1
|
Hussein M, Sun Z, Hawkey J, Allobawi R, Judd LM, Carbone V, Sharma R, Thombare V, Baker M, Rao GG, Li J, Holt KE, Velkov T. High-level nitrofurantoin resistance in a clinical isolate of Klebsiella pneumoniae: a comparative genomics and metabolomics analysis. mSystems 2024; 9:e0097223. [PMID: 38078757 PMCID: PMC10805014 DOI: 10.1128/msystems.00972-23] [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: 09/21/2023] [Accepted: 11/02/2023] [Indexed: 01/24/2024] Open
Abstract
Nitrofurantoin is a commonly used chemotherapeutic agent in the treatment of uncomplicated urinary tract infections caused by the problematic multidrug resistant Gram-negative pathogen Klebsiella pneumoniae. The present study aims to elucidate the mechanism of nitrofurantoin action and high-level resistance in K. pneumoniae using whole-genome sequencing (WGS), qPCR analysis, mutation structural modeling and untargeted metabolomic analysis. WGS profiling of evolved highly resistant mutants (nitrofurantoin minimum inhibitory concentrations > 256 mg/L) revealed modified expression of several genes related to membrane transport (porin ompK36 and efflux pump regulator oqxR) and nitroreductase activity (ribC and nfsB, involved in nitrofurantoin reduction). Untargeted metabolomics analysis of total metabolites extracted at 1 and 4 h post-nitrofurantoin treatment revealed that exposure to the drug caused a delayed effect on the metabolome which was most pronounced after 4 h. Pathway enrichment analysis illustrated that several complex interrelated metabolic pathways related to nitrofurantoin bacterial killing (aminoacyl-tRNA biosynthesis, purine metabolism, central carbohydrate metabolism, and pantothenate and CoA biosynthesis) and the development of nitrofurantoin resistance (riboflavin metabolism) were significantly perturbed. This study highlights for the first time the key role of efflux pump regulator oqxR in nitrofurantoin resistance and reveals global metabolome perturbations in response to nitrofurantoin, in K. pneumoniae.IMPORTANCEA quest for novel antibiotics and revitalizing older ones (such as nitrofurantoin) for treatment of difficult-to-treat Gram-negative bacterial infections has become increasingly popular. The precise antibacterial activity of nitrofurantoin is still not fully understood. Furthermore, although the prevalence of nitrofurantoin resistance remains low currently, the drug's fast-growing consumption worldwide highlights the need to comprehend the emerging resistance mechanisms. Here, we used multidisciplinary techniques to discern the exact mechanism of nitrofurantoin action and high-level resistance in Klebsiella pneumoniae, a common cause of urinary tract infections for which nitrofurantoin is the recommended treatment. We found that the expression of multiple genes related to membrane transport (including active efflux and passive diffusion of drug molecules) and nitroreductase activity was modified in nitrofurantoin-resistant strains, including oqxR, the transcriptional regulator of the oqxAB efflux pump. Furthermore, complex interconnected metabolic pathways that potentially govern the nitrofurantoin-killing mechanisms (e.g., aminoacyl-tRNA biosynthesis) and nitrofurantoin resistance (riboflavin metabolism) were significantly inhibited following nitrofurantoin treatment. Our study could help inform the improvement of nitrofuran derivatives, the development of new pharmacophores, or drug combinations to support the resurgence of nitrofurantoin in the management of multidrug resistant K. pneumouniae infection.
Collapse
Affiliation(s)
- Maytham Hussein
- Department of Pharmacology, Monash Biomedicine Discovery Institute,Monash University, Clayton, Victoria, Australia
| | - Zetao Sun
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Jane Hawkey
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Rafah Allobawi
- Department of Pharmacology, Monash Biomedicine Discovery Institute,Monash University, Clayton, Victoria, Australia
| | - Louise M. Judd
- Doherty Applied Microbial Genomics (DAMG), 12 Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Vincenzo Carbone
- AgResearch Limited, Grasslands Research Center, Tennent Drive, Palmerston North, New Zealand
| | - Rajnikant Sharma
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Varsha Thombare
- Department of Pharmacology, Monash Biomedicine Discovery Institute,Monash University, Clayton, Victoria, Australia
| | - Mark Baker
- Discipline of Biological 17 Sciences, Priority Research Center in Reproductive Biology, Faculty of Science and IT, University of Newcastle, University Drive, Callaghan, New South Wales, Australia
| | - Gauri G. Rao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jian Li
- Department of Pharmacology, Monash Biomedicine Discovery Institute,Monash University, Clayton, Victoria, Australia
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Kathryn E. Holt
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Tony Velkov
- Department of Pharmacology, Monash Biomedicine Discovery Institute,Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
2
|
Ohradanova-Repic A, Praženicová R, Gebetsberger L, Moskalets T, Skrabana R, Cehlar O, Tajti G, Stockinger H, Leksa V. Time to Kill and Time to Heal: The Multifaceted Role of Lactoferrin and Lactoferricin in Host Defense. Pharmaceutics 2023; 15:1056. [PMID: 37111542 PMCID: PMC10146187 DOI: 10.3390/pharmaceutics15041056] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Lactoferrin is an iron-binding glycoprotein present in most human exocrine fluids, particularly breast milk. Lactoferrin is also released from neutrophil granules, and its concentration increases rapidly at the site of inflammation. Immune cells of both the innate and the adaptive immune system express receptors for lactoferrin to modulate their functions in response to it. On the basis of these interactions, lactoferrin plays many roles in host defense, ranging from augmenting or calming inflammatory pathways to direct killing of pathogens. Complex biological activities of lactoferrin are determined by its ability to sequester iron and by its highly basic N-terminus, via which lactoferrin binds to a plethora of negatively charged surfaces of microorganisms and viruses, as well as to mammalian cells, both normal and cancerous. Proteolytic cleavage of lactoferrin in the digestive tract generates smaller peptides, such as N-terminally derived lactoferricin. Lactoferricin shares some of the properties of lactoferrin, but also exhibits unique characteristics and functions. In this review, we discuss the structure, functions, and potential therapeutic uses of lactoferrin, lactoferricin, and other lactoferrin-derived bioactive peptides in treating various infections and inflammatory conditions. Furthermore, we summarize clinical trials examining the effect of lactoferrin supplementation in disease treatment, with a special focus on its potential use in treating COVID-19.
Collapse
Affiliation(s)
- Anna Ohradanova-Repic
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Romana Praženicová
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| | - Laura Gebetsberger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Tetiana Moskalets
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| | - Rostislav Skrabana
- Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Ondrej Cehlar
- Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Gabor Tajti
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Hannes Stockinger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Vladimir Leksa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| |
Collapse
|
3
|
Bagel A, Lopez C, David-Briand E, Michel V, Douëllou T, Sergentet D. Serotype-dependent adhesion of Shiga toxin-producing Escherichia coli to bovine milk fat globule membrane proteins. Front Microbiol 2022; 13:1010665. [PMID: 36504830 PMCID: PMC9731836 DOI: 10.3389/fmicb.2022.1010665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
Shiga toxin-producing Escherichia coli (STEC) are food-borne pathogens that can cause severe symptoms for humans. Raw milk products are often incriminated as vehicule for human STEC infection. However, raw milk naturally contains molecules, such as the milk fat globule membrane and associated proteins, that could inhibit pathogen adhesion by acting as mimetic ligands. This study aimed to: (i) evaluate the capability of STEC cells to adhere to bovine milk fat globule membrane proteins (MFGMPs), (ii) highlight STEC surface proteins associated with adhesion and (iii) evaluate the variation between different STEC serotypes. We evaluated the physicochemical interactions between STEC and milk fat globules (MFGs) by analyzing hydrophobic properties and measuring the ζ-potential. We used a plate adhesion assay to assess adhesion between MFGMPs and 15 Escherichia coli strains belonging to three key serotypes (O157:H7, O26:H11, and O103:H2). A relative quantitative proteomic approach was conducted by mass spectrometry to identify STEC surface proteins that may be involved in STEC-MFG adhesion. The majority of E. coli strains showed a hydrophilic profile. The ζ-potential values were between -3.7 and - 2.9 mV for the strains and between -12.2 ± 0.14 mV for MFGs. Our results suggest that non-specific interactions are not strongly involved in STEC-MFG association and that molecular bonds could form between STEC and MFGs. Plate adhesion assays showed a weak adhesion of O157:H7 E. coli strains to MFGMPs. In contrast, O26:H11 and O103:H2 serotypes attached more to MFGMPs. Relative quantitative proteomic analysis showed that the O26:H11 str. 21,765 differentially expressed five outer membrane-associated proteins or lipoproteins compared with the O157:H7 str. EDL933. This analysis also found strain-specific differentially expressed proteins, including four O26:H11 str. 21,765-specific proteins/lipoproteins and eight O103:H2 str. PMK5-specific proteins. For the first time, we demonstrated STEC adhesion to MFGMPs and discovered a serotype effect. Several outer membrane proteins-OmpC and homologous proteins, intimin, Type 1 Fimbriae, and AIDA-I-that may be involved in STEC-MFG adhesion were highlighted. More research on STEC's ability to adhere to MFGMs in diverse biological environments, such as raw milk cheeses and the human gastrointestinal tract, is needed to confirm the anti-adhesion properties of the STEC-MFG complex.
Collapse
Affiliation(s)
- Arthur Bagel
- Bacterial Opportunistic Pathogens and Environment Research Group, UMR5557 Ecologie Microbienne Lyon, National Center of Scientific Research (CNRS), Université de Lyon, Marcy-l’Etoile, France
| | | | | | | | - Thomas Douëllou
- Bacterial Opportunistic Pathogens and Environment Research Group, UMR5557 Ecologie Microbienne Lyon, National Center of Scientific Research (CNRS), Université de Lyon, Marcy-l’Etoile, France
| | - Delphine Sergentet
- Bacterial Opportunistic Pathogens and Environment Research Group, UMR5557 Ecologie Microbienne Lyon, National Center of Scientific Research (CNRS), Université de Lyon, Marcy-l’Etoile, France,Laboratoire d’Etudes des Microorganismes Alimentaires Pathogènes, VetAgro Sup—Campus Vétérinaire, French National Reference Laboratory for Escherichia coli Including Shiga Toxin-Producing E. coli (NRL-STEC), Université de Lyon, Marcy-l‘Etoile, France,*Correspondence: Delphine Sergentet,
| |
Collapse
|
4
|
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
|
5
|
Enigk K, Jentsch H, Rodloff AC, Eschrich K, Stingu CS. Activity of five antimicrobial peptides against periodontal as well as non-periodontal pathogenic strains. J Oral Microbiol 2020; 12:1829405. [PMID: 33133417 PMCID: PMC7580719 DOI: 10.1080/20002297.2020.1829405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022] Open
Abstract
Background: Due to the increasing emergence of multi-resistant bacteria the search for alternative antimicrobial substances is of high interest. Promising agents are antimicrobial peptides which are host defense molecules of the innate immune system in a wide range of different species. Objectives: The aim of this study was to assess the activity of nisin, melittin, lactoferrin, parasin-1 and LL-37 against 35 oral bacteria and Candida albicans employing the gold standard method for anaerobic susceptibility testing. Methods: The activity of the peptides was determined by an agar dilution method under anaerobic and aerobic conditions. The test media contained final peptide concentrations between 0.125 µg/ml and 8 µg/ml (melittin, lactoferrin, parasin-1, LL-37) and between 0.125 µg/ml and 128 µg/ml (nisin). Results: Nisin completely inhibited the growth of Megasphaera sp., Bifidobacterium longum, Parvimonas micra, Actinomyces israelii, Actinomyces naeslundii, Actinomyces odontolyticus, Prevotella intermedia, Streptococcus anginosus, Streptococcus constellatus and Staphylococcus aureus. Melittin and lactoferrin reduced the growth of Megasphaera sp., P. micra, B. longum (melittin) and Selenomonas flueggei (lactoferrin). Parasin-1 and LL-37 showed no activity. Conclusion: AMPs, especially nisin and to a smaller degree lactoferrin, might be promising alternatives to antibiotics because of their antimicrobial activity, high resistance to environmental conditions and partially low costs.
Collapse
Affiliation(s)
- Katharina Enigk
- Institute for Medical Microbiology and Epidemiology of Infectious Diseases, University Hospital of Leipzig, Leipzig, Germany
| | - Holger Jentsch
- Center for Periodontology, Department of Cariology, Endodontology and Periodontology, University of Leipzig, Leipzig, Germany
| | - Arne C. Rodloff
- Institute for Medical Microbiology and Epidemiology of Infectious Diseases, University Hospital of Leipzig, Leipzig, Germany
| | - Klaus Eschrich
- Institute of Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Catalina-Suzana Stingu
- Institute for Medical Microbiology and Epidemiology of Infectious Diseases, University Hospital of Leipzig, Leipzig, Germany
| |
Collapse
|
6
|
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
|
7
|
Kepiro IE, Marzuoli I, Hammond K, Ba X, Lewis H, Shaw M, Gunnoo SB, De Santis E, Łapińska U, Pagliara S, Holmes MA, Lorenz CD, Hoogenboom BW, Fraternali F, Ryadnov MG. Engineering Chirally Blind Protein Pseudocapsids into Antibacterial Persisters. ACS NANO 2020; 14:1609-1622. [PMID: 31794180 DOI: 10.1021/acsnano.9b06814] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Antimicrobial resistance stimulates the search for antimicrobial forms that may be less subject to acquired resistance. Here we report a conceptual design of protein pseudocapsids exhibiting a broad spectrum of antimicrobial activities. Unlike conventional antibiotics, these agents are effective against phenotypic bacterial variants, while clearing "superbugs" in vivo without toxicity. The design adopts an icosahedral architecture that is polymorphic in size, but not in shape, and that is available in both l and d epimeric forms. Using a combination of nanoscale and single-cell imaging we demonstrate that such pseudocapsids inflict rapid and irreparable damage to bacterial cells. In phospholipid membranes they rapidly convert into nanopores, which remain confined to the binding positions of individual pseudocapsids. This mechanism ensures precisely delivered influxes of high antimicrobial doses, rendering the design a versatile platform for engineering structurally diverse and functionally persistent antimicrobial agents.
Collapse
Affiliation(s)
- Ibolya E Kepiro
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
| | - Irene Marzuoli
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
- Randall Centre for Cell and Molecular Biophysics , King's College London , London , SE1 1UL , U.K
| | - Katharine Hammond
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
- Department of Physics and Astronomy , University College London , London , WC1E 6BT , U.K
- London Centre for Nanotechnology , University College London , London , WC1H 0AH , U.K
| | - Xiaoliang Ba
- Department of Veterinary Medicine , University of Cambridge , Cambridge , CB3 0ES , U.K
| | - Helen Lewis
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
| | - Michael Shaw
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
- Department of Computer Science , University College London , London , WC1 6BT , U.K
| | - Smita B Gunnoo
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
| | - Emiliana De Santis
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
| | - Urszula Łapińska
- Living Systems Institute , University of Exeter , Exeter , EX4 4QD , U.K
| | - Stefano Pagliara
- Living Systems Institute , University of Exeter , Exeter , EX4 4QD , U.K
| | - Mark A Holmes
- Department of Veterinary Medicine , University of Cambridge , Cambridge , CB3 0ES , U.K
| | - Christian D Lorenz
- Department of Physics , King's College London , Strand Lane , London , WC2R 2LS , U.K
| | - Bart W Hoogenboom
- Department of Physics and Astronomy , University College London , London , WC1E 6BT , U.K
- London Centre for Nanotechnology , University College London , London , WC1H 0AH , U.K
| | - Franca Fraternali
- Randall Centre for Cell and Molecular Biophysics , King's College London , London , SE1 1UL , U.K
| | - Maxim G Ryadnov
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
- Department of Physics , King's College London , Strand Lane , London , WC2R 2LS , U.K
| |
Collapse
|
8
|
Rapid colorimetric lactoferrin-based sandwich immunoassay on cotton swabs for the detection of foodborne pathogenic bacteria. Talanta 2018; 185:275-280. [DOI: 10.1016/j.talanta.2018.03.072] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/22/2018] [Accepted: 03/24/2018] [Indexed: 11/24/2022]
|
9
|
Duong DA, Jensen RV, Stevens AM. Discovery of Pantoea stewartii ssp. stewartii genes important for survival in corn xylem through a Tn-Seq analysis. MOLECULAR PLANT PATHOLOGY 2018; 19:1929-1941. [PMID: 29480976 PMCID: PMC6638119 DOI: 10.1111/mpp.12669] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 05/29/2023]
Abstract
The bacterium Pantoea stewartii ssp. stewartii causes Stewart's wilt disease in corn. Pantoea stewartii is transmitted to plants via corn flea beetles, where it first colonizes the apoplast causing water-soaked lesions, and then migrates to the xylem and forms a biofilm that blocks water transport. Bacterial quorum sensing ensures that the exopolysaccharide production necessary for biofilm formation occurs only at high cell density. A genomic-level transposon sequencing (Tn-Seq) analysis was performed to identify additional bacterial genes essential for survival in planta and to provide insights into the plant-microbe interactions occurring during wilt disease. A mariner transposon library of approximately 40 000 mutants was constructed and used to inoculate corn seedlings through a xylem infection model. Cultures of the library grown in Luria-Bertani (LB) broth served as the in vitro pre-inoculation control. Tn-Seq analysis showed that the number of transposon mutations was reduced by more than 10-fold for 486 genes in planta compared with the library that grew in LB, suggesting that they are important for xylem survival. Interestingly, a small set of genes had a higher abundance of mutants in planta versus in vitro conditions, indicating enhanced strain fitness with loss of these genes inside the host. In planta competition assays retested the trends of the Tn-Seq data for several genes, including two outer membrane proteins, Lon protease and two quorum sensing-associated transcription factors, RcsA and LrhA. Virulence assays were performed to check for correlation between growth/colonization and pathogenicity. This study demonstrates the capacity of a Tn-Seq approach to advance our understanding of P. stewartii-corn interactions.
Collapse
Affiliation(s)
- Duy An Duong
- Department of Biological SciencesVirginia TechBlacksburgVA 24061USA
| | | | - Ann M. Stevens
- Department of Biological SciencesVirginia TechBlacksburgVA 24061USA
| |
Collapse
|
10
|
van Teeseling MCF, Benz R, de Almeida NM, Jetten MSM, Mesman RJ, van Niftrik L. Characterization of the first planctomycetal outer membrane protein identifies a channel in the outer membrane of the anammox bacterium Kuenenia stuttgartiensis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:767-776. [PMID: 29288627 DOI: 10.1016/j.bbamem.2017.12.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/30/2017] [Accepted: 12/25/2017] [Indexed: 01/27/2023]
Abstract
Planctomycetes are a bacterial phylum known for their complex intracellular compartmentalization. While most Planctomycetes have two compartments, the anaerobic ammonium oxidizing (anammox) bacteria contain three membrane-enclosed compartments. In contrast to a long-standing consensus, recent insights suggested the outermost Planctomycete membrane to be similar to a Gram-negative outer membrane (OM). One characteristic component that differentiates OMs from cytoplasmic membranes (CMs) is the presence of outer membrane proteins (OMPs) featuring a β-barrel structure that facilitates passage of molecules through the OM. Although proteomic and genomic evidence suggested the presence of OMPs in several Planctomycetes, no experimental verification existed of the pore-forming function and localization of these proteins in the outermost membrane of these exceptional microorganisms. Here, we show via lipid bilayer assays that at least two typical OMP-like channel-forming proteins are present in membrane preparations of the anammox bacterium Kuenenia stuttgartiensis. One of these channel-forming proteins, the highly abundant putative OMP Kustd1878, was purified to homogeneity. Analysis of the channel characteristics via lipid bilayer assays showed that Kustd1878 forms a moderately cation-selective channel with a high current noise and an average single-channel conductance of about 170-190pS in 1M KCl. Antibodies were raised against the purified protein and immunogold localization indicated Kustd1878 to be present in the outermost membrane. Therefore, this work clearly demonstrates the presence of OMPs in anammox Planctomycetes and thus firmly adds to the emerging view that Planctomycetes have a Gram-negative cell envelope.
Collapse
Affiliation(s)
- Muriel C F van Teeseling
- Department of Microbiology, Institute for Water and Wetland Research, Faculty of Science, Radboud University, Nijmegen, The Netherlands.
| | - Roland Benz
- Department of Life Sciences and Chemistry, Jacobs University, Bremen, Germany
| | - Naomi M de Almeida
- Department of Microbiology, Institute for Water and Wetland Research, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Mike S M Jetten
- Department of Microbiology, Institute for Water and Wetland Research, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Rob J Mesman
- Department of Microbiology, Institute for Water and Wetland Research, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Laura van Niftrik
- Department of Microbiology, Institute for Water and Wetland Research, Faculty of Science, Radboud University, Nijmegen, The Netherlands.
| |
Collapse
|
11
|
El-Hawiet A. A Simple, Sensitive, and Label-Free Platform for the Quantification of Lactoferrin in Camel and Goat Milk Based on Thin-Layer Chromatography. Chromatographia 2017. [DOI: 10.1007/s10337-017-3414-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
12
|
Rybarczyk J, Kieckens E, Vanrompay D, Cox E. In vitro and in vivo studies on the antimicrobial effect of lactoferrin against Escherichia coli O157:H7. Vet Microbiol 2017; 202:23-28. [DOI: 10.1016/j.vetmic.2016.05.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 05/11/2016] [Accepted: 05/18/2016] [Indexed: 10/21/2022]
|
13
|
Shimazaki KI, Kawai K. Advances in lactoferrin research concerning bovine mastitis. Biochem Cell Biol 2017; 95:69-75. [DOI: 10.1139/bcb-2016-0044] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Lactoferrin is a multifunctional, iron-binding glycoprotein found in milk and other exocrine secretions. Lactoferrin in milk plays vital roles in the healthy development of newborn mammals, and is also an innate resistance factor involved in the prevention of mammary gland infection by microorganisms. Inflammation of the udder because of bacterial infection is referred to as mastitis. There have been many investigations into the relationships between lactoferrin and mastitis, which fall into several categories. The main categories are fluctuations in the lactoferrin concentration of milk, lactoferrin activity against mastitis pathogens, elucidation of the processes underlying the onset of mastitis, participation of lactoferrin in the immune system, and utilization of lactoferrin in mastitis treatment and prevention. This minireview describes lactoferrin research concerning bovine mastitis. In the 1970s, many researchers reported that the lactoferrin concentration fluctuates in milk from cows with mastitis. From the late 1980s, many studies clarified the infection-defense mechanism in the udder and the contribution of lactoferrin to the immune system. After the year 2000, the processes underlying the onset of mastitis were elucidated in vivo and in vitro, and lactoferrin was applied for the treatment and prevention of mastitis.
Collapse
Affiliation(s)
| | - Kazuhiro Kawai
- School of Veterinary Medicine, Azabu University, Sagamihara, 252-5201, Japan
| |
Collapse
|
14
|
Kim WS, Kim PH, Shimazaki KI. Sensitivity of Pseudomonas syringae to Bovine Lactoferrin Hydrolysates and Identification of a Novel Inhibitory Peptide. Korean J Food Sci Anim Resour 2016; 36:487-93. [PMID: 27621689 PMCID: PMC5018508 DOI: 10.5851/kosfa.2016.36.4.487] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/04/2016] [Indexed: 01/06/2023] Open
Abstract
The antimicrobial activity of bovine lactoferrin hydrolysates (bLFH) was measured against Pseudomonas strains (P. syringae and P. fluorescens) in vitro. To compare susceptibility to bLFH, minimal inhibitory concentration (MIC) values were determined using chemiluminescence assays and paper disc plate assays. Antimicrobial effect against P. fluorescens was not observed by either assay, suggesting that bLFH did not exhibit antimicrobial activity against P. fluorescens. However, a significant inhibition of P. syringae growth was observed in the presence of bLFH. The addition of bLFH in liquid or solid medium inhibited growth of P. syringae in a dose-dependent manner. Furthermore, a bLFH peptide with antimicrobial activity toward P. syringae was isolated and identified. The N-terminal amino acid sequences of thus obtained antimicrobial bLFH peptides were analyzed by a protein sequencer and were found to be Leu-Arg-Ile-Pro-Ser-Lys-Val-Asp-Ser-Ala and Phe-Lys-Cys-Arg-Arg-Trp-Gln-Trp-Arg-Met. The latter peptide sequence is known to be characteristic of lactoferricin. Therefore, in the present study, we identified a new antimicrobial peptide against P. syringae, present within the N-terminus and possessing the amino acid sequence of Leu-Arg-Ile-Pro-Ser-Lys-Val-Asp-Ser-Ala.
Collapse
Affiliation(s)
- Woan-Sub Kim
- Department of Animal Life and Environmental Science, Hankyong National University, Anseong 17579, Korea
| | - Pyeung-Hyeun Kim
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 24341, Korea
| | - Kei-Ichi Shimazaki
- Dairy Food Science Laboratory, Research Faculty of Agriculture, Hokkaido University, Sapporo 060-8589, Japan
| |
Collapse
|
15
|
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
|
16
|
Murdock C, Chikindas ML, Matthews KR. The Pepsin Hydrolysate of Bovine Lactoferrin Causes a Collapse of the Membrane Potential in Escherichia coli O157:H7. Probiotics Antimicrob Proteins 2016; 2:112-9. [PMID: 26781120 DOI: 10.1007/s12602-010-9039-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In the present study, the ability of bovine lactoferrin hydrolysate (LfH) to disrupt the cytoplasmic membrane of Escherichia coli O157:H7 was investigated. Lactoferrin and LfH antimicrobial activities were compared against E. coli O157:H7 and E. coli O157:H7 spheroplasts. The effect of LfH on the cytoplasmic membrane of E. coli O157:H7 cells was determined by evaluating potassium efflux (K(+)), dissipation of ATP and membrane potential (ΔΨ). LfH produced a rapid efflux of potassium ions, a decrease in intracellular levels of ATP coupled with a substantial increase in extracellular ATP levels and a complete dissipation of the ΔΨ. The results suggest that LfH causes a collapse of the membrane integrity by pore formation in the inner membrane, leading to the death of the cell. Moreover, the mechanism of action of LfH on E. coli O157:H7 appears to involve an interference with the inner membrane integrity based on experiments using E. coli O157:H7 spheroplasts.
Collapse
Affiliation(s)
- Christopher Murdock
- Department of Food Science, Rutgers, The State University of New Jersey, 65 Dudley Road, New Brunswick, NJ, 08901-8520, USA
| | - Michael L Chikindas
- Department of Food Science, Rutgers, The State University of New Jersey, 65 Dudley Road, New Brunswick, NJ, 08901-8520, USA
| | - Karl R Matthews
- Department of Food Science, Rutgers, The State University of New Jersey, 65 Dudley Road, New Brunswick, NJ, 08901-8520, USA.
| |
Collapse
|
17
|
Hajishengallis G, Russell MW. Innate Humoral Defense Factors. Mucosal Immunol 2015. [PMCID: PMC7149745 DOI: 10.1016/b978-0-12-415847-4.00015-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although innate immunity came into the research spotlight in the late 1990s when its instructive role in the adaptive immune response was recognized, innate humoral defense factors have a much older history. The exocrine secretions of the body contain a plethora of distinct soluble factors (lysozyme, lactoferrin, peroxidases, proline-rich proteins, histatins, etc.) that protect the body from mucosal microbial pathogens. More recent studies have established that the humoral arm of innate immunity contains a heterogeneous group of pattern-recognition molecules (e.g., pentraxins, collectins, and ficolins), which perform diverse host-defense functions, such as agglutination and neutralization, opsonization, control of inflammation, and complement activation and regulation. These pattern-recognition molecules, which act as functional predecessors of antibodies (“ante-antibodies”), and the classic soluble innate defense factors form an integrated system with complementary specificity, action, and tissue distribution, and they are the subject of this chapter.
Collapse
|
18
|
Sandrini S, Masania R, Zia F, Haigh R, Freestone P. Role of porin proteins in acquisition of transferrin iron by enteropathogens. Microbiology (Reading) 2013; 159:2639-2650. [DOI: 10.1099/mic.0.071928-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Sara Sandrini
- Department of Infection, Immunity and Inflammation, University of Leicester, University Road, Leicester LE1 9HN, UK
| | - Rikesh Masania
- Department of Infection, Immunity and Inflammation, University of Leicester, University Road, Leicester LE1 9HN, UK
| | - Fatima Zia
- Department of Infection, Immunity and Inflammation, University of Leicester, University Road, Leicester LE1 9HN, UK
| | - Richard Haigh
- Department of Genetics, University of Leicester, University Road, Leicester LE1 9HN, UK
| | - Primrose Freestone
- Department of Infection, Immunity and Inflammation, University of Leicester, University Road, Leicester LE1 9HN, UK
| |
Collapse
|
19
|
Refolding and functional assembly of the Vibrio cholerae porin OmpU recombinantly expressed in the cytoplasm of Escherichia coli. Protein Expr Purif 2012; 85:204-10. [DOI: 10.1016/j.pep.2012.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 08/08/2012] [Indexed: 11/20/2022]
|
20
|
Thein M, Bonde M, Bunikis I, Denker K, Sickmann A, Bergström S, Benz R. DipA, a pore-forming protein in the outer membrane of Lyme disease spirochetes exhibits specificity for the permeation of dicarboxylates. PLoS One 2012; 7:e36523. [PMID: 22590556 PMCID: PMC3349712 DOI: 10.1371/journal.pone.0036523] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 04/03/2012] [Indexed: 11/18/2022] Open
Abstract
Lyme disease Borreliae are highly dependent on the uptake of nutrients provided by their hosts. Our study describes the identification of a 36 kDa protein that functions as putative dicarboxylate-specific porin in the outer membrane of Lyme disease Borrelia. The protein was purified by hydroxyapatite chromatography from Borrelia burgdorferi B31 and designated as DipA, for dicarboxylate-specific porin A. DipA was partially sequenced, and corresponding genes were identified in the genomes of B. burgdorferi B31, Borrelia garinii PBi and Borrelia afzelii PKo. DipA exhibits high homology to the Oms38 porins of relapsing fever Borreliae. B. burgdorferi DipA was characterized using the black lipid bilayer assay. The protein has a single-channel conductance of 50 pS in 1 M KCl, is slightly selective for anions with a permeability ratio for cations over anions of 0.57 in KCl and is not voltage-dependent. The channel could be partly blocked by different di- and tricarboxylic anions. Particular high stability constants up to about 28,000 l/mol (in 0.1 M KCl) were obtained among the 11 tested anions for oxaloacetate, 2-oxoglutarate and citrate. The results imply that DipA forms a porin specific for dicarboxylates which may play an important role for the uptake of specific nutrients in different Borrelia species.
Collapse
Affiliation(s)
- Marcus Thein
- Rudolf-Virchow-Center, DFG-Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Mari Bonde
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Ignas Bunikis
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Katrin Denker
- Rudolf-Virchow-Center, DFG-Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Albert Sickmann
- Rudolf-Virchow-Center, DFG-Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
- Department of Bioanalytics, Leibniz-Institut für Analytische Wissenschaften – ISAS, Dortmund, Germany
| | - Sven Bergström
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Roland Benz
- Rudolf-Virchow-Center, DFG-Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
- School of Engineering and Science, Jacobs University Bremen, Bremen, Germany
- * E-mail:
| |
Collapse
|
21
|
Teixeira V, Feio MJ, Bastos M. Role of lipids in the interaction of antimicrobial peptides with membranes. Prog Lipid Res 2012; 51:149-77. [DOI: 10.1016/j.plipres.2011.12.005] [Citation(s) in RCA: 461] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
22
|
Loss of outer membrane protein C in Escherichia coli contributes to both antibiotic resistance and escaping antibody-dependent bactericidal activity. Infect Immun 2012; 80:1815-22. [PMID: 22354022 DOI: 10.1128/iai.06395-11] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Outer membrane proteins (OMPs) serve as the permeability channels for nutrients, toxins, and antibiotics. In Escherichia coli, OmpA has been shown to be involved in bacterial virulence, and OmpC is related to multidrug resistance. However, it is unclear whether OmpC also has a role in the virulence of E. coli. The aims of this study were to characterize the role of OmpC in antimicrobial resistance and bacterial virulence in E. coli. The ompC deletion mutant showed significantly decreased susceptibility to carbapenems and cefepime. To investigate the survival of E. coli exposed to the innate immune system, a human blood bactericidal assay showed that the ompC mutant increased survival in blood and serum but not in complement-inactivated serum. These effects were also demonstrated in the natural selection of OmpC mutants. Also, C1q interacted with E. coli through a complex of antibodies bound to OmpC as a major target. Bacterial survival was increased in the wild-type strain in a dose-dependent manner by adding free recombinant OmpC protein or anti-C1q antibody to human serum. These results demonstrated that the interaction of OmpC-specific antibody and C1q was the key step in initiating the antibody-dependent classical pathway for the clearance of OmpC-expressing E. coli. Anti-OmpC antibody was detected in human sera, indicating that OmpC is an immunogen. These data indicate that the loss of OmpC in E. coli is resistant to not only antibiotics, but also the serum bactericidal effect, which is mediated from the C1q and anti-OmpC antibody-dependent classical pathway.
Collapse
|
23
|
Lee TT, Chang CC, Juang RS, Chen RB, Yang HY, Chu LW, Wang SR, Tseng TH, Wang CS, Chen LJ, Yu B. Porcine lactoferrin expression in transgenic rice and its effects as a feed additive on early weaned piglets. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2010; 58:5166-5173. [PMID: 20345096 DOI: 10.1021/jf903904s] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
The purpose of this study is to determine the growth performance and immune characteristics of early weaned piglets receiving rice bran expressing porcine lactoferrin as a feed additive. Full-length cDNA encoding porcine lactoferrin (LF) driven by a rice actin promoter was transformed into rice plants, and its integration into the rice genome was verified by Southern blot analysis. The expression of recombinant LF (rLF) in whole grains and rice bran was also confirmed, and the amount of rLF accumulated in rice bran was estimated by immunoblot assay to be approximately 0.1% of rice bran weight. An iron-binding assay showed that the rLF retained iron-binding activity and the binding capacity of 1 mg/mL rLF would be saturated by 100 microM of FeCl(3). Thirty-six early weaned piglets at 21 days old were randomly selected into two groups and fed a diet containing 5% transgenic rice bran containing 50 mg/kg rLF (rLF group) and 5% rice bran (control group) to investigate the piglets' growth performance and immune characteristics. The results showed no significant difference in growth performance between the groups during the feeding period. However, the aerobic bacteria, anaerobic bacteria, and coliform counts in the cecal contents of the rLF-fed group were significantly lower than those of the control group. Additional immune characteristics such as the IgG concentration in the rLF group was higher than the control group at the 28th day, but leukocyte counts and the peripheral lymphocyte ratio remained similar. In summary, porcine LF expressed in rice bran, a byproduct of rice, can be used as a functional additive to improve antimicrobial capabilities and IgG concentration of early weaned piglets.
Collapse
Affiliation(s)
- Tzu-Tai Lee
- Department of Biotechnology, Ming-Dao University, Changhau, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Antibacterial activity of recombinant human lactoferrin from rice: effect of heat treatment. Biosci Biotechnol Biochem 2009; 73:1301-7. [PMID: 19502737 DOI: 10.1271/bbb.80814] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The antibacterial activity of recombinant human lactoferrin from rice (rhLF) compared with that of human milk lactoferrin (hLF) was evaluated against Escherichia coli O157:H7, Salmonella Enteritidis and Listeria monocytogenes. The hydrolysates of rhLF and hLF were found to be more active than native proteins against E. coli O157:H7, and their activity was independent of their iron saturation. The effect of different heat treatments on the antibacterial activity of apo-rhLF was studied and compared with hLF. We observed that an HTST pasteurization treatment did not affect the antimicrobial activity of lactoferrin against the pathogens studied. Furthermore, the activity of apo-rhLF and hLF against E. coli O157:H7 and L. monocytogenes in UHT milk and whey was assayed, finding a decrease in the number of bacteria, although lower than that observed in a broth medium. This study shows the similar antibacterial activity of rhLF and hLF which is important in order to consider the addition of rhLF as a supplement in special products.
Collapse
|
25
|
Rosso ML, Chauvaux S, Dessein R, Laurans C, Frangeul L, Lacroix C, Schiavo A, Dillies MA, Foulon J, Coppée JY, Médigue C, Carniel E, Simonet M, Marceau M. Growth of Yersinia pseudotuberculosis in human plasma: impacts on virulence and metabolic gene expression. BMC Microbiol 2008; 8:211. [PMID: 19055764 PMCID: PMC2631605 DOI: 10.1186/1471-2180-8-211] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Accepted: 12/03/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In man, infection by the Gram-negative enteropathogen Yersinia pseudotuberculosis is usually limited to the terminal ileum. However, in immunocompromised patients, the microorganism may disseminate from the digestive tract and thus cause a systemic infection with septicemia. RESULTS To gain insight into the metabolic pathways and virulence factors expressed by the bacterium at the blood stage of pseudotuberculosis, we compared the overall gene transcription patterns (the transcriptome) of bacterial cells cultured in either human plasma or Luria-Bertani medium. The most marked plasma-triggered metabolic consequence in Y. pseudotuberculosis was the switch to high glucose consumption, which is reminiscent of the acetogenic pathway (known as "glucose overflow") in Escherichia coli. However, upregulation of the glyoxylate shunt enzymes suggests that (in contrast to E. coli) acetate may be further metabolized in Y. pseudotuberculosis. Our data also indicate that the bloodstream environment can regulate major virulence genes (positively or negatively); the yadA adhesin gene and most of the transcriptional units of the pYV-encoded type III secretion apparatus were found to be upregulated, whereas transcription of the pH6 antigen locus was strongly repressed. CONCLUSION Our results suggest that plasma growth of Y. pseudotuberculosis is responsible for major transcriptional regulatory events and prompts key metabolic reorientations within the bacterium, which may in turn have an impact on virulence.
Collapse
Affiliation(s)
- Marie-Laure Rosso
- Inserm U801, Lille, F-59019, Université Lille II, Faculté de Médecine Henri Warembourg), Lille, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Oms38 is the first identified pore-forming protein in the outer membrane of relapsing fever spirochetes. J Bacteriol 2008; 190:7035-42. [PMID: 18757545 DOI: 10.1128/jb.00818-08] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Relapsing fever is a worldwide, endemic disease caused by several spirochetal species belonging to the genus Borrelia. During the recurring fever peaks, borreliae proliferate remarkably quickly compared to the slow dissemination of Lyme disease Borrelia and therefore require efficient nutrient uptake from the blood of their hosts. This study describes the identification and characterization of the first relapsing fever porin, which is present in the outer membranes of B. duttonii, B. hermsii, B. recurrentis, and B. turicatae. The pore-forming protein was purified by hydroxyapatite chromatography and designated Oms38, for outer membrane-spanning protein of 38 kDa. Biophysical characterization of Oms38 was done by using the black lipid bilayer method, demonstrating that Oms38 forms small, water-filled channels of 80 pS in 1 M KCl that did not exhibit voltage-dependent closure. The Oms38 channel is slightly selective for anions and shows a ratio of permeability for cations over anions of 0.41 in KCl. Analysis of the deduced amino acid sequences demonstrated that Oms38 contains an N-terminal signal sequence which is processed under in vivo conditions. Oms38 is highly conserved within the four studied relapsing fever species, sharing an overall amino acid identity of 58% and with a strong indication for the presence of amphipathic beta-sheets.
Collapse
|
27
|
COLAK HILAL, HAMPIKYAN HAMPARSUN, BINGOL ENVERBARIS, AKSU HARUN. THE EFFECT OF NISIN AND BOVINE LACTOFERRIN ON THE MICROBIOLOGICAL QUALITY OF TURKISH-STYLE MEATBALL (TEKIRDAĞ KÖFTE). J Food Saf 2008. [DOI: 10.1111/j.1745-4565.2008.00105.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
28
|
Lactoferrin Structure and Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 606:163-94. [DOI: 10.1007/978-0-387-74087-4_6] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
29
|
Min S, Krochta JM. Inhibition ofPenicillium communeby Edible Whey Protein Films Incorporating Lactoferrin, Lacto-ferrin Hydrolysate, and Lactoperoxidase Systems. J Food Sci 2006. [DOI: 10.1111/j.1365-2621.2005.tb07108.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
30
|
Kida K, Baba E, Torii R, Kawate N, Hatoya S, Wijewardana V, Sugiura K, Sawada T, Tamada H, Inaba T. Lactoferrin expression in the canine uterus during the estrous cycle and with pyometra. Theriogenology 2006; 66:1325-33. [PMID: 16730786 DOI: 10.1016/j.theriogenology.2006.04.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2006] [Accepted: 04/26/2006] [Indexed: 10/24/2022]
Abstract
The expression of lactoferrin, a non-specific antimicrobial defence, in the canine uterus during the normal estrous cycle and in bitches with pyometra was examined. Using polymerase chain reaction analysis, lactoferrin gene transcripts were detected in the endometrium at all stages of the estrous cycle, with the highest levels in estrus. In normal bitches, endometrial lactoferrin mRNA increased from proestrus to estrus (P<0.05). Thereafter, it dramatically decreased from estrus to Day 10 of diestrus (P<0.05), and stayed low at Day 35 of diestrus and anestrus; this was consistent with blood estrogen concentrations. Levels of lactoferrin mRNA were higher in bitches with pyometra than in normal diestrus (P<0.05). With immunohistochemistry, distinct staining of lactoferrin was detected in the luminal and glandular epithelial cells of the endometrium at proestrus and estrus, but little staining was detected at Day 10 of diestrus. At Day 35 of diestrus and anestrus, a partial and weak reaction was present in the same region. In bitches with pyometra, the glandular epithelial cells and many cells in the uterine stroma were strongly stained. Staining cells in the stroma were morphologically similar to neutrophils. No lactoferrin staining was seen in the uterine stromal cells or myometrium in any section. These results suggest that, in the canine uterus, lactoferrin expression is related to the blood concentration of estrogen, and that the dramatic reduction in lactoferrin observed at the early stage of diestrus may impair antimicrobial defense. Also, enhanced expression of lactoferrin mRNA in the endometrium with pyometra may be associated with neutrophil invasion into the uterus to combat the infection.
Collapse
Affiliation(s)
- K Kida
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka 599-8531, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Hunter HN, Demcoe AR, Jenssen H, Gutteberg TJ, Vogel HJ. Human lactoferricin is partially folded in aqueous solution and is better stabilized in a membrane mimetic solvent. Antimicrob Agents Chemother 2005; 49:3387-95. [PMID: 16048952 PMCID: PMC1196233 DOI: 10.1128/aac.49.8.3387-3395.2005] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lactoferricins are highly basic bioactive peptides that are released in the stomach through proteolytic cleavage of various lactoferrin proteins. Here we have determined the solution structure of human lactoferricin (LfcinH) by conventional two-dimensional nuclear magnetic resonance methods in both aqueous solution and a membrane mimetic solvent. Unlike the 25-residue bovine lactoferricin (LfcinB), which adopts a somewhat distorted antiparallel beta sheet, the longer LfcinH peptide shows a helical content from Gln14 to Lys29 in the membrane mimetic solvent but a nonexistent beta-sheet character in either the N- or C-terminal regions of the peptide. The helical characteristic of the LfcinH peptide resembles the conformation that this region adopts in the crystal structure of the intact protein. The LfcinH structure determined in aqueous solution displays a nascent helix in the form of a coiled conformation in the region from Gln14 to Lys29. Numerous hydrophobic interactions create the basis for the better-defined overall structure observed in the membrane mimetic solvent. The 49-residue LfcinH peptide isolated for these studies was found to be slightly longer than previously reported peptide preparations and was found to have an intact peptide bond between residues Ala11 and Val12. The distinct solution structures of LfcinH and LfcinB represent a novel difference in the physical properties of these two peptides, which contributes to their unique physiological activities.
Collapse
Affiliation(s)
- Howard N Hunter
- Department of Biological Sciences, University of Calgary, 2500 University N.W., Calgary, Alberta, Canada
| | | | | | | | | |
Collapse
|
32
|
Sundara Baalaji N, Acharya KR, Singh TP, Krishnaswamy S. High-resolution diffraction from crystals of a membrane-protein complex: bacterial outer membrane protein OmpC complexed with the antibacterial eukaryotic protein lactoferrin. Acta Crystallogr Sect F Struct Biol Cryst Commun 2005; 61:773-5. [PMID: 16511154 PMCID: PMC1952365 DOI: 10.1107/s1744309105022086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2005] [Accepted: 07/08/2005] [Indexed: 11/10/2022]
Abstract
Crystals of the complex formed between the outer membrane protein OmpC from Escherichia coli and the eukaryotic antibacterial protein lactoferrin from Camelus dromedarius (camel) have been obtained using a detergent environment. Initial data processing suggests that the crystals belong to the hexagonal space group P6, with unit-cell parameters a = b = 116.3, c = 152.4 A, alpha = beta = 90, gamma = 120 degrees. This indicated a Matthews coefficient (VM) of 3.3 A3 Da(-1), corresponding to a possible molecular complex involving four molecules of lactoferrin and two porin trimers in the unit cell (4832 amino acids; 533.8 kDa) with 63% solvent content. A complete set of diffraction data was collected to 3 A resolution at 100 K. Structure determination by molecular replacement is in progress. Structural study of this first surface-exposed membrane-protein complex with an antibacterial protein will provide insights into the mechanism of action of OmpC as well as lactoferrin.
Collapse
Affiliation(s)
- N. Sundara Baalaji
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai 625021, India
| | - K. Ravi Acharya
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, England
| | - T. P. Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - S. Krishnaswamy
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai 625021, India
- Correspondence e-mail: ,
| |
Collapse
|
33
|
|
34
|
Viejo-Díaz M, Andrés MT, Fierro JF. Modulation of in vitro fungicidal activity of human lactoferrin against Candida albicans by extracellular cation concentration and target cell metabolic activity. Antimicrob Agents Chemother 2004; 48:1242-8. [PMID: 15047526 PMCID: PMC375254 DOI: 10.1128/aac.48.4.1242-1248.2004] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The anti-Candida activity of the innate defense protein human lactoferrin was investigated. Lactoferrin displayed a clear fungicidal effect against Candida albicans only under low-strength conditions. This candidacidal activity was inversely correlated with the extracellular concentration of the monovalent cations and was prevented by Na(+) and K(+) (> or 30 mM) and by divalent cations (Ca(2+) and Mg(2+) at > or 4 mM). A slight cellular release of K(+), cytosolic acidification, and a change in the membrane potential were observed in C. albicans cells treated with lactoferrin, suggesting that this protein directly or indirectly interacts with the cytoplasmic membrane. Mitochondrial inhibitors (carbonyl cyanide m-chlorophenylhydrazone, 2,4-dinitrophenol, azide, and antimycin) as well as anaerobic conditions significantly reduced the killing effect of lactoferrin. These results suggest that low-strength conditions and the cellular metabolic state may modulate the candidacidal activity of human lactoferrin.
Collapse
Affiliation(s)
- Mónica Viejo-Díaz
- Department of Functional Biology (Microbiology), Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
| | | | | |
Collapse
|
35
|
Farnaud S, Spiller C, Moriarty L, Patel A, Gant V, Odell EW, Evans R. Interactions of lactoferricin-derived peptides with LPS and antimicrobial activity. FEMS Microbiol Lett 2004. [DOI: 10.1111/j.1574-6968.2004.tb09482.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
36
|
Velliyagounder K, Kaplan JB, Furgang D, Legarda D, Diamond G, Parkin RE, Fine DH. One of two human lactoferrin variants exhibits increased antibacterial and transcriptional activation activities and is associated with localized juvenile periodontitis. Infect Immun 2003; 71:6141-7. [PMID: 14573629 PMCID: PMC219577 DOI: 10.1128/iai.71.11.6141-6147.2003] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The iron-binding protein lactoferrin is a ubiquitous and abundant constituent of human exocrine secretions. Lactoferrin inhibits bacterial growth by sequestering essential iron and also exhibits non-iron-dependent antibacterial, antifungal, antiviral, antitumor, anti-inflammatory, and immunoregulatory activities. All of these non-iron-dependent activities are mediated by the highly charged N terminus of lactoferrin. In this study we characterized a Lys/Arg polymorphism at position 29 in the N-terminal region of human lactoferrin that results from a single nucleotide polymorphism in exon 1 of the human lactoferrin gene. We expressed cDNAs encoding both lactoferrin variants in insect cells and purified the two proteins by ion exchange chromatography. The two lactoferrin variants exhibited nearly identical iron-binding and iron-releasing activities and equivalent bactericidal activities against a strain of the gram-negative bacterium Actinobacillus actinomycetemcomitans. When tested against the gram-positive species Streptococcus mutans and Streptococcus mitis, however, lactoferrin containing Lys at position 29 exhibited significantly greater bactericidal activity than did lactoferrin containing Arg. In addition, the Lys-containing lactoferrin stimulated bovine tracheal epithelial cells to synthesize much higher levels of tracheal antimicrobial peptide mRNA than did the Arg-containing variant. A genotyping assay that distinguished between the two alleles based on a polymorphic EarI restriction site showed that the Lys and Arg alleles had frequencies of 24% and 76%, respectively, among 17 healthy human subjects, and 72% and 28%, respectively, among nine patients with localized juvenile periodontitis. Our findings suggest that these two lactoferrin variants are functionally different and that these differences may contribute to the pathogenesis of localized juvenile periodontitis.
Collapse
Affiliation(s)
- Kabilan Velliyagounder
- Department of Oral Biology, New Jersey Dental School, Graduate School of Biomedical Sciences, University of Medicine and Dentistry of New Jersey, Newark, New Jersey 07103, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Murdock CA, Matthews KR. Antibacterial activity of pepsin-digested lactoferrin on foodborne pathogens in buffered broth systems and ultra-high temperature milk with EDTA. J Appl Microbiol 2003; 93:850-6. [PMID: 12392532 DOI: 10.1046/j.1365-2672.2002.01762.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
AIMS To evaluate the antimicrobial activity in peptone yeast extract glucose (PYG) broth and ultra-high temperature (UHT) milk of bovine lactoferrin hydrolysate (LFH) with pepsin against the foodborne pathogens Salmonella Stanley, Escherichia coli, Listeria monocytogenes and Staphylococcus aureus. METHODS AND RESULTS The LFH was suspended in PYG and the minimum inhibitory concentration for each pathogen determined. The LFH was also suspended in UHT milk adjusted to pH 4 or 7, samples incubated at 4 or 35 degrees C and the change in bacterial cell population determined. Experiments in UHT milk were conducted using L. monocytogenes and E. coli O157:H7. At pH 4 LFH reduced the population of E. coli O157:H7 and L. monocytogenes by approx. 2 log; however, only E. coli O157:H7 was inhibited in samples adjusted to pH 7. The addition of EDTA (10 mg ml(-1)) to UHT milk supplemented with LFH did not markedly influence the growth of E. coli O157:H7 or L. monocytogenes. CONCLUSIONS The results suggest that, under low pH and refrigeration conditions, LFH can limit the growth or reduce the population of pathogenic bacteria in a dairy product. SIGNIFICANCE AND IMPACT OF THE STUDY Natural preservatives that are active against Gram-negative and Gram-positive bacteria are desirable to the food industry. This study demonstrates that LFH is effective in a complex food system. Moreover, the LFH used was not purified, making its use by industry more attractive.
Collapse
Affiliation(s)
- C A Murdock
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901-8520, USA
| | | |
Collapse
|
38
|
Ostberg Y, Pinne M, Benz R, Rosa P, Bergström S. Elimination of channel-forming activity by insertional inactivation of the p13 gene in Borrelia burgdorferi. J Bacteriol 2002; 184:6811-9. [PMID: 12446631 PMCID: PMC135472 DOI: 10.1128/jb.184.24.6811-6819.2002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
P13 is a chromosomally encoded 13-kDa integral outer membrane protein of the Lyme disease agent, Borrelia burgdorferi. The aim of this study was to investigate the function of the P13 protein. Here, we inactivated the p13 gene by targeted mutagenesis and investigated the porin activities of outer membrane proteins by using lipid bilayer experiments. Channel-forming activity was lost in the p13 mutant compared to wild-type B. burgdorferi, indicating that P13 may function as a porin. We purified native P13 to homogeneity by fast performance liquid chromatography and demonstrated that pure P13 has channel-forming activity with a single-channel conductance in 1 M KCl of 3.5 nS, the same as the porin activity that was lost in the p13 mutant. Further characterization of the channel formed by P13 suggested that it is cation selective and voltage independent. In addition, no major physiological effects of the inactivated p13 gene could be detected under normal growth conditions. The inactivation of p13 is the first reported inactivation of a gene encoding an integral outer membrane protein in B. burgdorferi. Here, we describe both genetic and biophysical experiments indicating that P13 in B. burgdorferi is an outer membrane protein with porin activity.
Collapse
Affiliation(s)
- Yngve Ostberg
- Department of Molecular Biology, Umeå University, Sweden
| | | | | | | | | |
Collapse
|
39
|
Gomez HF, Ochoa TJ, Herrera-Insua I, Carlin LG, Cleary TG. Lactoferrin protects rabbits from Shigella flexneri-induced inflammatory enteritis. Infect Immun 2002; 70:7050-3. [PMID: 12438385 PMCID: PMC132992 DOI: 10.1128/iai.70.12.7050-7053.2002] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Shigella species cause bacillary dysentery in humans by invasion, intracellular multiplication, spread to adjacent cells, and induction of brisk inflammatory responses in the intestinal epithelium. In vitro data suggest that lactoferrin, a glycoprotein present in human mucosal secretions, has a role in protection from bacterial enteric infections. We sought to determine the activity of lactoferrin in vivo, using the concentration present in human colostrum, to investigate its effect on the development of clinical and pathological evidence of inflammation in a rabbit model of enteritis. Lactoferrin protected rabbits infected with Shigella flexneri from developing inflammatory intestinal disease. Typical histological changes in ill animals included villous blunting with sloughing of epithelial cells, submucosal edema, infiltration of leukocytes, venous congestion, and hemorrhage. Lactoferrin at a concentration normally found in human colostrum blocks development of S. flexneri-induced inflammatory enteritis.
Collapse
Affiliation(s)
- Henry F Gomez
- Division of Infectious Diseases, Department of Pediatrics, University of Texas-Houston Medical School, 77030, USA
| | | | | | | | | |
Collapse
|
40
|
The lipid bilayer concept and its experimental realization: from soap bubbles, kitchen sink, to bilayer lipid membranes. J Memb Sci 2001. [DOI: 10.1016/s0376-7388(01)00394-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
41
|
Deriy LV, Chor J, Thomas LL. Surface expression of lactoferrin by resting neutrophils. Biochem Biophys Res Commun 2000; 275:241-6. [PMID: 10944471 DOI: 10.1006/bbrc.2000.3284] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We examined the surface expression of lactoferrin by human neutrophils. Western blot analysis with anti-lactoferrin antibodies demonstrated the presence of a 78- to 79-kDa band in plasma membranes isolated from resting neutrophils that corresponded to the 78- to 79-kDa protein in neutrophil secondary granules. Flow cytometry using FITC-conjugated anti-lactoferrin antibodies confirmed that lactoferrin is expressed on the neutrophil surface. Preincubating the neutrophils in acidic (pH 3.9) buffer did not alter staining of the cells by the antibodies. Surface expression of lactoferrin was also detected on neutrophils in whole blood. Neutrophil activation by C5a or the calcium ionophore A23187 did not increase the surface expression of lactoferrin. Instead, the level of lactoferrin expression detected with one of two monoclonal antibodies was diminished after neutrophil activation, suggesting a possible conformational change in the lactoferrin. The surface-expressed lactoferrin may provide a mechanism for the interaction between lactoferrin-binding microorganisms and neutrophils.
Collapse
Affiliation(s)
- L V Deriy
- Department of Immunology/Microbiology, Rush-Presbyterian-St. Luke's Medical Center, 1653 W. Congress Parkway, Chicago, Illinois, 60612, USA
| | | | | |
Collapse
|
42
|
Recio I, Visser S. Antibacterial and binding characteristics of bovine, ovine and caprine lactoferrins: a comparative study. Int Dairy J 2000. [DOI: 10.1016/s0958-6946(00)00089-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|