tests were positive, the specificity increased to 94%, with an overall sensitivity of 94%. The present study has yielded insights into pathophysiological changes in mares susceptible to PMIE and identified robust diagnostic markers (EBD1, LYZ and SLPI) for susceptibility to this disease.
Collapse
Affiliation(s)
- Christina D Marth
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Corner Park Drive and Flemington Road, Parkville, Vic. 3010, Australia
| | - Simon M Firestone
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Corner Park Drive and Flemington Road, Parkville, Vic. 3010, Australia
| | - Dave Hanlon
- Matamata Veterinary Services, 26 Tainui Street, Matamata, New Zealand
| | - Lisa Y Glenton
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Corner Park Drive and Flemington Road, Parkville, Vic. 3010, Australia
| | - Glenn F Browning
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Corner Park Drive and Flemington Road, Parkville, Vic. 3010, Australia
| | - Neil D Young
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Corner Park Drive and Flemington Road, Parkville, Vic. 3010, Australia
| | - Natali Krekeler
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Corner Park Drive and Flemington Road, Parkville, Vic. 3010, Australia
| |
Collapse
|
41
|
Telang S. Lactoferrin: A Critical Player in Neonatal Host Defense. Nutrients 2018; 10:nu10091228. [PMID: 30181493 PMCID: PMC6165050 DOI: 10.3390/nu10091228] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/30/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
Newborn infants are at a high risk for infection due to an under-developed immune system, and human milk has been shown to exhibit substantial anti-infective properties that serve to bolster neonatal defenses against multiple infections. Lactoferrin is the dominant whey protein in human milk and has been demonstrated to perform a wide array of antimicrobial and immunomodulatory functions and play a critical role in protecting the newborn infant from infection. This review summarizes data describing the structure and important functions performed by lactoferrin in protecting the neonate from infection and contributing to the maturation of the newborn innate and adaptive immune systems. We also briefly discuss clinical trials examining the utility of lactoferrin supplementation in the prevention of sepsis and necrotizing enterocolitis in newborn infants. The data reviewed provide rationale for the continuation of studies to examine the effects of lactoferrin administration on the prevention of sepsis in the neonate.
Collapse
Affiliation(s)
- Sucheta Telang
- Division of Neonatology, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA.
- Division of Hematology/Oncology, Department of Medicine, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
42
|
Vargas Buonfiglio LG, Borcherding JA, Frommelt M, Parker GJ, Duchman B, Vanegas Calderón OG, Fernandez-Ruiz R, Noriega JE, Stone EA, Gerke AK, Zabner J, Comellas AP. Airway surface liquid from smokers promotes bacterial growth and biofilm formation via iron-lactoferrin imbalance. Respir Res 2018. [PMID: 29524964 PMCID: PMC5845328 DOI: 10.1186/s12931-018-0743-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Smoking is a leading cause of respiratory infections worldwide. Tobacco particulate matter disrupts iron homeostasis in the lungs and increases the iron content in the airways of smokers. The airway epithelia secrete lactoferrin to quench iron required for bacteria to proliferate and cause lung infections. We hypothesized that smokers would have increased bacterial growth and biofilm formation via iron lactoferrin imbalance. Methods We collected bronchoalveolar lavage (BAL) samples from non-smokers and smokers. We challenged these samples using a standard inoculum of Staphylococcus aureus and Pseudomonas aeruginosa and quantified bacterial growth and biofilm formation. We measured both iron and lactoferrin in the samples. We investigated the effect of supplementing non-smoker BAL with cigarette smoke extract (CSE) or ferric chloride and the effect of supplementing smoker BAL with lactoferrin on bacterial growth and biofilm formation. Results BAL from smokers had increased bacterial growth and biofilm formation compared to non-smokers after both S. aureus and P. aeruginosa challenge. In addition, we found that samples from smokers had a higher iron to lactoferrin ratio. Supplementing the BAL of non-smokers with cigarette smoke extract and ferric chloride increased bacterial growth. Conversely, supplementing the BAL of smokers with lactoferrin had a concentration-dependent decrease in bacterial growth and biofilm formation. Conclusion Cigarette smoking produces factors which increase bacterial growth and biofilm formation in the BAL. We propose that smoking disrupts the iron-to-lactoferrin in the airways. This finding offers a new avenue for potential therapeutic interventions to prevent respiratory infections in smokers. Electronic supplementary material The online version of this article (10.1186/s12931-018-0743-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Luis G Vargas Buonfiglio
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 6312 Pappajohn Biomedical Discovery Building. Newton Road, Iowa City, IA, 52242, USA
| | - Jennifer A Borcherding
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 6312 Pappajohn Biomedical Discovery Building. Newton Road, Iowa City, IA, 52242, USA
| | - Mark Frommelt
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 6312 Pappajohn Biomedical Discovery Building. Newton Road, Iowa City, IA, 52242, USA
| | - Gavin J Parker
- Department of Chemistry, College of Liberal Arts & Sciences, University of Iowa, Iowa City, IA, USA
| | - Bryce Duchman
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 6312 Pappajohn Biomedical Discovery Building. Newton Road, Iowa City, IA, 52242, USA
| | - Oriana G Vanegas Calderón
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ruth Fernandez-Ruiz
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 6312 Pappajohn Biomedical Discovery Building. Newton Road, Iowa City, IA, 52242, USA
| | - Julio E Noriega
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 6312 Pappajohn Biomedical Discovery Building. Newton Road, Iowa City, IA, 52242, USA
| | - Elizabeth A Stone
- Department of Chemistry, College of Liberal Arts & Sciences, University of Iowa, Iowa City, IA, USA
| | - Alicia K Gerke
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 6312 Pappajohn Biomedical Discovery Building. Newton Road, Iowa City, IA, 52242, USA
| | - Joseph Zabner
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 6312 Pappajohn Biomedical Discovery Building. Newton Road, Iowa City, IA, 52242, USA
| | - Alejandro P Comellas
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 6312 Pappajohn Biomedical Discovery Building. Newton Road, Iowa City, IA, 52242, USA.
| |
Collapse
|
43
|
Abstract
Human and bovine lactoferrin (hLf and bLf) are multifunctional iron-binding glycoprotein constitutively synthesized and secreted by glandular epithelial cells and by neutrophils following induction. HLf and bLf possess very high similarity of sequence. Therefore, most of the in vitro and in vivo studies are carried out with commercial bLf (cbLf), available in large quantities and recognized by Food and Drug Administration (FDA, USA) as a safe substance. Physico-chemical heterogeneity of different cbLf preparations influences their effectiveness. CbLf iron-saturation affects thermal stability and resistance to proteolysis. Moreover, other metal ions such as Al(III), Cu(II), Mg(II), Mn(II), Zn(II) are chelated by cbLf, even if at lower affinity than Fe(III). Ca(II) is also sequestered by the carboxylate groups of sialic acid present on glycan chains of cbLf thus provoking the release of LPS, contributing to bactericidal activity. Similarly to more than 50% of eukaryotic proteins, cbLf possesses five N-glycosylation sites, also contributing to the resistance to proteolysis and, putatively, to the protection of intestinal mucosa from pathogens. CbLfs possess several functions as anti-microbial, anti-biofilm, anti-adhesive, anti-invasive and anti-inflammatory activities. They are also relevant modulators of iron and inflammatory homeostasis. However, the efficacy of cbLfs in exerting several functions can be erratic mainly depending from integrity, degree of iron and other metal ions saturation, N-glycosylation sites and chains, desialylated forms, Ca(II) sequestration, presence of contaminants and finally the ability to enter inside nucleus.
Collapse
|
44
|
Valenti P, Rosa L, Capobianco D, Lepanto MS, Schiavi E, Cutone A, Paesano R, Mastromarino P. Role of Lactobacilli and Lactoferrin in the Mucosal Cervicovaginal Defense. Front Immunol 2018; 9:376. [PMID: 29545798 PMCID: PMC5837981 DOI: 10.3389/fimmu.2018.00376] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/09/2018] [Indexed: 01/26/2023] Open
Abstract
The innate defense system of the female mucosal genital tract involves a close and complex interaction among the healthy vaginal microbiota, different cells, and various proteins that protect the host from pathogens. Vaginal lactobacilli and lactoferrin represent two essential actors in the vaginal environment. Lactobacilli represent the dominant bacterial species able to prevent facultative and obligate anaerobes outnumber in vaginal microbiota maintaining healthy microbial homeostasis. Several mechanisms underlie the protection exerted by lactobacilli: competition for nutrients and tissue adherence, reduction of the vaginal pH, modulation of immunity, and production of bioactive compounds. Among bioactive factors of cervicovaginal mucosa, lactoferrin, an iron-binding cationic glycoprotein, is a multifunctional glycoprotein with antibacterial, antifungal, antiviral, and antiparasitic activities, recently emerging as an important modulator of inflammation. Lactobacilli and lactoferrin are largely under the influence of female hormones and of paracrine production of various cytokines. Lactoferrin is strongly increased in lower genital tract mucosal fluid of women affected by Neisseria gonorrheae, Chlamydia trachomatis, and Trichomonas vaginalis infections promoting both innate and adaptive immune responses. In vaginal dysbiosis characterized by low amounts of vaginal lactobacilli and increased levels of endogenous anaerobic bacteria, the increase in lactoferrin could act as an immune modulator assuming the role normally played by the healthy microbiota in vaginal mucosa. Then lactoferrin and lactobacilli may be considered as biomarkers of altered microbial homeostasis at vaginal level. Considering the shortage of effective treatments to counteract recurrent and/or antibiotic-resistant bacterial infections, the intravaginal administration of lactobacilli and lactoferrin could be a novel efficient therapeutic strategy and a valuable tool to restore mucosal immune homeostasis.
Collapse
Affiliation(s)
- Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, Rome, Italy
| | - Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, Rome, Italy
| | - Daniela Capobianco
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, Rome, Italy
| | - Maria Stefania Lepanto
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, Rome, Italy
| | - Elisa Schiavi
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, Rome, Italy
| | - Antimo Cutone
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, Rome, Italy
| | - Rosalba Paesano
- Department of Gynecological-Obstetric and Urological Sciences, University of Rome La Sapienza, Rome, Italy
| | - Paola Mastromarino
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, Rome, Italy
| |
Collapse
|
45
|
Kieckens E, Rybarczyk J, Cox E, Vanrompay D. Antibacterial and immunomodulatory activities of bovine lactoferrin against Escherichia coli O157:H7 infections in cattle. Biometals 2018; 31:321-330. [PMID: 29442205 DOI: 10.1007/s10534-018-0082-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 02/06/2018] [Indexed: 12/27/2022]
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 is a zoonotic pathogen that causes food-borne disease in humans ranging from watery diarrhea to bloody diarrhea and severe hemorrhagic colitis, renal failure and hemolytic uremic syndrome. Cattle, the most important source of E. coli O157:H7 transmission to humans, harbor the bacteria in their gastrointestinal tract without showing clinical symptoms. Prevention of E. coli O157:H7 infections in ruminants could diminish the public health risk. However, there is no specific treatment available nor a vaccine or a therapeutic agent which completely prevents E. coli O157:H7 infections in cattle. This paper provides an overview of latest research data on eradicating enterohemorrhagic E. coli O157:H7 in ruminants by use of bovine lactoferrin administration. The article provides insights into the anti-microbial and immunomodulatory activities of bovine lactoferrin against E. coli O157:H7 infections in cattle.
Collapse
Affiliation(s)
- Evelien Kieckens
- Laboratory of Immunology and Animal Biotechnology, Department of Animal Production, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Joanna Rybarczyk
- Laboratory of Immunology and Animal Biotechnology, Department of Animal Production, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Eric Cox
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Daisy Vanrompay
- Laboratory of Immunology and Animal Biotechnology, Department of Animal Production, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium.
| |
Collapse
|
46
|
Figueroa‐Lozano S, Valk‐Weeber RL, van Leeuwen SS, Dijkhuizen L, de Vos P. Dietary N-Glycans from Bovine Lactoferrin and TLR Modulation. Mol Nutr Food Res 2018; 62:1700389. [PMID: 28971586 PMCID: PMC6120133 DOI: 10.1002/mnfr.201700389] [Citation(s) in RCA: 28] [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: 05/05/2017] [Revised: 07/22/2017] [Indexed: 12/29/2022]
Abstract
SCOPE Bovine lactoferrin (bLF) is an ingredient of food supplements and infant formulas given its antimicrobial and antiviral properties. We modified bLF enzymatically to alter its N-glycosylation and to isolate the glycan chains. The aims of this study include (1) to evaluate whether such derivates induce responses via pattern recognition receptors namely Toll-like receptors (TLRs) and (2) to relate those responses to their different glycosylation profiles. METHODS AND RESULTS The unmodified and modified bLF fractions are incubated with reporter cell lines expressing pattern recognition receptors. Afterwards, we screen for TLRs and analyze for nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) activation. Activation of reporter cell lines show that signaling is highly dependent on TLRs. The activation pattern of bLF is reduced with the desialylated form and increased with the demannosylated form. In reporter cells for TLR, bLF activate TLR-4 and inhibit TLR-3. The isolated glycans from bLF inhibit TLR-8. TLR-2, TLR-5, TLR-7, and TLR-9 are not significantly altered. CONCLUSION The profile of glycosylation is key for the biological activity of bLF. By understanding how this affects the human defense responses, the bLF glycan profile can be modified to enhance its immunomodulatory effects when used as a dietary ingredient.
Collapse
Affiliation(s)
- Susana Figueroa‐Lozano
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Rivca L. Valk‐Weeber
- Microbial PhysiologyGroningen Biomolecular Sciences and Biotechnology Institute (GBB)GroningenThe Netherlands
| | - Sander S. van Leeuwen
- Microbial PhysiologyGroningen Biomolecular Sciences and Biotechnology Institute (GBB)GroningenThe Netherlands
| | - Lubbert Dijkhuizen
- Microbial PhysiologyGroningen Biomolecular Sciences and Biotechnology Institute (GBB)GroningenThe Netherlands
| | - Paul de Vos
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| |
Collapse
|
47
|
In vitro and in vivo anticandidal activities of alginate-enclosed chitosan-calcium phosphate-loaded Fe-bovine lactoferrin nanocapsules. Future Sci OA 2017; 4:FSO257. [PMID: 29379633 PMCID: PMC5778379 DOI: 10.4155/fsoa-2017-0085] [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: 07/05/2017] [Accepted: 09/27/2017] [Indexed: 12/28/2022] Open
Abstract
Aim: To study the in vitro and in vivo anticandidal activity of nanocapsulated bovine lactoferrin. Materials & methods: In vitro and in vivo antimicrobial activities were conducted to study the anticandidal activities of nanocapsules (NCs). Results: The NCs showed good anticandidal activities. The disruption of cell wall and cell membrane was noted via microscopy studies. The NCs changed the normal growth profile of Candida albicans. NCs reduced the colony forming unit in kidney and blood samples. Histopathological examination showed better cell structure and coordination compared with untreated mice kidney. NCs also enhanced the natural killing properties of C. albicans by epithelial cells. Conclusion: NCs have effective anticandidal properties and have the potential as a therapeutic agent against candidiasis. Previous study revealed that lactoferrin had potent anticandidal action against C. albicans. However, encapsulated lactoferrin has never been tested for anticandidal activity in detail. In the present study, we evaluate nanocapsulated lactoferrin for anticandidal effects. To observe the anticandidal properties of encapsulated lactoferrin, various studies were conducted. Our findings showed that encapsulated lactoferrin demonstrates remarkable efficacy against C. albicans.
Collapse
|
48
|
Sharma N, Huynh DL, Kim SW, Ghosh M, Sodhi SS, Singh AK, Kim NE, Lee SJ, Hussain K, Oh SJ, Jeong DK. A PiggyBac mediated approach for lactoferricin gene transfer in bovine mammary epithelial stem cells for management of bovine mastitis. Oncotarget 2017; 8:104272-104285. [PMID: 29262639 PMCID: PMC5732805 DOI: 10.18632/oncotarget.22210] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 09/21/2017] [Indexed: 12/31/2022] Open
Abstract
The antibacterial and anti-inflammatory properties of lactoferricin have been ascribed to its ability to sequester essential iron. The objective of the study was to clone bovine lactoferricin (LFcinB) gene into PiggyBac Transposon vector, expression study in the bovine mammary epithelial stem cells (bMESCs) and also to determine the antimicrobial property of recombinant LFcinB against bovine mastitis-causing organisms. The PiggyBac-LFcinB was transfected into bMESCs by electroporation and a three fold of LFcinB secretion was observed in the transfected bMESCs medium by ELISA assay. Furthermore, the assessment of antimicrobial activity against mastitis causing pathogens Staphylococcus aureus and Escherichia coli demonstrated convincing evidence to prove strong antibacterial activity of LFcinB with 14.0±1.0 mm and 18.0±1.5 mm zone of inhibition against both organisms, respectively. The present study provides the convincing evidence to suggest the potential of PiggyBac transposon system to transfer antibacterial peptide into bMESCs or cow mammary gland and also pave the way to use bovine mammary gland as the bioreactors. Simultaneously, it also suggest toward commercial utilization of LFcinB bioreactor system in pharmaceutical industry.
Collapse
Affiliation(s)
- Neelesh Sharma
- Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Republic of Korea.,Division of Veterinary Medicine, Faculty of Veterinary Science & Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences & Technology of Jammu, Jammu, India
| | - Do Luong Huynh
- Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Republic of Korea
| | - Sung Woo Kim
- Animal Genetic Resources Station, National Institute of Animal Science, Rural Development Administration, Namwon, Republic of Korea
| | - Mrinmoy Ghosh
- Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Republic of Korea
| | - Simrinder Singh Sodhi
- Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Republic of Korea
| | - Amit Kumar Singh
- Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Republic of Korea
| | - Nam Eun Kim
- Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Republic of Korea
| | - Sung Jin Lee
- Department of Animal Biotechnology, College of Animal Bioscience and Technology, Kangwon National University, Chuncheon, Republic of Korea
| | - Kafil Hussain
- Division of Veterinary Medicine, Faculty of Veterinary Science & Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences & Technology of Jammu, Jammu, India
| | - Sung Jong Oh
- National Institute of Animal Science, Wanju-gun, Republic of Korea
| | - Dong Kee Jeong
- Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
49
|
Rosa L, Cutone A, Lepanto MS, Paesano R, Valenti P. Lactoferrin: A Natural Glycoprotein Involved in Iron and Inflammatory Homeostasis. Int J Mol Sci 2017; 18:1985. [PMID: 28914813 PMCID: PMC5618634 DOI: 10.3390/ijms18091985&n948647=v984776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human lactoferrin (hLf), an iron-binding multifunctional cationic glycoprotein secreted by exocrine glands and by neutrophils, is a key element of host defenses. HLf and bovine Lf (bLf), possessing high sequence homology and identical functions, inhibit bacterial growth and biofilm dependently from iron binding ability while, independently, bacterial adhesion to and the entry into cells. In infected/inflamed host cells, bLf exerts an anti-inflammatory activity against interleukin-6 (IL-6), thus up-regulating ferroportin (Fpn) and transferrin receptor 1 (TfR1) and down-regulating ferritin (Ftn), pivotal actors of iron and inflammatory homeostasis (IIH). Consequently, bLf inhibits intracellular iron overload, an unsafe condition enhancing in vivo susceptibility to infections, as well as anemia of inflammation (AI), re-establishing IIH. In pregnant women, affected by AI, bLf oral administration decreases IL-6 and increases hematological parameters. This surprising effect is unrelated to iron supplementation by bLf (80 μg instead of 1-2 mg/day), but to its role on IIH. AI is unrelated to the lack of iron, but to iron delocalization: cellular/tissue overload and blood deficiency. BLf cures AI by restoring iron from cells to blood through Fpn up-expression. Indeed, anti-inflammatory activity of oral and intravaginal bLf prevents preterm delivery. Promising bLf treatments can prevent/cure transitory inflammation/anemia/oral pathologies in athletes.
Collapse
Affiliation(s)
- Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Antimo Cutone
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Maria Stefania Lepanto
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Rosalba Paesano
- Department of Gynecological-Obstetric and Urological Sciences, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| |
Collapse
|
50
|
Rosa L, Cutone A, Lepanto MS, Paesano R, Valenti P. Lactoferrin: A Natural Glycoprotein Involved in Iron and Inflammatory Homeostasis. Int J Mol Sci 2017; 18:E1985. [PMID: 28914813 PMCID: PMC5618634 DOI: 10.3390/ijms18091985] [Citation(s) in RCA: 227] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 02/07/2023] Open
Abstract
Human lactoferrin (hLf), an iron-binding multifunctional cationic glycoprotein secreted by exocrine glands and by neutrophils, is a key element of host defenses. HLf and bovine Lf (bLf), possessing high sequence homology and identical functions, inhibit bacterial growth and biofilm dependently from iron binding ability while, independently, bacterial adhesion to and the entry into cells. In infected/inflamed host cells, bLf exerts an anti-inflammatory activity against interleukin-6 (IL-6), thus up-regulating ferroportin (Fpn) and transferrin receptor 1 (TfR1) and down-regulating ferritin (Ftn), pivotal actors of iron and inflammatory homeostasis (IIH). Consequently, bLf inhibits intracellular iron overload, an unsafe condition enhancing in vivo susceptibility to infections, as well as anemia of inflammation (AI), re-establishing IIH. In pregnant women, affected by AI, bLf oral administration decreases IL-6 and increases hematological parameters. This surprising effect is unrelated to iron supplementation by bLf (80 μg instead of 1-2 mg/day), but to its role on IIH. AI is unrelated to the lack of iron, but to iron delocalization: cellular/tissue overload and blood deficiency. BLf cures AI by restoring iron from cells to blood through Fpn up-expression. Indeed, anti-inflammatory activity of oral and intravaginal bLf prevents preterm delivery. Promising bLf treatments can prevent/cure transitory inflammation/anemia/oral pathologies in athletes.
Collapse
Affiliation(s)
- Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Antimo Cutone
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Maria Stefania Lepanto
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Rosalba Paesano
- Department of Gynecological-Obstetric and Urological Sciences, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| |
Collapse
|