1
|
Gao N, Bai P, Fang C, Wu W, Bi C, Wang J, Shan A. Biomimetic Peptide Nanonets: Exploiting Bacterial Entrapment and Macrophage Rerousing for Combatting Infections. ACS NANO 2024; 18:25446-25464. [PMID: 39240217 DOI: 10.1021/acsnano.4c03669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
The alarming rise in global antimicrobial resistance underscores the urgent need for effective antibacterial drugs. Drawing inspiration from the bacterial-entrapment mechanism of human defensin 6, we have fabricated biomimetic peptide nanonets composed of multiple functional fragments for bacterial eradication. These biomimetic peptide nanonets are designed to address antimicrobial resistance challenges through a dual-approach strategy. First, the resulting nanofibrous networks trap bacteria and subsequently kill them by loosening the membrane structure, dissipating proton motive force, and causing multiple metabolic perturbations. Second, these trapped bacterial clusters reactivate macrophages to scavenge bacteria through enhanced chemotaxis and phagocytosis via the PI3K-AKT signaling pathway and ECM-receptor interaction. In vivo results have proven that treatment with biomimetic peptide nanonets can alleviate systemic bacterial infections without causing noticeable systemic toxicity. As anticipated, the proposed strategy can address stubborn infections by entrapping bacteria and awakening antibacterial immune responses. This approach might serve as a guide for the design of bioinspired materials for future clinical applications.
Collapse
Affiliation(s)
- Nan Gao
- College of animal science and technology, Northeast Agricultural University, Harbin 150030, China
| | - Pengfei Bai
- College of animal science and technology, Northeast Agricultural University, Harbin 150030, China
| | - Chunyang Fang
- College of animal science and technology, Northeast Agricultural University, Harbin 150030, China
| | - Wanpeng Wu
- College of animal science and technology, Northeast Agricultural University, Harbin 150030, China
| | - Chongpeng Bi
- College of animal science and technology, Northeast Agricultural University, Harbin 150030, China
| | - Jiajun Wang
- College of animal science and technology, Northeast Agricultural University, Harbin 150030, China
| | - Anshan Shan
- College of animal science and technology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
2
|
Watanabe M, Toyomura T, Wake H, Nishinaka T, Hatipoglu OF, Takahashi H, Nishibori M, Mori S. Cationic ribosomal proteins can inhibit pro-inflammatory action stimulated by LPS+HMGB1 and are hindered by advanced glycation end products. Biotechnol Appl Biochem 2024; 71:264-271. [PMID: 38010900 DOI: 10.1002/bab.2538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/12/2023] [Indexed: 11/29/2023]
Abstract
We previously found that ribosomal protein L9 (RPL9) is a novel advanced glycation end product (AGE)-binding protein that can decrease pro-inflammatory TNF-α expression stimulated by lipopolysaccharide (LPS) plus high-mobility group box 1 (HMGB1), suggesting that RPL9 has a role in regulating LPS+HMGB1-stimulated inflammatory reactions. Among the various ribosomal proteins, it was found that RPS5 reproduced the regulatory activity of RPL9 on LPS+HMGB1-stimulated TNF-α expression in macrophage-like RAW264.7 cells. RPL9 and RPS5 share a common feature as cationic proteins. Polylysine, a cationic polypeptide, and a synthetic peptide of the cationic region from RPL9 also exhibited reducing activity on LPS+HMGB1-induced TNF-α expression. By pull-down assay, RPL9 and RPS5 were confirmed to interact with AGEs. When AGEs coexisted with LPS, HMGB1, plus RPL9 or RPS5, the reducing effect of TNF-α expression by these cationic ribosomal proteins was shown to be abrogated. The results suggest that cationic ribosomal proteins have a regulatory role in the pro-inflammatory response induced by LPS+HMGB1, and in the pathophysiological condition of accumulating AGEs, this regulatory effect is abolished, which exacerbates inflammation.
Collapse
Affiliation(s)
- Masahiro Watanabe
- Department of Pharmacology, School of Pharmacy, Shujitsu University, Okayama, Japan
| | - Takao Toyomura
- Department of Pharmacology, School of Pharmacy, Shujitsu University, Okayama, Japan
| | - Hidenori Wake
- Department of Pharmacology, Faculty of Medicine, Kindai University, Osaka-Sayama, Japan
| | - Takashi Nishinaka
- Department of Pharmacology, Faculty of Medicine, Kindai University, Osaka-Sayama, Japan
| | - Omer Faruk Hatipoglu
- Department of Pharmacology, Faculty of Medicine, Kindai University, Osaka-Sayama, Japan
| | - Hideo Takahashi
- Department of Pharmacology, Faculty of Medicine, Kindai University, Osaka-Sayama, Japan
| | - Masahiro Nishibori
- Department of Translational Research and Drug Development, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Shuji Mori
- Department of Pharmacology, School of Pharmacy, Shujitsu University, Okayama, Japan
| |
Collapse
|
3
|
Wang Z, Xu J, Zeng X, Du Q, Lan H, Zhang J, Pan D, Tu M. Recent Advances on Antimicrobial Peptides from Milk: Molecular Properties, Mechanisms, and Applications. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:80-93. [PMID: 38152984 DOI: 10.1021/acs.jafc.3c07217] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Traditional antibiotics are facing a tremendous challenge due to increased antimicrobial resistance; hence, there is an urgent need to find novel antibiotic alternatives. Milk protein-derived antimicrobial peptides (AMPs) are currently attracting substantial attention considering that they showcase an extensive spectrum of antimicrobial activities, with slower development of antimicrobial resistance and safety of raw materials. This review summarizes the molecular properties, and activity mechanisms and highlights the applications and limitations of AMPs derived from milk proteins comprehensively. Also the analytical technologies, especially bioinformatics methodologies, applied in the process of screening, identification, and mechanism illustration of AMPs were underlined. This review will give some ideas for further research and broadening of the applications of milk protein-derived AMPs in the food field.
Collapse
Affiliation(s)
- Zhicheng Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China
- Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
| | - Jue Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China
- Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
| | - Xiaoqun Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China
- Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
| | - Qiwei Du
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China
- Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
| | - Hangzhen Lan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China
- Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
| | - Jianming Zhang
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310016, China
| | - Daodong Pan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China
- Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
| | - Maolin Tu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China
- Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
| |
Collapse
|
4
|
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
|
5
|
Ostrówka M, Duda-Madej A, Pietluch F, Mackiewicz P, Gagat P. Testing Antimicrobial Properties of Human Lactoferrin-Derived Fragments. Int J Mol Sci 2023; 24:10529. [PMID: 37445717 DOI: 10.3390/ijms241310529] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Lactoferrin, an iron-binding glycoprotein, plays a significant role in the innate immune system, with antibacterial, antivirial, antifungal, anticancer, antioxidant and immunomodulatory functions reported. It is worth emphasizing that not only the whole protein but also its derived fragments possess antimicrobial peptide (AMP) activity. Using AmpGram, a top-performing AMP classifier, we generated three novel human lactoferrin (hLF) fragments: hLF 397-412, hLF 448-464 and hLF 668-683, predicted with high probability as AMPs. For comparative studies, we included hLF 1-11, previously confirmed to kill some bacteria. With the four peptides, we treated three Gram-negative and three Gram-positive bacterial strains. Our results indicate that none of the three new lactoferrin fragments have antimicrobial properties for the bacteria tested, but hLF 1-11 was lethal against Pseudomonas aeruginosa. The addition of serine protease inhibitors with the hLF fragments did not enhance their activity, except for hLF 1-11 against P. aeruginosa, which MIC dropped from 128 to 64 µg/mL. Furthermore, we investigated the impact of EDTA with/without serine protease inhibitors and the hLF peptides on selected bacteria. We stress the importance of reporting non-AMP sequences for the development of next-generation AMP prediction models, which suffer from the lack of experimentally validated negative dataset for training and benchmarking.
Collapse
Affiliation(s)
- Michał Ostrówka
- Faculty of Biotechnology, University of Wrocław, Fryderyka Joliot-Curie 14a, 50-137 Wrocław, Poland
| | - Anna Duda-Madej
- Department of Microbiology, Faculty of Medicine, Wrocław Medical University, Chałubińskiego 4, 50-368 Wrocław, Poland
| | - Filip Pietluch
- Faculty of Biotechnology, University of Wrocław, Fryderyka Joliot-Curie 14a, 50-137 Wrocław, Poland
| | - Paweł Mackiewicz
- Faculty of Biotechnology, University of Wrocław, Fryderyka Joliot-Curie 14a, 50-137 Wrocław, Poland
| | - Przemysław Gagat
- Faculty of Biotechnology, University of Wrocław, Fryderyka Joliot-Curie 14a, 50-137 Wrocław, Poland
| |
Collapse
|
6
|
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
|
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
|
Yang H, Wang L, Yuan L, Du H, Pan B, Lu K. Antimicrobial Peptides with Rigid Linkers against Gram-Negative Bacteria by Targeting Lipopolysaccharide. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:15903-15916. [PMID: 36511360 DOI: 10.1021/acs.jafc.2c05921] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
A series of hybrid peptides were designed by connecting an antimicrobial peptide Ce(1-8) with a lipopolysaccharide (LPS)-targeting peptide Lf(28-34) via different linkers. Antimicrobial experimental results indicated that linkers play an essential role in the anti-Gram-negative bacterial activity of the hybrid peptides. Among these hybrid peptides, peptide CL5 with dipeptide rigid linker LP exhibited excellent activity and selectivity against Gram-negative bacteria. The minimum inhibitory concentrations of CL5 against the tested Gram-negative bacteria were 4-32 μM, while the toxicity toward HEK-293 cells was relatively low. It was found that the interactions of the peptides with LPS were crucial for peptide activity against Gram-negative bacteria. Antimicrobial mechanistic studies showed that peptide CL5 contributed to the death of Gram-negative bacterial cells by disrupting the integrity of the bacterial membranes. This study revealed the importance of linker selection in the design of hybrid peptides and provides the basis for the further development of antimicrobial peptides.
Collapse
Affiliation(s)
- Hongyan Yang
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Lan Wang
- School of Chemical Engineering and Food Science, Zhengzhou University of Technology, Zhengzhou 450044, China
| | - Libo Yuan
- School of Chemistry and Chemical Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Heng Du
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Boyuan Pan
- School of Chemistry and Chemical Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Kui Lu
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, China
- School of Chemical Engineering and Food Science, Zhengzhou University of Technology, Zhengzhou 450044, China
| |
Collapse
|
9
|
Rivera-Jiménez J, Berraquero-García C, Pérez-Gálvez R, García-Moreno PJ, Espejo-Carpio FJ, Guadix A, Guadix EM. Peptides and protein hydrolysates exhibiting anti-inflammatory activity: sources, structural features and modulation mechanisms. Food Funct 2022; 13:12510-12540. [PMID: 36420754 DOI: 10.1039/d2fo02223k] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inflammation is the response of the immune system to harmful stimuli such as tissue injury, infection or toxic chemicals, which has the aim of eliminating irritants or pathogenic microorganisms and enhancing tissue repair. Uncontrolled long-lasting acute inflammation can gradually progress to chronic, causing a variety of chronic inflammatory diseases that are usually treated with anti-inflammatory drugs, but most of them are inadequate to control chronic responses and are also associated with adverse side effects. Thus, many efforts are being directed to develop alternative and more selective anti-inflammatory therapies from natural products. One main field of interest is the obtaining of bioactive peptides exhibiting anti-inflammatory activity from sustainable protein sources like edible insects or agroindustry and fishing by-products. This work highlighted the structure-activity relationship of anti-inflammatory peptides. Small peptides with molecular weight under 1 kDa and amino acid chain length between 2 to 20 residues are generally the most active because of the higher probability to be absorbed in the intestine and penetrate into cells when compared with the larger size peptides. The presence of hydrophobic (Val, Ile, Pro) and positively charged (His, Arg, Lys) amino acids is another common occurrence for anti-inflammatory peptides. Interestingly, a high percentage (77%) of these bioactive peptides can be found in alternative sustainable protein sources such as Tenebrio molitor or sunflower, apart from its original protein source. However, not all of these peptides with anti-inflammatory potential in vitro achieve good scores by the in silico bioactivity predictors studied. Therefore, it is essential to implement current bioinformatics tools, in order to complement in vitro experiments with prior prediction of potential bioactive peptides.
Collapse
Affiliation(s)
- Julia Rivera-Jiménez
- Department of Chemical Engineering, University of Granada, 18071, Granada, Spain.
| | | | - Raúl Pérez-Gálvez
- Department of Chemical Engineering, University of Granada, 18071, Granada, Spain.
| | | | | | - Antonio Guadix
- Department of Chemical Engineering, University of Granada, 18071, Granada, Spain.
| | - Emilia M Guadix
- Department of Chemical Engineering, University of Granada, 18071, Granada, Spain.
| |
Collapse
|
10
|
Martínez-García JJ, Canizalez-Roman A, Angulo-Zamudio UA, Velazquez-Roman J, Flores-Villaseñor H, Valdez-Flores MA, Rios-Burgueño E, Moran-Portela D, León-Sicairos N. Lactoferrin and Metoprolol Supplementation Increase Mouse Survival in an Experimental LPS-Induced Sepsis Model. Int J Pept Res Ther 2022; 28:141. [DOI: 10.1007/s10989-022-10447-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2022] [Indexed: 11/27/2022]
|
11
|
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
|
12
|
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
|
13
|
The milk-derived lactoferrin inhibits V-ATPase activity by targeting its V1 domain. Int J Biol Macromol 2021; 186:54-70. [PMID: 34237360 DOI: 10.1016/j.ijbiomac.2021.06.200] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/20/2021] [Accepted: 06/29/2021] [Indexed: 11/20/2022]
Abstract
Lactoferrin (Lf), a bioactive milk protein, exhibits strong anticancer and antifungal activities. The search for Lf targets and mechanisms of action is of utmost importance to enhance its effective applications. A common feature among Lf-treated cancer and fungal cells is the inhibition of a proton pump called V-ATPase. Lf-driven V-ATPase inhibition leads to cytosolic acidification, ultimately causing cell death of cancer and fungal cells. Given that a detailed elucidation of how Lf and V-ATPase interact is still missing, herein we aimed to fill this gap by employing a five-stage computational approach. Molecular dynamics simulations of both proteins were performed to obtain a robust sampling of their conformational landscape, followed by clustering, which allowed retrieving representative structures, to then perform protein-protein docking. Subsequently, molecular dynamics simulations of the docked complexes and free binding energy calculations were carried out to evaluate the dynamic binding process and build a final ranking based on the binding affinities. Detailed atomist analysis of the top ranked complexes clearly indicates that Lf binds to the V1 cytosolic domain of V-ATPase. Particularly, our data suggest that Lf binds to the interfaces between A/B subunits, where the ATP hydrolysis occurs, thus inhibiting this process. The free energy decomposition analysis further identified key binding residues that will certainly aid in the rational design of follow-up experimental studies, hence bridging computational and experimental biochemistry.
Collapse
|
14
|
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
|
15
|
Almehdar HA, El-Baky NA, Alhaider AA, Almuhaideb SA, Alhaider AA, Albiheyri RS, Uversky VN, Redwan EM. Bacteriostatic and Bactericidal Activities of Camel Lactoferrins Against Salmonella enterica Serovar Typhi. Probiotics Antimicrob Proteins 2021; 12:18-31. [PMID: 30723884 DOI: 10.1007/s12602-019-9520-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lactoferrin is an iron-binding glycoprotein present in various secretions (e.g., milk, tears, saliva, pancreatic juice), which performs multiple functions, with one of them being the antimicrobial defense. Purified camel lactoferrins (cLfs) from different Saudi camel clans, as well as human and bovine lactoferrins (hLf or bLf) were tested as antimicrobial agents against Salmonella enterica serovar Typhi (S. Typhi). All cLfs showed superior antibacterial potentials relative to hLf or bLf, while there was no noticeable difference in the antimicrobial capabilities between the cLfs from different camel clans. We observed synergy between the inhibitory activities of Lfs and antibiotics against bacterial growth. Expression of numerous bacterial proteins was affected by the treatment with Lf and its combinations, giving insight into the molecular mechanisms of the Lf action. Furthermore, several bacterial proteins were shown to interact with cLf-biotin. Scanning and transmission electron microscopy revealed the presence of obvious extracellular and intracellular changes after S. Typhi treatment by antibiotic (carbenicillin) or cLf alone, and in combination. The effects of antibiotics and Lf were synergistic, supporting the potential of the use of Lf-antibiotic combinations.
Collapse
Affiliation(s)
- Hussein A Almehdar
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, P.O. Box 80203, Jeddah, Saudi Arabia
| | - Nawal Abd El-Baky
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, 21934, Egypt
| | - Abdulqader A Alhaider
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, P.O. Box 80203, Jeddah, Saudi Arabia
| | | | - Abdullah A Alhaider
- Medical school, King Saud bin Abdulaziz University for health Science, Riyadh, Saudi Arabia
| | - Raed S Albiheyri
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, P.O. Box 80203, Jeddah, Saudi Arabia
| | - Vladimir N Uversky
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, P.O. Box 80203, Jeddah, Saudi Arabia.
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Moscow region, 142290, Russia.
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, P.O. Box 80203, Jeddah, Saudi Arabia.
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, 21934, Egypt.
| |
Collapse
|
16
|
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
|
17
|
El-Fakharany EM. Nanoformulation of lactoferrin potentiates its activity and enhances novel biotechnological applications. Int J Biol Macromol 2020; 165:970-984. [PMID: 33011258 DOI: 10.1016/j.ijbiomac.2020.09.235] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/08/2023]
|
18
|
Icriverzi M, Dinca V, Moisei M, Evans RW, Trif M, Roseanu A. Lactoferrin in Bone Tissue Regeneration. Curr Med Chem 2020; 27:838-853. [PMID: 31258057 DOI: 10.2174/0929867326666190503121546] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 11/15/2018] [Accepted: 12/13/2018] [Indexed: 11/22/2022]
Abstract
Among the multiple properties exhibited by lactoferrin (Lf), its involvement in bone regeneration processes is of great interest at the present time. A series of in vitro and in vivo studies have revealed the ability of Lf to promote survival, proliferation and differentiation of osteoblast cells and to inhibit bone resorption mediated by osteoclasts. Although the mechanism underlying the action of Lf in bone cells is still not fully elucidated, it has been shown that its mode of action leading to the survival of osteoblasts is complemented by its mitogenic effect. Activation of several signalling pathways and gene expression, in an LRPdependent or independent manner, has been identified. Unlike the effects on osteoblasts, the action on osteoclasts is different, with Lf leading to a total arrest of osteoclastogenesis. Due to the positive effect of Lf on osteoblasts, the potential use of Lf alone or in combination with different biologically active compounds in bone tissue regeneration and the treatment of bone diseases is of great interest. Since the bioavailability of Lf in vivo is poor, a nanotechnology- based strategy to improve the biological properties of Lf was developed. The investigated formulations include incorporation of Lf into collagen membranes, gelatin hydrogel, liposomes, loading onto nanofibers, porous microspheres, or coating onto silica/titan based implants. Lf has also been coupled with other biologically active compounds such as biomimetic hydroxyapatite, in order to improve the efficacy of biomaterials used in the regulation of bone homeostasis. This review aims to provide an up-to-date review of research on the involvement of Lf in bone growth and healing and on its use as a potential therapeutic factor in bone tissue regeneration.
Collapse
Affiliation(s)
- Madalina Icriverzi
- Ligand-Receptor Interaction Department, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania.,University of Bucharest, Faculty of Biology, Bucharest, Romania
| | - Valentina Dinca
- National Institute for Laser, Plasma and Radiation Physics, Magurele RO-077125, Romania
| | - Magdalena Moisei
- Ligand-Receptor Interaction Department, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Robert W Evans
- Brunel University, School of Engineering and Design, London, United Kingdom
| | - Mihaela Trif
- Ligand-Receptor Interaction Department, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Anca Roseanu
- Ligand-Receptor Interaction Department, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| |
Collapse
|
19
|
Understanding the Elements of Maternal Protection from Systemic Bacterial Infections during Early Life. Nutrients 2020; 12:nu12041045. [PMID: 32290170 PMCID: PMC7230816 DOI: 10.3390/nu12041045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023] Open
Abstract
Late-onset sepsis (LOS) and other systemic bloodstream infections are notable causes of neonatal mortality, particularly in prematurely born very low birth weight infants. Breastfeeding in early life has numerous health benefits, impacting the health of the newborn in both the short-term and in the long-term. Though the known benefits of an exclusive mother's own milk diet in early life have been well recognized and described, it is less understood how breastfed infants enjoy a potential reduction in risk of LOS and other systemic infections. Here we review how gut residing pathogens within the intestinal microbiota of infants can cause a subset of sepsis cases and the components of breastmilk that may prevent the dissemination of pathogens from the intestine.
Collapse
|
20
|
Synergistic Killing of Pathogenic Escherichia coli Using Camel Lactoferrin from Different Saudi Camel Clans and Various Antibiotics. Protein J 2020; 38:479-496. [PMID: 30963371 DOI: 10.1007/s10930-019-09828-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Current study aimed to analyze the synergistic killing of pathogenic Escherichia coli using camel lactoferrin from different Saudi camel clans and various antibiotics. Methods: using multiple microbiological and protein analysis techniques, the results were shown that the purified camel lactoferrins (cLfs) from different Saudi camel have strong antimicrobial potentials against two strains of E. coli. Although all cLfs were superior relative to human or bovine lactoferrins (hLf or bLf), there was no noticeable difference in the antimicrobial potentials of cLfs from different camel clans. The effects of antibiotics and cLfs were synergistic, indicating the superiority of using cLf-antibiotic combinations against E. coli growth. Since these combinations possessed distinguished synergy profiles, it is likely that they can be used to enhance the low efficacy of antibiotics, as well as to control the problems associated with bacterial resistance. Furthermore, these combinations can reduce the cost of cure of bacterial infections, especially in the developing countries. The analysis of the molecular mechanisms of lactoferrin action revealed that expression of several E. coli proteins was affected by the treatment with these antibacterial factors. Several proteins of different molecular weights interacting with cLf-biotin were found. Scanning and transmission electron microscopy analysis revealed the presence of noticeable morphological changes associated with the treatment of E. coli strains by antibiotic carbenicillin or cLf alone, and in combination. Camel lactoferrin has superior potential killing of E. coli over bovine and human lactoferrin, and this potential can be further synergistically enhanced of cLF is combined with antibiotics.
Collapse
|
21
|
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
|
22
|
Alhalwani AY, Davey RL, Kaul N, Barbee SA, Alex Huffman J. Modification of lactoferrin by peroxynitrite reduces its antibacterial activity and changes protein structure. Proteins 2019; 88:166-174. [PMID: 31295370 DOI: 10.1002/prot.25782] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/19/2019] [Accepted: 07/06/2019] [Indexed: 01/02/2023]
Abstract
Lactoferrin (LF) is a multifunctional protein that plays important physiological roles as one of the most concentrated proteins in many human and other mammalian fluids and tissues. In particular, LF provides antibacterial properties to human milk, saliva, and tear fluid. LF also protects against stress-induced lipid peroxidation at inflammation sites through its iron-binding ability. Previous studies have shown that LF can be efficiently nitrated via biologically relevant mediators such as peroxynitrite (ONOO- ), which are also present at high intracellular concentrations during inflammation and nitrosative stress. Here, we examine changes in antibacterial properties and structure of LF following ONOO- treatment. The reaction induces nitration of tyrosine and tryptophan residues, which are commonly used as biomarker molecules for several diseases. Treatment with ONOO- at a 10/1 M ratio of ONOO- to tyrosine inhibited all antibacterial activity exhibited by native LF. Secondary structural changes in LF were assessed using circular dichroism spectroscopy. Nitration products with and without the addition of Fe3+ show significant reduction in alpha-helical properties, suggesting partial protein unfolding. Iron-binding capacity of LF was also reduced after treatment with ONOO- , suggesting a decreased ability of LF to protect against cellular damage. LC-MS/MS spectrometry was used to identify LF peptide fragments nitrated by ONOO- , including tyrosine residue Y92 located in the iron-binding region. These results suggest that posttranslational modification of LF by ONOO- could be an important pathway to exacerbate infection, for example, in inflamed tissues and to reduce the ability of LF to act as an immune responder and decrease oxidative damage.
Collapse
Affiliation(s)
- Amani Y Alhalwani
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado
| | - Rachel L Davey
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado
| | - Navneeta Kaul
- Department of Biological Sciences, University of Denver, Denver, Colorado
| | - Scott A Barbee
- Department of Biological Sciences, University of Denver, Denver, Colorado
| | - J Alex Huffman
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado
| |
Collapse
|
23
|
Assessment of bioactivities of the human milk lactoferrin–osteopontin complex in vitro. J Nutr Biochem 2019; 69:10-18. [DOI: 10.1016/j.jnutbio.2019.03.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/28/2019] [Accepted: 03/14/2019] [Indexed: 02/02/2023]
|
24
|
Polysialic Acid Modulates the Binding of External Lactoferrin in Neutrophil Extracellular Traps. BIOLOGY 2019; 8:biology8020020. [PMID: 30925725 PMCID: PMC6627751 DOI: 10.3390/biology8020020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/28/2019] [Accepted: 03/21/2019] [Indexed: 02/07/2023]
Abstract
Neutrophil extracellular traps (NETs) are formed by neutrophils during inflammation. Among other things, these DNA constructs consist of antimicrobial proteins such as lactoferrin and histones. With these properties, NETs capture and destroy invading microorganisms. The carbohydrate polysialic acid (polySia) interacts with both lactoferrin and histones. Previous experiments demonstrated that, in humans, lactoferrin inhibits the release of NET and that this effect is supported by polySia. In this study, we examined the interplay of lactoferrin and polySia in already-formed NETs from bovine neutrophils. The binding of polySia was considered to occur at the lactoferricin (LFcin)-containing domain of lactoferrin. The interaction with the peptide LFcin was studied in more detail using groups of defined polySia chain lengths, which suggested a chain-length-dependent interaction mechanism with LFcin. The LFcin domain of lactoferrin was found to interact with DNA. Therefore, the possibility that polySia influences the integration of lactoferrin into the DNA-structures of NETs was tested by isolating bovine neutrophils and inducing NETosis. Experiments with NET fibers saturated with lactoferrin demonstrated that polySia initiates the incorporation of external lactoferrin in already-loaded NETs. Thus, polySia may modulate the constituents of NET.
Collapse
|
25
|
Vargas-Casanova Y, Rodríguez-Mayor AV, Cardenas KJ, Leal-Castro AL, Muñoz-Molina LC, Fierro-Medina R, Rivera-Monroy ZJ, García-Castañeda JE. Synergistic bactericide and antibiotic effects of dimeric, tetrameric, or palindromic peptides containing the RWQWR motif against Gram-positive and Gram-negative strains. RSC Adv 2019; 9:7239-7245. [PMID: 35519960 PMCID: PMC9061098 DOI: 10.1039/c9ra00708c] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 02/21/2019] [Indexed: 01/12/2023] Open
Abstract
Dimeric and tetrameric peptides derived from LfcinB (20-25): RRWQWR, LfcinB (20-30): RRWQWRMKKLG, LfcinB (17-31): FKARRWQWRMKKLGA, or the palindromic sequence LfcinB (21-25)Pal: RWQWRWQWR were obtained by means of the SPPS-Fmoc/tBu methodology. The antibacterial activity of these molecules was evaluated against Escherichia coli (ATCC 25922 and ATCC 11775), Staphylococcus aureus (ATCC 25923), Enterococcus faecalis (ATCC 29212), and Pseudomonas aeruginosa (ATCC 27853). The dimer LfcinB (20-25)2: (RRWQWR)2K-Ahx, the tetramer LfcinB (20-25)4: (RRWQWR)4K2-Ahx2-C2, and the palindromic sequence LfcinB (21-25)Pal exhibited the highest antibacterial activity against the tested bacterial strains. In all cases, the antibacterial activity was dependent on peptide concentration. The polyvalent molecules LfcinB (20-25)2 and LfcinB (20-25)4 exhibited bacteriostatic and bactericidal activity against E. coli, P. aeruginosa, and S. aureus strains; additionally, this dimer and this tetramer combined with ciprofloxacin exhibited a synergistic antibacterial effect against E. coli ATCC 25922 and P. aeruginosa, respectively. Furthermore, the peptides LfcinB (20-30)4, LfcinB (20-25)4, and LfcinB (21-25)Pal combined with vancomycin exhibited a synergistic antibacterial effect against S. aureus and E. faecalis, respectively. This study showed that polyvalent peptides derived from LfcinB exhibit significant antibacterial activity, suggesting that these peptides could have a therapeutic application. Furthermore, our results suggest that polyvalent peptide synthesis could be considered as an innovative and viable strategy for obtaining promising antimicrobial molecules.
Collapse
Affiliation(s)
- Yerly Vargas-Casanova
- Biotechnology Institute, Universidad Nacional de Colombia Carrera 45 No 26-85 Bogotá 11321 Colombia
| | | | - Karen Johanna Cardenas
- Pharmacy Department, Universidad Nacional de Colombia Carrera 45 No 26-85, Building 450, Office 213 Bogotá 11321 Colombia
| | - Aura Lucía Leal-Castro
- Medicine Faculty, Universidad Nacional de Colombia Carrera 45 No 26-85, Building 450, Office 213 Bogotá 11321 Colombia
| | | | - Ricardo Fierro-Medina
- Chemistry Department, Universidad Nacional de Colombia Carrera 45 No 26-85, Building 450, Office 213 Bogotá 11321 Colombia
| | - Zuly Jenny Rivera-Monroy
- Chemistry Department, Universidad Nacional de Colombia Carrera 45 No 26-85, Building 450, Office 213 Bogotá 11321 Colombia
| | | |
Collapse
|
26
|
Schüller SS, Kramer BW, Villamor E, Spittler A, Berger A, Levy O. Immunomodulation to Prevent or Treat Neonatal Sepsis: Past, Present, and Future. Front Pediatr 2018; 6:199. [PMID: 30073156 PMCID: PMC6060673 DOI: 10.3389/fped.2018.00199] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
Despite continued advances in neonatal medicine, sepsis remains a leading cause of death worldwide in neonatal intensive care units. The clinical presentation of sepsis in neonates varies markedly from that in older children and adults, and distinct acute inflammatory responses results in age-specific inflammatory and protective immune response to infection. This review first provides an overview of the neonatal immune system, then covers current mainstream, and experimental preventive and adjuvant therapies in neonatal sepsis. We also discuss how the distinct physiology of the perinatal period shapes early life immune responses and review strategies to reduce neonatal sepsis-related morbidity and mortality. A summary of studies that characterize immune ontogeny and neonatal sepsis is presented, followed by discussion of clinical trials assessing interventions such as breast milk, lactoferrin, probiotics, and pentoxifylline. Finally, we critically appraise future treatment options such as stem cell therapy, other antimicrobial protein and peptides, and targeting of pattern recognition receptors in an effort to prevent and/or treat sepsis in this highly vulnerable neonatal population.
Collapse
Affiliation(s)
- Simone S. Schüller
- Division of Neonatology, Pediatric Intensive Care & Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Precision Vaccines Program, Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Boris W. Kramer
- Department of Pediatrics, Maastricht University Medical Centre (MUMC+), Maastricht, Netherlands
- School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Centre (MUMC+), Maastricht, Netherlands
- School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Andreas Spittler
- Department of Surgery, Research Labs & Core Facility Flow Cytometry, Medical University of Vienna, Vienna, Austria
| | - Angelika Berger
- Division of Neonatology, Pediatric Intensive Care & Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Boston, MA, United States
| |
Collapse
|
27
|
Bovine Lactoferrin Modulates Dendritic Cell Differentiation and Function. Nutrients 2018; 10:nu10070848. [PMID: 29966271 PMCID: PMC6073808 DOI: 10.3390/nu10070848] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/19/2018] [Accepted: 06/26/2018] [Indexed: 11/16/2022] Open
Abstract
Lactoferrin is an abundant glycoprotein in bovine milk that has immunomodulatory effects on human cells. Bovine lactoferrin (LF) binds lipopolysaccharides (LPS) with high affinity and is postulated to act via TLR4-dependent and -independent mechanisms. It has been shown that LF modulates differentiation of human monocytes into tolerogenic dendritic cells. However, in a previous study, we showed that LPS also mediates differentiation into tolerogenic dendritic cells (DC). Since LF binds LPS with high affinity, it remains to be investigated whether LF or LPS is mediating these effects. We, therefore, further investigated the LPS-independent effect of LF on differentiation of human monocytes into dendritic cells (DC). Human monocytes were isolated by magnetic cell sorting from freshly isolated PBMCs and cultured for six days in the presence of IL-4 and GM-CSF with or without LF or proteinase K treated LF to generate DC. These immature DC were stimulated for 48 h with LPS or Poly I:C + R848. Cell surface marker expression and cytokine production were measured by flow cytometry. DC differentiated in the presence of LF produced higher IL-6 and IL-8 levels during differentiation and showed a lower expression of CD1a and HLA-DR. These LFDCs showed to be hyporesponsive towards TLR ligands as shown by their semi-mature phenotype and reduced cytokine production. The effect of LF was abrogated by proteinase K treatment, showing that the functional effects of LF were not mediated by LPS contamination. Thus, LF alters DC differentiation and dampens responsiveness towards TLR ligands. This study indicates that LF can play a role in immune homeostasis in the human GI tract.
Collapse
|
28
|
Guha S, Majumder K. Structural-features of food-derived bioactive peptides with anti-inflammatory activity: A brief review. J Food Biochem 2018; 43:e12531. [PMID: 31353488 DOI: 10.1111/jfbc.12531] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 02/01/2018] [Accepted: 02/02/2018] [Indexed: 02/06/2023]
Abstract
Chronic inflammatory conditions such as obesity, type-2 diabetes, and cardiovascular diseases are the leading causes of mortality worldwide. Hence, much research interest in bioactive peptides has been stimulated due to lack of potent pharmacological interventions. Although many such peptides have been identified from food proteins, insufficient information is available on their structure-function relationship. Presence of hydrophobic and positively charged amino acids is a common occurrence for the peptides with anti-inflammatory properties. However, inconsistent findings have also been reported. Most of the food-derived peptides exhibited their anti-inflammatory activities primarily by inhibiting signaling components of either NF-κB or MAPK pathway, which are the two major pathways involved in chronic inflammation following uncontrolled signal activation. This review highlighted the structural requirements of the peptides to exhibit anti-inflammatory activity based on the current knowledge about food-derived anti-inflammatory peptides and their underlying molecular mechanisms of action. PRACTICAL APPLICATIONS: While research in the food-derived bioactive peptide is gaining momentum, but the ability to translate these new findings into the commercial product such as nutraceuticals and functional foods, remains delayed. The most prominent reasons for this delay are the lack of detailed research on, (i) the structure-function relationship of the peptide and the underlying molecular mechanisms of these bioactive peptides, and (ii) the interaction of these peptides with different cellular elements in the disease pathophysiology. This review gives an insight into the structure-activity relationship of bioactive peptides involved in anti-inflammatory responses. The information provided here would be highly beneficial to describe the possible anti-inflammatory activity of any newly identified peptides from different food sources.
Collapse
Affiliation(s)
- Snigdha Guha
- Department of Food Science and Technology, University of Nebraska, Lincoln, Nebraska
| | - Kaustav Majumder
- Department of Food Science and Technology, University of Nebraska, Lincoln, Nebraska
| |
Collapse
|
29
|
Inhibitory Effect of Bovine Lactoferrin on Catechol-O-Methyltransferase. Molecules 2017; 22:molecules22081373. [PMID: 28825621 PMCID: PMC6152271 DOI: 10.3390/molecules22081373] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/08/2017] [Accepted: 08/16/2017] [Indexed: 12/22/2022] Open
Abstract
Lactoferrin (LF) is a well-known multifunctional protein. In this study, we report the inhibitory potency of bovine LF (bLF) on catechol-O-methyltransferase (COMT), which catalyzes methylation of catechol substrates. We found that bLF binds to and inhibits COMT using its N-terminal region. An N-terminal peptide fragment obtained from bLF by trypsin digestion showed a higher inhibitory activity than intact bLF. A synthetic fragment of the bLF N-terminal residues 6-50, with two pairs of disulfide bonds, also showed higher inhibitory activity than intact bLF. Enzyme kinetic studies proved that bLF did not compete with S-adenosylmethionine (the methyl donor substrate) as well as methyl acceptor substrates such as dihydroxybenzoic acid, (-)-epicatechin, norepinephrine, or l-3,4-dihydroxyphenylalanine. The inhibitory potency of bLF decreased against a COMT preparation pretreated with dithiothreitol, suggesting that the oxidation status of COMT is relevant to interaction with bLF. We further confirmed that COMT activity in the cell extracts form Caco-2 and HepG2 cells was inhibited by bLF and by the synthesized fragment. Enzyme kinetic study indicated that bLF functions as a non-competitive inhibitor by binding to an allosteric surface of COMT.
Collapse
|
30
|
Hu L, Hu X, Long K, Gao C, Dong HL, Zhong Q, Gao XM, Gong FY. Extraordinarily potent proinflammatory properties of lactoferrin-containing immunocomplexes against human monocytes and macrophages. Sci Rep 2017; 7:4230. [PMID: 28652573 PMCID: PMC5484712 DOI: 10.1038/s41598-017-04275-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 05/14/2017] [Indexed: 12/25/2022] Open
Abstract
Lactoferrin (LTF), an important first line defense molecule against infection, is a common target for humoral autoimmune reactions in humans. Since LTF is a multifunctional protein capable of activating innate immune cells via various surface receptors, we hypothesized that LTF-containing immune complexes (ICs) (LTF-ICs), likely formed in patients with high titer anti-LTF autoantibodies, could possess unique monocyte/macrophage-activating properties compared with other ICs. ELISA analysis on serum samples from rheumatoid arthritis (RA) patients (n = 80) and healthy controls (n = 35) for anti-LTF autoantibodies confirmed a positive correlation between circulating LTF-specific IgG and RA. ICs between human LTF and LTF-specific IgG purified from patient sera or immunized rabbits and mice, but not control ICs, LTF or Abs alone, elicited strong production of TNF-α and IL-1β by freshly fractionated human peripheral blood monocytes and monocytes-derived macrophages. Furthermore, LTF-ICs utilized both membrane-anchored CD14 and CD32a (FcγRIIa) to trigger monocyte activation in an internalization-, Toll-like receptor (TLR)4- and TLR9-dependent manner, and also that LTF-IC-induced cytokine production was blocked by specific inhibitors of caspase-1, NF-κB and MAPK. These results uncover a possible pathway for LTF-ICs perpetuating local inflammation and contributing to the pathogenesis of autoimmune diseases by triggering activation of infiltrating monocytes or tissue macrophages in vivo.
Collapse
Affiliation(s)
- Lulu Hu
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xiaomin Hu
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Kai Long
- Department of Immunology, Peking University Health Science Center, Beijing, China.,Department of Physiology, Jiujiang College, Jiangxi Province, China
| | - Chenhui Gao
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Hong-Liang Dong
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Qiao Zhong
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xiao-Ming Gao
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China.
| | - Fang-Yuan Gong
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
31
|
Huertas NDJ, Monroy ZJR, Medina RF, Castañeda JEG. Antimicrobial Activity of Truncated and Polyvalent Peptides Derived from the FKCRRQWQWRMKKGLA Sequence against Escherichia coli ATCC 25922 and Staphylococcus aureus ATCC 25923. Molecules 2017; 22:molecules22060987. [PMID: 28613262 PMCID: PMC6152618 DOI: 10.3390/molecules22060987] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 06/08/2017] [Accepted: 06/12/2017] [Indexed: 12/29/2022] Open
Abstract
Peptides derived from LfcinB were designed and synthesized, and their antibacterial activity was tested against Escherichia coli ATCC 25922 and Staphylococcus aureus ATCC 25923. Specifically, a peptide library was constructed by systemically removing the flanking residues (N or C-terminal) of Lfcin 17–31 (17FKCRRWQWRMKKLGA31), maintaining in all peptides the 20RRWQWR25 sequence that corresponds to the minimal antimicrobial motif. For this research, also included were (i) a peptide containing an Ala instead of Cys ([Ala19]-LfcinB 17–31) and (ii) polyvalent peptides containing the RRWQWR sequence and a non-natural amino acid (aminocaproic acid). We established that the lineal peptides LfcinB 17–25 and LfcinB 17–26 exhibited the greatest activity against E. coli ATCC 25922 and S. aureus ATCC 25923, respectively. On the other hand, polyvalent peptides, a dimer and a tetramer, exhibited the greatest antibacterial activity, indicating that multiple copies of the sequence increase the activity. Our results suggest that the dimeric and tetrameric sequence forms potentiate the antibacterial activity of lineal sequences that have exhibited moderate antibacterial activity.
Collapse
Affiliation(s)
- Nataly de Jesús Huertas
- Chemistry Department, Universidad Nacional de Colombia, Bogotá Carrera 45 No. 26-85, Building 451, Office 409, Laboratory 334, Bogotá 11321, Colombia.
| | - Zuly Jenny Rivera Monroy
- Chemistry Department, Universidad Nacional de Colombia, Bogotá Carrera 45 No. 26-85, Building 451, Office 409, Laboratory 334, Bogotá 11321, Colombia.
| | - Ricardo Fierro Medina
- Chemistry Department, Universidad Nacional de Colombia, Bogotá Carrera 45 No. 26-85, Building 451, Office 409, Laboratory 334, Bogotá 11321, Colombia.
| | - Javier Eduardo García Castañeda
- Pharmacy Department, Universidad Nacional de Colombia, Bogotá Carrera 45 No. 26-85, Building 450, office 203, Bogotá 11321, Colombia.
| |
Collapse
|
32
|
Tsou YA, Tung YT, Wu TF, Chang GRL, Chen HC, Lin CD, Lai CH, Chen HL, Chen CM. Lactoferrin interacts with SPLUNC1 to attenuate lipopolysaccharide-induced inflammation of human nasal epithelial cells via down-regulated MEK1/2-MAPK signaling. Biochem Cell Biol 2017; 95:394-399. [PMID: 28178421 DOI: 10.1139/bcb-2016-0047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The short palate, lung, and nasal epithelium clone 1 (SPLUNC1) protein is an important innate material in the upper airway, and lactoferrin (LF) aids the innate functions in humans. In this study, a nasal epithelial model was used to investigate how LF modulates SPLUNC1 to reduce the inflammatory process mediated by lipopolysaccharide (LPS). The inflammation of human RPMI-2650 cells was induced with LPS to evaluate SPLUNC1 expression after treating the cells with bovine LF (bLF). The interaction pathway between LF and SPLUNC1 in LPS-induced inflammation was further investigated. Our study reveals that the addition of bLF results in the recovery of SPLUNC1 expression in nasal epithelial cells under LPS-induced inflammation. MAPK is involved in the main pathway for the SPLUNC1 and bLF interaction. Decreased SPLUNC1 function could be recovered by addition of bLF. The MEK1/2-MAPK signaling pathway is crucial for the SPLUNC1 and bLF interaction. Therefore, LF could support SPLUNC1 in the innate immunity recovery process.
Collapse
Affiliation(s)
- Yung-An Tsou
- a Department of Life Sciences, and Agricultural Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan.,b Department of Otolaryngology-Head and Neck Surgery, China Medical University and Hospital, Taichung 40402, Taiwan.,c Graduate Institute of Biomedicine Sciences, China Medical University, Taichung 40402, Taiwan
| | - Yu-Tong Tung
- a Department of Life Sciences, and Agricultural Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan
| | - Tsu-Fang Wu
- c Graduate Institute of Biomedicine Sciences, China Medical University, Taichung 40402, Taiwan
| | - Gary Ro-Lin Chang
- a Department of Life Sciences, and Agricultural Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan
| | - Han-Chien Chen
- a Department of Life Sciences, and Agricultural Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan
| | - Chia-Der Lin
- b Department of Otolaryngology-Head and Neck Surgery, China Medical University and Hospital, Taichung 40402, Taiwan.,c Graduate Institute of Biomedicine Sciences, China Medical University, Taichung 40402, Taiwan
| | - Chih-Ho Lai
- d Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| | - Hsiao-Ling Chen
- e Department of Bioresources, Da-Yeh University, Changhua 51591, Taiwan
| | - Chuan-Mu Chen
- a Department of Life Sciences, and Agricultural Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan.,f Rong-Hsing Translational Medicine Center, iEGG Center, National Chung Hsing University, Taichung 40227, Taiwan
| |
Collapse
|
33
|
Investigation of the enhanced antimicrobial activity of combination dry powder inhaler formulations of lactoferrin. Int J Pharm 2016; 514:399-406. [DOI: 10.1016/j.ijpharm.2016.09.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 09/08/2016] [Accepted: 09/09/2016] [Indexed: 11/23/2022]
|
34
|
Stoleru E, Zaharescu T, Hitruc EG, Vesel A, Ioanid EG, Coroaba A, Safrany A, Pricope G, Lungu M, Schick C, Vasile C. Lactoferrin-Immobilized Surfaces onto Functionalized PLA Assisted by the Gamma-Rays and Nitrogen Plasma to Create Materials with Multifunctional Properties. ACS APPLIED MATERIALS & INTERFACES 2016; 8:31902-31915. [PMID: 27933972 DOI: 10.1021/acsami.6b09069] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Both cold nitrogen radiofrequency plasma and gamma irradiation have been applied to activate and functionalize the polylactic acid (PLA) surface and the subsequent lactoferrin immobilization. Modified films were comparatively characterized with respect to the procedure of activation and also with unmodified sample by water contact angle measurements, mass loss, X-ray photoelectron spectroscopy (XPS), attenuated total reflectance-Fourier transform infrared spectroscopy (ATR-FTIR), atomic force microscopy (AFM), and chemiluminescence measurements. All modified samples exhibit enhanced surface properties mainly those concerning biocompatibility, antimicrobial, and antioxidant properties, and furthermore, they are biodegradable and environmentally friendly. Lactoferrin deposited layer by covalent coupling using carbodiimide chemistry showed a good stability. It was found that the lactoferrin-modified PLA materials present significantly increased oxidative stability. Gamma-irradiated samples and lactoferrin-functionalized samples show higher antioxidant, antimicrobial, and cell proliferation activity than plasma-activated and lactoferrin-functionalized ones. The multifunctional materials thus obtained could find application as biomaterials or as bioactive packaging films.
Collapse
Affiliation(s)
- Elena Stoleru
- "P. Poni" Institute of Macromolecular Chemistry, Physical Chemistry Department, Iasi 700487, Romania
| | - Traian Zaharescu
- National Institute for R&D in Electrical Engineering , Bucharest 030138, Romania
| | - Elena Gabriela Hitruc
- "P. Poni" Institute of Macromolecular Chemistry, Physical Chemistry Department, Iasi 700487, Romania
| | - Alenka Vesel
- Jožef Stefan Institute , Ljubljana 1000, Slovenia
| | - Emil G Ioanid
- "P. Poni" Institute of Macromolecular Chemistry, Physical Chemistry Department, Iasi 700487, Romania
| | - Adina Coroaba
- "P. Poni" Institute of Macromolecular Chemistry, Physical Chemistry Department, Iasi 700487, Romania
| | - Agnes Safrany
- Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna 1400, Austria
| | - Gina Pricope
- Veterinary and Food Safety Laboratory, Food Safety Department, Iasi 700487, Romania
| | - Maria Lungu
- National Institute of Research and Development for Biological Sciences , Bucharest 060031, Romania
| | - Christoph Schick
- Universität Rostock, Institut für Physik , Rostock 18059, Germany
| | - Cornelia Vasile
- "P. Poni" Institute of Macromolecular Chemistry, Physical Chemistry Department, Iasi 700487, Romania
| |
Collapse
|
35
|
Azumi S, Tanamoto KI. Anti-endotoxin properties of a cinnamon bark-derived compound and its effect on the endotoxin shock model. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519990050030401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
An endotoxin inhibitor derived from cinnamon bark was characterized chemically and tested for anti-LPS properties. Chemical analysis suggested that the active center of the inhibitor was in the lipid portion. Upon incubation with LPS molecule, the inhibitor reduced the ability of LPS to induce TNFα and generate nitric oxide from various cells in vitro, and Limulus gelation activity. The lethal toxicity of LPS in galactosamine-sensitized mice and pyrogenicity of LPS in a rabbit model were reduced 1000- and 100-fold by pre-incubation with the inhibitor, respectively. Simultaneous but separate injection of the inhibitor with a lethal dose of LPS also protected the majority of mice. Protection against LPS was seen when the inhibitor was given to mice 1 h before the LPS challenge. Furthermore, the inhibitor significantly suppressed the induction of fever by simultaneous administration with LPS without prior mixing. These results suggest that the inhibitor may be a useful potent blocker of bacterial endotoxin.
Collapse
Affiliation(s)
- Satoko Azumi
- Division of Microbiology, National Institute of Health Sciences, Tokyo, Japan
| | - Ken-ichi Tanamoto
- Division of Microbiology, National Institute of Health Sciences, Tokyo, Japan,
| |
Collapse
|
36
|
Andrä J, Gutsmann T, Garidel P, Brandenburg K. Invited review: Mechanisms of endotoxin neutralization by synthetic cationic compounds. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519060120050201] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A basic challenge in the treatment of septic patients in critical care units is the release of bacterial pathogenicity factors such as lipopolysaccharide (LPS, endotoxin) from the cell envelope of Gram-negative bacteria due to killing by antibiotics. LPS aggregates may interact with serum and membrane proteins such as LBP (lipopolysaccharide-binding protein) and CD14 leading to the observed strong reaction of the immune system. Thus, an effective treatment of patients infected by Gram-negative bacteria must comprise beside bacterial killing the neutralization of endotoxins. Here, data are summarized for synthetic compounds indicating the stepwise development to very effective LPS-neutralizing agents. These data include synthetic peptides, based on the endotoxin-binding domains of natural binding proteins such as lactoferrin, Limulus anti-LPS factor, NK-lysin, and cathelicidins or based on LPS sequestering polyamines. Many of these compounds could be shown to act not only in vitro, but also in vivo (e.g . in animal models of sepsis), and might be useful in future clinical trials and in sepsis therapy.
Collapse
Affiliation(s)
- Jörg Andrä
- Forschungszentrum Borstel, Biophysics Division, Leibniz-Zentrum für Medizin und Biowissenschaften, Borstel, Germany
| | - Thomas Gutsmann
- Forschungszentrum Borstel, Biophysics Division, Leibniz-Zentrum für Medizin und Biowissenschaften, Borstel, Germany
| | - Patrick Garidel
- Institut für Physikalische Chemie, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Klaus Brandenburg
- Forschungszentrum Borstel, Biophysics Division, Leibniz-Zentrum für Medizin und Biowissenschaften, Borstel, Germany,
| |
Collapse
|
37
|
Satoh M, Iwahori T, Sugawara N, Yamazaki M. Liver argininosuccinate synthase binds to bacterial lipopolysaccharides and lipid A and inactivates their biological activities. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519060120010301] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The liver is known to clear and detoxify circulating lipopolysaccharide (LPS). To characterize the molecules involved in this process in the liver, we attempted to purify mouse liver protein(s) that can interact with lipid A, a biologically active portion of LPS. By partially purifying the inactivating activity against a synthetic lipid A analog, we observed the enrichment of a 45-kDa protein in the active fractions. The internal amino acid sequences of the protein were identical with those of argininosuccinate synthase (EC 6.3.4.5). To examine whether argininosuccinate synthase can interact with lipid A, we purified the enzyme from mouse liver and found the co-elevation of the specific enzyme activity and specific lipid A-inactivating activity, indicating that argininosuccinate synthase is the major lipid A-interacting protein in liver. Argininosuccinate synthase also inhibited the biological activities (macrophage activation and Limulus test) of natural lipid A and rough-type LPS but not smooth-type LPS. The enzyme activity was inhibited by lipid A and rough-type LPS and also by smooth-type LPS. Native gel electrophoresis of a mixture of argininosuccinate synthase and LPS and immunoprecipitation of a mixture of argininosuccinate synthase and [3H]-LPS with anti-argininosuccinate synthase antiserum showed that argininosuccinate synthase stably bound lipid A and LPS. These findings, together, indicate that argininosuccinate synthase can effectively bind LPS in the liver.
Collapse
Affiliation(s)
- Motonobu Satoh
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Teikyo University, Sagamiko, Kanagawa, Japan,-u.ac.jp
| | - Tsuguya Iwahori
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Teikyo University, Sagamiko, Kanagawa, Japan
| | - Naoki Sugawara
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Teikyo University, Sagamiko, Kanagawa, Japan
| | - Masatoshi Yamazaki
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Teikyo University, Sagamiko, Kanagawa, Japan
| |
Collapse
|
38
|
Kim WS, Ohashi M, Shimazaki KI. Inhibitory Effects of Synthetic Peptides Containing Bovine Lactoferrin C-lobe Sequence on Bacterial Growth. Korean J Food Sci Anim Resour 2016; 36:452-7. [PMID: 27621684 PMCID: PMC5018503 DOI: 10.5851/kosfa.2016.36.4.452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/15/2016] [Accepted: 06/15/2016] [Indexed: 11/12/2022] Open
Abstract
Lactoferrin is a glycoprotein with various biological effects, with antibacterial activity being one of the first effects reported. This glycoprotein suppresses bacterial growth through bacteriostatic or bactericidal action. It also stimulates the growth of certain kinds of bacteria such as lactic acid bacteria and bifidobacteria. In this study, Asn-Leu-Asn-Arg was selected and chemically synthesized based on the partial sequences of bovine lactoferrin tryptic fragments. Synthetic Asn-Leu-Asn-Arg suppressed the growth of Pseudomonas fluorescens, P. syringae and Escherichia coli. P. fluorescens is a major psychrotrophic bacteria found in raw and pasteurized milk, which decreases milk quality. P. syringae is a harmful infectious bacterium that damages plants. However, synthetic Asn-Leu-Asn-Arg did not inhibit the growth of Lactobacillus acidophilus. It is expected that this synthetic peptide would be the first peptide sequence from the bovine lactoferrin C-lobe that shows antibacterial activity.
Collapse
Affiliation(s)
- Woan-Sub Kim
- Department of Animal Life and Environmental Science, Hankyong National University, Anseong 17579, Korea
| | - Midori Ohashi
- Research Faculty of Agriculture, Hokkaido University, Sapporo 060-8589, Japan
| | - Kei-Ichi Shimazaki
- Research Faculty of Agriculture, Hokkaido University, Sapporo 060-8589, Japan
| |
Collapse
|
39
|
Kim H, Jang JH, Kim SC, Cho JH. Enhancement of the antimicrobial activity and selectivity of GNU7 against Gram-negative bacteria by fusion with LPS-targeting peptide. Peptides 2016; 82:60-66. [PMID: 27242337 DOI: 10.1016/j.peptides.2016.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/25/2016] [Accepted: 05/25/2016] [Indexed: 01/18/2023]
Abstract
Antimicrobial peptides (AMPs) provide a potential source of new antimicrobial therapeutics for the treatment of multidrug-resistant pathogens. To develop Gram-negative selective AMPs that can inhibit the effects of lipopolysaccharide (LPS)-induced sepsis, we added various rationally designed LPS-targeting peptides [amino acids 28-34 of lactoferrin (Lf28-34), amino acids 84-99 of bactericidal/permeability increasing protein (BPI84-99), and de novo peptide (Syn)] to the potent AMP, GNU7 (RLLRPLLQLLKQKLR). Compared to our original starting peptide GNU7, hybrid peptides had an 8- to 32-fold improvement in antimicrobial activity against Gram-negative bacteria, such as Escherichia coli and Salmonella typhimurium. Among them, Syn-GNU7 showed the strongest LPS-binding and -neutralizing activities, thus allowing it to selectively eliminate Gram-negative bacteria from within mixed cultures. Our results suggest that LPS-targeting peptides would be useful to increase the antimicrobial activity and selectivity of other AMPs against Gram-negative bacteria.
Collapse
Affiliation(s)
- Hyun Kim
- Division of Life Science, Gyeongsang National University, Jinju 52828, South Korea
| | - Ju Hye Jang
- Research Institute of Life Science, Gyeongsang National University, Jinju 52828, South Korea
| | - Sun Chang Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, South Korea
| | - Ju Hyun Cho
- Division of Life Science, Gyeongsang National University, Jinju 52828, South Korea; Research Institute of Life Science, Gyeongsang National University, Jinju 52828, South Korea.
| |
Collapse
|
40
|
Lizzi AR, Carnicelli V, Clarkson MM, Nazzicone C, Segatore B, Celenza G, Aschi M, Dolo V, Strom R, Amicosante G. Bovine lactoferrin and its tryptic peptides: Antibacterial activity against different species. APPL BIOCHEM MICRO+ 2016. [DOI: 10.1134/s0003683816040116] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
41
|
Guerville M, Boudry G. Gastrointestinal and hepatic mechanisms limiting entry and dissemination of lipopolysaccharide into the systemic circulation. Am J Physiol Gastrointest Liver Physiol 2016; 311:G1-G15. [PMID: 27151941 DOI: 10.1152/ajpgi.00098.2016] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/30/2016] [Indexed: 01/31/2023]
Abstract
The human microbiota consists of 100 trillion microorganisms that provide important metabolic and biological functions benefiting the host. However, the presence in host plasma of a gut-derived bacteria component, the lipopolysaccharide (LPS), has been identified as a causal or complicating factor in multiple serious diseases such as sepsis and septic shock and, more recently, obesity-associated metabolic disorders. Understanding the precise mechanisms by which gut-derived LPS is transported from the gut lumen to the systemic circulation is crucial to advance our knowledge of LPS-associated diseases and elaborate targeted strategies for their prevention. The aim of this review is to synthetize current knowledge on the host mechanisms limiting the entry and dissemination of LPS into the systemic circulation. To prevent bacterial colonization and penetration, the intestinal epithelium harbors multiple defense mechanisms including the secretion of antimicrobial peptides and mucins as well as detoxification enzymes. Despite this first line of defense, LPS can reach the apical site of intestinal epithelial cells (IECs) and, because of its large size, likely crosses IECs via transcellular transport, either lipid raft- or clathrin-mediated endocytosis or goblet cell-associated passage. However, the precise pathway remains poorly described. Finally, if LPS crosses the gut mucosa, it is directed via the portal vein to the liver, where major detoxification processes occur by deacetylation and excretion through the bile. If this disposal process is not sufficient, LPS enters the systemic circulation, where it is handled by numerous transport proteins that clear it back to the liver for further excretion.
Collapse
Affiliation(s)
| | - Gaëlle Boudry
- INRA UR1341 ADNC, Domaine de la Prise, Saint-Gilles, France
| |
Collapse
|
42
|
Abstract
Lactoferrin is thought to be the most polyvalent protein present in host defense against tissue injuries and infections in vertebrates. Owing to the propensity of its basic N-terminal domain to interact with various microbial and host targets, lactoferrin not only has antimicrobial properties, but also modulates the innate and adaptive immune responses. Lactoferrin may indeed up- and downregulate immune cell activation, migration, and growth. Whereas the immunomodulatory properties of lactoferrin are evidenced from in vivo studies using either lactoferrin-knockout, lactoferrin-overexpressing transgenic models, and dietary lactoferrin, few mechanisms from in vitro studies have been proposed to explain these properties. The best characterized lactoferrin targets are negatively charged molecules. They encompass pro-inflammatory microbial molecules, such as pathogen-associated molecular patterns (eg, lipopolysaccharide), but also host components such as DNA, the glycosaminoglycan chains of proteoglycans, and surface cell receptors. Signaling through these receptors is thought to be the main lever used by lactoferrin to influence immune cells and cytokine-balance-controlling cell activity. This article aims to review our current understanding, though incomplete, of the many ways lactoferrin influences the complex immune machinery and the known and putative mechanisms that may explain its properties.
Collapse
Affiliation(s)
- Dominique Legrand
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France.
| |
Collapse
|
43
|
Barber MF, Kronenberg Z, Yandell M, Elde NC. Antimicrobial Functions of Lactoferrin Promote Genetic Conflicts in Ancient Primates and Modern Humans. PLoS Genet 2016; 12:e1006063. [PMID: 27203426 PMCID: PMC4874600 DOI: 10.1371/journal.pgen.1006063] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 04/27/2016] [Indexed: 01/10/2023] Open
Abstract
Lactoferrin is a multifunctional mammalian immunity protein that limits microbial growth through sequestration of nutrient iron. Additionally, lactoferrin possesses cationic protein domains that directly bind and inhibit diverse microbes. The implications for these dual functions on lactoferrin evolution and genetic conflicts with microbes remain unclear. Here we show that lactoferrin has been subject to recurrent episodes of positive selection during primate divergence predominately at antimicrobial peptide surfaces consistent with long-term antagonism by bacteria. An abundant lactoferrin polymorphism in human populations and Neanderthals also exhibits signatures of positive selection across primates, linking ancient host-microbe conflicts to modern human genetic variation. Rapidly evolving sites in lactoferrin further correspond to molecular interfaces with opportunistic bacterial pathogens causing meningitis, pneumonia, and sepsis. Because microbes actively target lactoferrin to acquire iron, we propose that the emergence of antimicrobial activity provided a pivotal mechanism of adaptation sparking evolutionary conflicts via acquisition of new protein functions. Immunity genes can evolve rapidly in response to antagonism by microbial pathogens, but how the emergence of new protein functions impacts such evolutionary conflicts remains unclear. Here we have traced the evolutionary history of the lactoferrin gene in primates, which in addition to an ancient iron-binding function, acquired antimicrobial peptide activity in mammals. We show that, in contrast to the related gene transferrin, lactoferrin has rapidly evolved at protein domains that mediate iron-independent antimicrobial functions. We also pinpoint signatures of natural selection acting on lactoferrin in human populations, suggesting that lactoferrin genetic diversity has impacted the evolutionary success of both ancient primates and humans. Our work demonstrates how the emergence of new host immune protein functions can drastically alter evolutionary and molecular interactions with microbes.
Collapse
Affiliation(s)
- Matthew F Barber
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Zev Kronenberg
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Mark Yandell
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Nels C Elde
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| |
Collapse
|
44
|
Abouhmad A, Mamo G, Dishisha T, Amin MA, Hatti-Kaul R. T4 lysozyme fused with cellulose-binding module for antimicrobial cellulosic wound dressing materials. J Appl Microbiol 2016; 121:115-25. [PMID: 27028513 DOI: 10.1111/jam.13146] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 03/13/2016] [Accepted: 03/23/2016] [Indexed: 12/19/2022]
Abstract
AIMS Bacterial infection is a major challenge in wound care. Antimicrobial wound dressings are of great value for treating wound infections. Endolysins are evolving as a new class of antimicrobials with multiple applications. This study describes the production and evaluation of T4 lysozyme (T4Lyz), product of gene e of the T4 bacteriophage, fused with Cellulose Binding Module (CBM) for facile immobilization to cellulosic wound dressing. METHODS AND RESULTS Genes encoding T4Lyz-CBM and T4Lyz were cloned and expressed in Escherichia coli and the enzymes were purified by cation exchange chromatography. While the CBM tag did not alter the optimum pH and stability features of T4Lyz, the lytic activity of the fusion protein was lowered. The bactericidal activity of T4Lyz-CBM, determined by viable count plating assay after 1 h incubation with Micrococcus lysodeikticus was 97·5% with 10 μg ml(-1) , and 99·96% and 95% for E. coli and Pseudomonas mendocina, respectively, with 200 μg ml(-1) enzyme. T4Lyz-CBM was immobilized to wound dressing gauze with a capacity of 5·5 μg mg(-1) matrix, whereas the unmodified T4Lyz did not exhibit any binding. The immobilized protein retained its bactericidal activity against Gram-positive and Gram-negative bacteria. Both free and immobilized T4Lyz-CBM, after heat denaturation, retained their bactericidal activities against Gram-negative bacteria only. The immobilized enzyme exhibited higher stability than the free enzyme when stored in dry form or in the presence of polyol stabilizers. CONCLUSION Tagging T4Lyz with CBM provides a facile, irreversible binding to cellulosic wound dressing while retaining its activity. This approach may be suitable even for other antimicrobial enzymes and -peptides. SIGNIFICANCE AND IMPACT OF THE STUDY The spread of antibiotic resistance requires innovative strategies for discovery and development of effective antimicrobial alternatives. This report presents a novel strategy for producing antimicrobial wound dressing materials.
Collapse
Affiliation(s)
- A Abouhmad
- Division of Biotechnology, Center for Chemistry and Chemical Engineering, Lund University, Lund, Sweden.,Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - G Mamo
- Division of Biotechnology, Center for Chemistry and Chemical Engineering, Lund University, Lund, Sweden
| | - T Dishisha
- Division of Biotechnology, Center for Chemistry and Chemical Engineering, Lund University, Lund, Sweden.,Department of Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - M A Amin
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - R Hatti-Kaul
- Division of Biotechnology, Center for Chemistry and Chemical Engineering, Lund University, Lund, Sweden
| |
Collapse
|
45
|
Hu L, Gao CH, Hong C, Zhong Q, Dong HL, Gao XM. Expression, purification, and breast cancer cell inhibiting effect of recombinant human lactoferrin C-lobe. Biosci Biotechnol Biochem 2015; 80:257-63. [PMID: 26405758 DOI: 10.1080/09168451.2015.1088376] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Lactoferrin (LTF), a multifunctional glycoprotein of the transferrin family mainly found in exotic secretions in mammals, is an important defense molecule against not only microbial invasion but also tumors. It folds into two globular domains (N- and C-lobes) each containing an iron-binding site. The cationic antimicrobial peptide in N-lobe is known to exert anti-tumor effect via a non-receptor-mediated pathway. However, whether LTF C-lobe also contributes to its anti-tumor activity remains to be investigated. In this study, a human LTF fragment (amino acid residues 343-682) covering the C-lobe was expressed with a histidine tag in E. coli and the purified polypeptide refolded through a series of buffer changing procedure. The resultant recombinant protein caused significant growth arrest of breast carcinoma cells MDA-MB-231 in a dose- and time-dependent manner, evidently via induction of apoptosis of the cell. Our data suggest a positive role for the C-lobe of human LTF in controlling tumors in vitro.
Collapse
Affiliation(s)
- Lulu Hu
- a Institute of Biology and Medical Sciences, School of Biology and Basic Medical Science, Soochow University , Suzhou , China
| | - Chen-Hui Gao
- a Institute of Biology and Medical Sciences, School of Biology and Basic Medical Science, Soochow University , Suzhou , China
| | - Chao Hong
- a Institute of Biology and Medical Sciences, School of Biology and Basic Medical Science, Soochow University , Suzhou , China
| | - Qiao Zhong
- a Institute of Biology and Medical Sciences, School of Biology and Basic Medical Science, Soochow University , Suzhou , China
| | - Hong-Liang Dong
- a Institute of Biology and Medical Sciences, School of Biology and Basic Medical Science, Soochow University , Suzhou , China
| | - Xiao-Ming Gao
- a Institute of Biology and Medical Sciences, School of Biology and Basic Medical Science, Soochow University , Suzhou , China
| |
Collapse
|
46
|
Padrão J, Machado R, Casal M, Lanceros-Méndez S, Rodrigues LR, Dourado F, Sencadas V. Antibacterial performance of bovine lactoferrin-fish gelatine electrospun membranes. Int J Biol Macromol 2015; 81:608-14. [PMID: 26314908 DOI: 10.1016/j.ijbiomac.2015.08.047] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 08/20/2015] [Accepted: 08/21/2015] [Indexed: 11/17/2022]
Abstract
The increase of antibiotic resistant microorganisms urged the development and synthesis of novel antimicrobial biomaterials to be employed in a broad range of applications, ranging from food packaging to medical devices. This work describes the production and characterization of a protein-based electrospun fibrous membranes bearing antimicrobial properties. Its composition is exclusively comprised of proteins, with fish gelatine as structural matrix and bovine lactoferrin (bLF) as the active antimicrobial agent. The bLF bactericidal effect was determined against clinical isolates of Escherichia coli and Staphylococcus aureus through microdilution assays. Two distinctive methods were used to incorporate bLF into the fish gelatine nanofibres: (i) as a filler in the electrospinning formulation with concentrations of 2, 5 and 10 (wt%), and cross-linked with glutaraldehyde vapour, in order to achieve stability in aqueous solution; and (ii) through adsorption in a solution with 40mgmL(-1) bLF. Fourier transform infrared spectroscopy analysis showed that the structure of both proteins remained intact through the electrospinning blending and cross-linking procedure. Remarkable antibacterial properties were obtained with membranes containing 5% and 10% bLF with a bacterial reduction of approximately 90% and 100%, respectively.
Collapse
Affiliation(s)
- Jorge Padrão
- Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Raul Machado
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, 4710-057 Braga, Portugal
| | - Margarida Casal
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, 4710-057 Braga, Portugal
| | | | - Ligia R Rodrigues
- Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Fernando Dourado
- Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Vitor Sencadas
- School of Mechanical, Materials and Mechatronics Engineering, University of Wollongong, Wollongong, NSW 2522, Australia.
| |
Collapse
|
47
|
Kanwar JR, Roy K, Patel Y, Zhou SF, Singh MR, Singh D, Nasir M, Sehgal R, Sehgal A, Singh RS, Garg S, Kanwar RK. Multifunctional iron bound lactoferrin and nanomedicinal approaches to enhance its bioactive functions. Molecules 2015; 20:9703-31. [PMID: 26016555 PMCID: PMC6272382 DOI: 10.3390/molecules20069703] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/13/2015] [Indexed: 02/08/2023] Open
Abstract
Lactoferrin (Lf), an iron-binding protein from the transferrin family has been reported to have numerous functions. Even though Lf was first isolated from milk, it is also found in most exocrine secretions and in the secondary granules of neutrophils. Antimicrobial and anti-inflammatory activity reports on lactoferrin identified its significance in host defense against infection and extreme inflammation. Anticarcinogenic reports on lactoferrin make this protein even more valuable. This review is focused on the structural configuration of iron-containing and iron-free forms of lactoferrin obtained from different sources such as goat, camel and bovine. Apart for emphasizing on the specific beneficial properties of lactoferrin from each of these sources, the general antimicrobial, immunomodulatory and anticancer activities of lactoferrin are discussed here. Implementation of nanomedicinial strategies that enhance the bioactive function of lactoferrin are also discussed, along with information on lactoferrin in clinical trials.
Collapse
Affiliation(s)
- Jagat R Kanwar
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Molecular and Medical Research (MMR) Strategic Research Centre, Faculty of Health, Deakin University, Waurn Ponds, Victoria 3217, Australia.
| | - Kislay Roy
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Molecular and Medical Research (MMR) Strategic Research Centre, Faculty of Health, Deakin University, Waurn Ponds, Victoria 3217, Australia.
| | - Yogesh Patel
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Molecular and Medical Research (MMR) Strategic Research Centre, Faculty of Health, Deakin University, Waurn Ponds, Victoria 3217, Australia.
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA.
| | - Manju Rawat Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur 492 010, India.
| | - Deependra Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur 492 010, India.
| | - Muhammad Nasir
- Department of Food Science & Human Nutrition, Faculty of Bio-Sciences, University of Veterinary & Animal Sciences, Lahore, Punjab 54000, Pakistan.
| | - Rakesh Sehgal
- Department of Medical Parasitology, Postgraduate Institute of Medical Education & Research, Chandigarh 160012, India.
| | - Alka Sehgal
- Department of Obstetrics & Gynecology, Government Medical College & Hospital, Sector 32, Chandigarh 160031, India.
| | - Ram Sarup Singh
- Carbohydrate and Protein Biotechnology Laboratory, Department of Biotechnology, Punjabi University, Patiala 147002, India.
| | - Sanjay Garg
- Centre for Pharmaceutical Innovation and Development (CPID), School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5000, Australia.
| | - Rupinder K Kanwar
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine (SoM), Molecular and Medical Research (MMR) Strategic Research Centre, Faculty of Health, Deakin University, Waurn Ponds, Victoria 3217, Australia.
| |
Collapse
|
48
|
Abstract
Lactoferrin is one of a number of multifunctional proteins that are present in or on all mucosal surfaces throughout the body. Levels of lactoferrin are consistently elevated in inflammatory diseases such as arthritis, inflammatory bowel diseases, corneal disease, and periodontitis. Single-nucleotide polymorphisms (SNPs) in lactoferrin have been shown to be present in individuals susceptible to Escherichia coli-induced travelers' diarrhea and in tear fluid derived from virally associated corneal disease. Here, we review data showing a lactoferrin SNP in amino acid position 29 in the antimicrobial region of lactoferrin that acts against caries associated bacteria. This SNP was initially discovered in African American subjects with localized aggressive periodontitis (LAP) who had proximal bone loss but minimal proximal caries. Results were confirmed in a genetic association study of children from Brazil with this same SNP who showed a reduced level of caries. In vitro data indicate that lactoferrin from whole saliva derived from subjects with this SNP, recombinant human lactoferrin containing this SNP, or an 11-mer peptide designed for this SNP kills mutans streptococci associated with caries by >1 log. In contrast, the SNP has minimal effect on Gram-negative species associated with periodontitis. Moreover, periodontally healthy subjects homozygous for this lysine (K) SNP have lactoferrin in their saliva that kills mutans streptococci and have reduced proximal decay. The review summarizes data supporting the ecologic plaque hypothesis and suggests that a genetic variant in lactoferrin with K in position 29 when found in saliva and crevice fluid can influence community biofilm composition. We propose that, for caries, this SNP is ethnicity independent and protective by directly killing caries-provoking bacteria (reducing proximal decay). However, the clinical effect of this SNP in LAP is ethnicity dependent, destructive (increases LAP incidence), and complex with mechanisms still to be determined.
Collapse
Affiliation(s)
- D H Fine
- Department of Oral Biology, Rutgers School of Dental Medicine, Rutgers University, Newark, NJ, USA
| |
Collapse
|
49
|
Redwan EM, Uversky VN, El-Fakharany EM, Al-Mehdar H. Potential lactoferrin activity against pathogenic viruses. C R Biol 2014; 337:581-95. [PMID: 25282173 DOI: 10.1016/j.crvi.2014.08.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 08/10/2014] [Accepted: 08/11/2014] [Indexed: 10/24/2022]
Abstract
Lactoferrin (LF) is an 80-kDa globular glycoprotein with high affinity for metal ions, particularly for iron. This protein possesses many biological functions, including the binding and release of iron and serves as one of the important components of the innate immune system, where it acts as a potent inhibitor of several pathogens. LF has efficacious antibacterial and antiviral activities against a wide range of Gram-positive and Gram-negative bacteria and against both naked and enveloped DNA and RNA viruses. In its antiviral pursuit, LF acts predominantly at the acute phase of the viral infection or even at the intracellular stage, as in hepatitis C virus infection. LF inhibits the entry of viral particles into host cells, either by direct attachment to the viral particles or by blocking their cellular receptors. This wide range of activities may be attributed to the capacity of LF to bind iron and its ability to interfere with the cellular receptors of both hosts and pathogenic microbes.
Collapse
Affiliation(s)
- Elrashdy M Redwan
- Biology Department, Faculty of Science, King Abdulaziz University, PO Box 80203, Jeddah 21589, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technological Applications, New Borg EL-Arab 21394, Alexandria, Egypt.
| | - Vladimir N Uversky
- Biology Department, Faculty of Science, King Abdulaziz University, PO Box 80203, Jeddah 21589, Saudi Arabia; Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; Institute for Biological Instrumentation, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia.
| | - Esmail M El-Fakharany
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technological Applications, New Borg EL-Arab 21394, Alexandria, Egypt.
| | - Hussein Al-Mehdar
- Biology Department, Faculty of Science, King Abdulaziz University, PO Box 80203, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
50
|
Redwan EM, EL-Fakharany EM, Uversky VN, Linjawi MH. Screening the anti infectivity potentials of native N- and C-lobes derived from the camel lactoferrin against hepatitis C virus. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:219. [PMID: 24993815 PMCID: PMC4086701 DOI: 10.1186/1472-6882-14-219] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 06/30/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Hepatitis C virus (HCV) infection represents a worldwide health threat that still needs efficient protective vaccine and/or effective drug. The traditional medicine, such as camel milk, is heavily used by the large sector of HCV patients to control the infection due to the high cost of the available standard therapy. Camel milk contains lactoferrin, which plays an important and multifunctional role in innate immunity and specific host defense against microbial infection. Continuing the analysis of the effectiveness of camel lactoferrin against HCV, the current study aimed to separate and purify the native N- and C-lobes from the proteolytically cleaved camel lactoferrin (cLF) and to compare their in vitro activities against the HCV infection in Huh7.5 cells in order to determine the most active domain. METHODS Lactoferrin and its digested N- and C-lobes were purified by Mono S 5/50 GL column and Superdex 200 5/150 column. The purified proteins were assessed through three venues: 1. To inhibit intracellular replication, HCV infected cells were treated with the proteins at different concentrations and time intervals; 2. The proteins were directly incubated with the viral particles (neutralization) and then such neutralized viruses were used to infect cells; 3. The cells were protected with proteins before exposure to the virus. The antiviral potentials of the cLf and its lobes were determined using three techniques: 1. RT-nested PCR, 2. Real-time PCR, and 3. Flow cytometry. RESULTS N- and C-lobes were purified in two consecutive steps; using Mono-S and Superdex 200 columns. The molecular mass of N- and C-lobes was about 40 kDa. cLF and its lobes could prevent HCV entry into Huh 7.5 cells with activity reached 100% through direct interaction with the virus. The inhibition of intracellular viral replication by N-lobe is 2-fold and 3-fold more effective than that of the cLF and C-lobe, respectively. CONCLUSION Generated native N- and C-lobes from camel lactoferrin demonstrated a range of noticeably different potentials against HCV cellular infectivity. The anti-HCV activities were sorted as N-lobe > cLf > C-lobe.
Collapse
|