1
|
Lou J, Wu X, Ji W, Yu J, Xu Y, Xiao W, Lu W, Xin K, Chen T, Tang Q, Liang G, Gao Y, Wu D. N-Terminal random curl-tandam α-helical peptide 7W: A potent antibacterial and anti-inflammatory dual-effect agent through tryptophan substitution. Eur J Med Chem 2025; 292:117686. [PMID: 40319576 DOI: 10.1016/j.ejmech.2025.117686] [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: 01/10/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
This study investigates the impact of tryptophan substitution on the properties of the Medisin family peptide MS-PT. By substituting hydrophobic amino acids in MS-PT1 with tryptophan, a series of derivative peptides were synthesized. Among them, the 7W peptide stood out with its unique N-terminal random curl and α-helix structure. In vitro, 7W effectively inhibited the secretion of pro-inflammatory cytokines like IL-6 and TNF-α in LPS-induced Membrane-Proximal Macrophages (MPMs) by blocking the MAPK/NF-κB signaling pathway. It also exhibited stronger antimicrobial activity against Gram-positive bacteria compared to the parent peptide MS-PT1, with good safety as indicated by a low hemolysis rate. In vivo, in the CLP-induced sepsis mouse model, 7W alleviated lung and liver injury, suppressed the expression of inflammatory factors in serum and tissues, and had a relatively long plasma half-life of 46.8 h. Mechanistically, 7W interacted preferentially with bacterial mimic membranes and LPS, and its anti-inflammatory effect might be mediated by binding to TLR4. These findings not only clarify the role of tryptophan substitution in modulating peptide properties but also offer a new strategy for the development of multifunctional antimicrobial peptides, suggesting that 7W has great potential as a therapeutic agent for sepsis and other inflammatory diseases.
Collapse
Affiliation(s)
- Jietao Lou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xinyi Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China; Department of Pharmacy, The First People's Hospital of Jiande, Hangzhou, 311600, China
| | - Wenwen Ji
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jiaye Yu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yanyan Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wanyang Xiao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China; College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Weijie Lu
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Kaiyun Xin
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Tianbao Chen
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Qidong Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310053, China
| | - Yitian Gao
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| | - Di Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
2
|
Moita T, Pedroso L, Santos I, Lima A. Casein and Casein-Derived Peptides: Antibacterial Activities and Applications in Health and Food Systems. Nutrients 2025; 17:1615. [PMID: 40431356 PMCID: PMC12114164 DOI: 10.3390/nu17101615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
The growing threat of antimicrobial resistance has intensified the search for alternative strategies to conventional antibiotics and preservatives. Casein-derived antimicrobial peptides (CDAMPs), generated through proteolysis, exhibit potent activity against a broad spectrum of pathogens, including antibiotic-resistant strains, revealing strong potential as natural preservatives and therapeutic agents in food and medical applications. Furthermore, casein can be an ideal source for peptide production in these sectors due to its abundance, disordered structure, which enhances enzymatic cleavage, and its amino acid profile, which favors bioactivity. Nonetheless, there is limited literature addressing real-life applications in veterinary medicine, food safety, and public health. This review provides a structured synthesis of current knowledge on the antibacterial properties of CDPs. We classify the main types of these peptides, describe their production methods, and summarize their mechanisms of action against Gram-positive and Gram-negative bacteria. Furthermore, we examine their potential applications in clinical, veterinary, and food-related contexts, and discuss key aspects related to delivery systems, safety, and regulatory considerations. Overall, our findings highlight the potential of CDPs in addressing antimicrobial resistance, reducing antibiotic use in livestock and humans, and contributing to sustainable food safety and functional food production.
Collapse
Affiliation(s)
- Tomás Moita
- Research in Veterinary Medicine (I-MVET), Faculty of Veterinary Medicine, Lusófona University, Lisbon University Centre, Campo Grande, 376, 1749-024 Lisbon, Portugal; (T.M.); (L.P.); (I.S.)
| | - Laurentina Pedroso
- Research in Veterinary Medicine (I-MVET), Faculty of Veterinary Medicine, Lusófona University, Lisbon University Centre, Campo Grande, 376, 1749-024 Lisbon, Portugal; (T.M.); (L.P.); (I.S.)
- Veterinary and Animal Research Centre (CECAV), Faculty of Veterinary Medicine, Lusófona University, Lisbon University Centre, 1749-024 Lisbon, Portugal
- IPLUSO—Polytechnic Institute of Lusofonia, School of Health, Protection and Animal Welfare, Campo Grande 400, 1700-098 Lisbon, Portugal
| | - Isabel Santos
- Research in Veterinary Medicine (I-MVET), Faculty of Veterinary Medicine, Lusófona University, Lisbon University Centre, Campo Grande, 376, 1749-024 Lisbon, Portugal; (T.M.); (L.P.); (I.S.)
- Veterinary and Animal Research Centre (CECAV), Faculty of Veterinary Medicine, Lusófona University, Lisbon University Centre, 1749-024 Lisbon, Portugal
| | - Ana Lima
- Research in Veterinary Medicine (I-MVET), Faculty of Veterinary Medicine, Lusófona University, Lisbon University Centre, Campo Grande, 376, 1749-024 Lisbon, Portugal; (T.M.); (L.P.); (I.S.)
- Veterinary and Animal Research Centre (CECAV), Faculty of Veterinary Medicine, Lusófona University, Lisbon University Centre, 1749-024 Lisbon, Portugal
- IPLUSO—Polytechnic Institute of Lusofonia, School of Health, Protection and Animal Welfare, Campo Grande 400, 1700-098 Lisbon, Portugal
| |
Collapse
|
3
|
Amann V, Kissmann AK, Firacative C, Rosenau F. Biofilm-Associated Candidiasis: Pathogenesis, Prevalence, Challenges and Therapeutic Options. Pharmaceuticals (Basel) 2025; 18:460. [PMID: 40283897 PMCID: PMC12030374 DOI: 10.3390/ph18040460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/11/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
The rising prevalence of fungal infections, especially those caused by Candida species, presents a major risk to global health. With approximately 1.5 million deaths annually, the urgency for effective treatment options has never been greater. Candida spp. are the leading cause of invasive infections, significantly impacting immunocompromised patients and those in healthcare settings. C. albicans, C. parapsilosis and the emerging species C. auris are categorized as highly dangerous species because of their pathogenic potential and increasing drug resistance. This review comparatively describes the formation of microbial biofilms of both bacterial and fungal origin, including major pathogens, thereby creating a novel focus. Biofilms can further complicate treatment, as these structures provide enhanced resistance to antifungal therapies. Traditional antifungal agents, including polyenes, azoles and echinocandins, have shown effectiveness, yet resistance development continues to rise, necessitating the exploration of novel therapeutic approaches. Antimicrobial peptides (AMPs) such as the anti-biofilm peptides Pom-1 and Cm-p5 originally isolated from snails represent promising candidates due to their unique mechanisms of action and neglectable cytotoxicity. This review article discusses the challenges posed by Candida infections, the characteristics of important species, the role of biofilms in virulence and the potential of new therapeutic options like AMPs.
Collapse
Affiliation(s)
- Valerie Amann
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| | - Ann-Kathrin Kissmann
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| | - Carolina Firacative
- Studies in Translational Microbiology and Emerging Diseases (MICROS) Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogota 111221, Colombia;
| | - Frank Rosenau
- Institute of Pharmaceutical Biotechnology, Ulm University, 89081 Ulm, Germany; (V.A.); (A.-K.K.)
| |
Collapse
|
4
|
Zhang Q, Choi K, Wang X, Xi L, Lu S. The Contribution of Human Antimicrobial Peptides to Fungi. Int J Mol Sci 2025; 26:2494. [PMID: 40141139 PMCID: PMC11941821 DOI: 10.3390/ijms26062494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/07/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Various species of fungi can be detected in the environment and within the human body, many of which may become pathogenic under specific conditions, leading to various forms of fungal infections. Antimicrobial peptides (AMPs) are evolutionarily ancient components of the immune response that are quickly induced in response to infections with many pathogens in almost all tissues. There is a wide range of AMP classes in humans, many of which exhibit broad-spectrum antimicrobial function. This review provides a comprehensive overview of the mechanisms of action of AMPs, their distribution in the human body, and their antifungal activity against a range of both common and rare clinical fungal pathogens. It also discusses the current research status of promising novel antifungal strategies, highlighting the challenges that must be overcome in the development of these therapies. The hope is that antimicrobial peptides, as a class of antimicrobial agents, will soon progress through large-scale clinical trials and be implemented in clinical practice, offering new treatment options for patients suffering from infections.
Collapse
Affiliation(s)
| | | | | | | | - Sha Lu
- Department of Dermatology and Venereology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, #107 Yanjiang West Rd., Guangzhou 510120, China; (Q.Z.); (K.C.); (X.W.); (L.X.)
| |
Collapse
|
5
|
Hetta HF, Sirag N, Alsharif SM, Alharbi AA, Alkindy TT, Alkhamali A, Albalawi AS, Ramadan YN, Rashed ZI, Alanazi FE. Antimicrobial Peptides: The Game-Changer in the Epic Battle Against Multidrug-Resistant Bacteria. Pharmaceuticals (Basel) 2024; 17:1555. [PMID: 39598464 PMCID: PMC11597525 DOI: 10.3390/ph17111555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
The rapid progress of antibiotic resistance among bacteria has prompted serious medical concerns regarding how to manage multidrug-resistant (MDR) bacterial infections. One emerging strategy to combat antibiotic resistance is the use of antimicrobial peptides (AMPs), which are amino acid chains that act as broad-spectrum antimicrobial molecules and are essential parts of the innate immune system in mammals, fungi, and plants. AMPs have unique antibacterial mechanisms that offer benefits over conventional antibiotics in combating drug-resistant bacterial infections. Currently, scientists have conducted multiple studies on AMPs for combating drug-resistant bacterial infections and found that AMPs are a promising alternative to conventional antibiotics. On the other hand, bacteria can develop several tactics to resist and bypass the effect of AMPs. Therefore, it is like a battle between the bacterial community and the AMPs, but who will win? This review provides thorough insights into the development of antibiotic resistance as well as detailed information about AMPs in terms of their history and classification. Furthermore, it addresses the unique antibacterial mechanisms of action of AMPs, how bacteria resist these mechanisms, and how to ensure AMPs win this battle. Finally, it provides updated information about FDA-approved AMPs and those that were still in clinical trials. This review provides vital information for researchers for the development and therapeutic application of novel AMPs for drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Helal F. Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (H.F.H.); (A.A.A.); (T.T.A.)
| | - Nizar Sirag
- Division of Pharmacognosy, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Shumukh M. Alsharif
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (H.F.H.); (A.A.A.); (T.T.A.)
| | - Ahmad A. Alharbi
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (H.F.H.); (A.A.A.); (T.T.A.)
| | - Tala T. Alkindy
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (H.F.H.); (A.A.A.); (T.T.A.)
| | - Alanoud Alkhamali
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (A.A.); (A.S.A.)
| | - Abdullah S. Albalawi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (A.A.); (A.S.A.)
| | - Yasmin N. Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt; (Y.N.R.); (Z.I.R.)
| | - Zainab I. Rashed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt; (Y.N.R.); (Z.I.R.)
| | - Fawaz E. Alanazi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| |
Collapse
|
6
|
Rizvi SFA, Zhang H, Fang Q. Engineering peptide drug therapeutics through chemical conjugation and implication in clinics. Med Res Rev 2024; 44:2420-2471. [PMID: 38704826 DOI: 10.1002/med.22046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/21/2024] [Accepted: 04/21/2024] [Indexed: 05/07/2024]
Abstract
The development of peptide drugs has made tremendous progress in the past few decades because of the advancements in modification chemistry and analytical technologies. The novel-designed peptide drugs have been modified through various biochemical methods with improved diagnostic, therapeutic, and drug-delivery strategies. Researchers found it a helping hand to overcome the inherent limitations of peptides and bring continued advancements in their applications. Furthermore, the emergence of peptide-drug conjugates (PDCs)-utilizes target-oriented peptide moieties as a vehicle for cytotoxic payloads via conjugation with cleavable chemical agents, resulting in the key foundation of the new era of targeted peptide drugs. This review summarizes the various classifications of peptide drugs, suitable chemical modification strategies to improve the ADME (adsorption, distribution, metabolism, and excretion) features of peptide drugs, and recent (2015-early 2024) progress/achievements in peptide-based drug delivery systems as well as their fruitful implication in preclinical and clinical studies. Furthermore, we also summarized the brief description of other types of PDCs, including peptide-MOF conjugates and peptide-UCNP conjugates. The principal aim is to provide scattered and diversified knowledge in one place and to help researchers understand the pinching knots in the science of PDC development and progress toward a bright future of novel peptide drugs.
Collapse
Affiliation(s)
- Syed Faheem Askari Rizvi
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Haixia Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Quan Fang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
7
|
Oliver-Cervelló L, López-Gómez P, Martin-Gómez H, Marion M, Ginebra MP, Mas-Moruno C. Functionalization of Alginate Hydrogels with a Multifunctional Peptide Supports Mesenchymal Stem Cell Adhesion and Reduces Bacterial Colonization. Chemistry 2024; 30:e202400855. [PMID: 39031737 DOI: 10.1002/chem.202400855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/07/2024] [Accepted: 06/19/2024] [Indexed: 07/22/2024]
Abstract
Hydrogels with cell adhesive moieties stand out as promising materials to enhance tissue healing and regeneration. Nonetheless, bacterial infections of the implants represent an unmet major concern. In the present work, we developed an alginate hydrogel modified with a multifunctional peptide containing the RGD cell adhesive motif in combination with an antibacterial peptide derived from the 1-11 region of lactoferrin (LF). The RGD-LF branched peptide was successfully anchored to the alginate backbone by carbodiimide chemistry, as demonstrated by 1H NMR and fluorescence measurements. The functionalized hydrogel presented desirable physicochemical properties (porosity, swelling and rheological behavior) to develop biomaterials for tissue engineering. The viability of mesenchymal stem cells (MSCs) on the peptide-functionalized hydrogels was excellent, with values higher than 85 % at day 1, and higher than 95 % after 14 days in culture. Moreover, the biological characterization demonstrated the ability of the hydrogels to significantly enhance ALP activity of MSCs as well as to decrease bacterial colonization of both Gram-positive and Gram-negative models. Such results prove the potential of the functionalized hydrogels as novel biomaterials for tissue engineering, simultaneously displaying cell adhesive activity and the capacity to prevent bacterial contamination, a dual bioactivity commonly not found for these types of hydrogels.
Collapse
Affiliation(s)
- Lluís Oliver-Cervelló
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
| | - Patricia López-Gómez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
| | - Helena Martin-Gómez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
| | - Mahalia Marion
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, 08028, Spain
| | - Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| |
Collapse
|
8
|
Krupińska AM, Bogucki Z. Lactoferrin as a potential therapeutic for the treatment of Candida-associated denture stomatitis. J Oral Biosci 2024; 66:308-313. [PMID: 38777122 DOI: 10.1016/j.job.2024.05.007] [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: 04/16/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND The use of prostheses in the oral cavity creates favorable conditions for Candida colonization, which may subsequently lead to Candida-associated denture stomatitis (CADS). Due to its many contributing factors and frequent relapses, CADS is difficult to manage. Given the rise in drug resistance among fungal species, it is critical to develop new therapeutic approaches, reduce the required dosage of medications, and minimize the toxicity and side effects of therapy. HIGHLIGHT Salivary lactoferrin, a multifunctional glycoprotein, is thought to be the first line of defense against microbial invasion of mucosal surfaces. CONCLUSION Current research emphasizes the capability of lactoferrin and its derivatives to eliminate a broad spectrum of Candida species. It may be an appealing option for use in monotherapy or in combination with common medications for oral stomatitis treatment. This review provides an overview of the current understanding of lactoferrin's anti-fungal effects in oral candidiasis.
Collapse
Affiliation(s)
| | - Zdzisław Bogucki
- Department and Division of Dental Prosthetics, Wroclaw Medical University, Wyb. Ludwika Pasteura 1, 50-367, Wrocław, Poland
| |
Collapse
|
9
|
Tan T, Hou Y, Zhang Y, Wang B. Double-Network Hydrogel with Strengthened Mechanical Property for Controllable Release of Antibacterial Peptide. Biomacromolecules 2024; 25:1850-1860. [PMID: 38416425 DOI: 10.1021/acs.biomac.3c01290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Developing double-network (DN) hydrogels with high mechanical properties and antibacterial efficacy to combat multidrug-resistant bacterial infections and serve as scaffolds for cell culture still remains an ongoing challenge. In this study, an ion-responsive antibacterial peptide (AMP) (C16-WIIIKKK, termed as IK7) was synergistically combined with a photoresponsive gelatin methacryloyl (GelMA) polymer to fabricate a biocompatible DN hydrogel. The GelMA-IK7 DN hydrogel showed enhanced mechanical properties in contrast to the individual IK7 and GelMA hydrogels and demonstrated substantial antibacterial efficacy. Further investigations revealed that the DN hydrogel effectively inhibited bacterial growth by the controlled and sustained release of the IK7 peptide. In addition, the formation of the DN hydrogel was also found to protect AMP IK7 from rapid degradation by proteinase K. Our findings suggested that the developed GelMA-IK7 DN hydrogel holds great potential for next-generation antibacterial hydrogels for three-dimensional cell culture and tissue regeneration.
Collapse
Affiliation(s)
- Tingyuan Tan
- Research Institute of Interdisciplinary Sciences & School of Materials Science and Engineering, Dongguan University of Technology, Dongguan 523808, China
| | - Yangqian Hou
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Yi Zhang
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Biao Wang
- Research Institute of Interdisciplinary Sciences & School of Materials Science and Engineering, Dongguan University of Technology, Dongguan 523808, China
- School of Physics, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
10
|
Azadi S, Yazdanpanah MA, Afshari A, Alahdad N, Chegeni S, Angaji A, Rezayat SM, Tavakol S. Bioinspired synthetic peptide-based biomaterials regenerate bone through biomimicking of extracellular matrix. J Tissue Eng 2024; 15:20417314241303818. [PMID: 39670180 PMCID: PMC11635874 DOI: 10.1177/20417314241303818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
There have been remarkable advancements in regenerative medicine for bone regeneration, tackling the worldwide health concern of tissue loss. Tissue engineering uses the body's natural capabilities and applies biomaterials and bioactive molecules to replace damaged or lost tissues and restore their functionality. While synthetic ceramics have overcome some challenges associated with allografts and xenografts, they still need essential growth factors and biomolecules. Combining ceramics and bioactive molecules, such as peptides derived from biological motifs of vital proteins, is the most effective approach to achieve optimal bone regeneration. These bioactive peptides induce various cellular processes and modify scaffold properties by mimicking the function of natural osteogenic, angiogenic and antibacterial biomolecules. The present review aims to consolidate the latest and most pertinent information on the advancements in bioactive peptides, including angiogenic, osteogenic, antimicrobial, and self-assembling peptide nanofibers for bone tissue regeneration, elucidating their biological effects and potential clinical implications.
Collapse
Affiliation(s)
- Sareh Azadi
- Department of Medical Biotechnology, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Yazdanpanah
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Ali Afshari
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Niloofar Alahdad
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Solmaz Chegeni
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Abdolhamid Angaji
- Department of Medical Biotechnology, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Research and Development, Tavakol Biomimetic Technologies Company, Tehran, Iran
| |
Collapse
|
11
|
Machado M, Silva S, Costa EM. Are Antimicrobial Peptides a 21st-Century Solution for Atopic Dermatitis? Int J Mol Sci 2023; 24:13460. [PMID: 37686269 PMCID: PMC10488019 DOI: 10.3390/ijms241713460] [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: 08/01/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disorder that is the result of various environmental, bacterial and genetic stimuli, which culminate in the disruption of the skin's barrier function. Characterized by highly pruritic skin lesions, xerosis and an array of comorbidities among which skin infections are the most common, this condition results in both a significant loss of quality of life and in the need for life-long treatments (e.g., corticosteroids, monoclonal antibodies and regular antibiotic intake), all of which may have harmful secondary effects. This, in conjunction with AD's rising prevalence, made the development of alternative treatment strategies the focus of both the scientific community and the pharmaceutical industry. Given their potential to both manage the skin microbiome, fight infections and even modulate the local immune response, the use of antimicrobial peptides (AMPs) from more diverse origins has become one of the most promising alternative solutions for AD management, with some being already used with some success towards this end. However, their production and use also exhibit some limitations. The current work seeks to compile the available information and provide a better understanding of the state of the art in the understanding of AMPs' true potential in addressing AD.
Collapse
Affiliation(s)
| | - Sara Silva
- CBQF Centro de Biotecnologia e Química Fina Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal;
| | - Eduardo M. Costa
- CBQF Centro de Biotecnologia e Química Fina Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal;
| |
Collapse
|
12
|
Chen N, Jiang C. Antimicrobial peptides: Structure, mechanism, and modification. Eur J Med Chem 2023; 255:115377. [PMID: 37099837 DOI: 10.1016/j.ejmech.2023.115377] [Citation(s) in RCA: 97] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/28/2023]
Affiliation(s)
- Na Chen
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Cheng Jiang
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China.
| |
Collapse
|
13
|
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: 0.5] [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
|
14
|
Trivedi A, Maheshwari R, Tarnow-Mordi WO, Saxena N. Lactoferrin for the postoperative management of term neonates after gastrointestinal surgery. Cochrane Database Syst Rev 2023; 5:CD012218. [PMID: 37233609 PMCID: PMC10215284 DOI: 10.1002/14651858.cd012218.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
BACKGROUND Neonates who have undergone gastrointestinal surgery are particularly susceptible to infectious complications in the postoperative period. This may be due in part to disruption of the integrity of the gut and its altered intestinal microflora. Lactoferrin is a whey protein found in milk and is an important innate mammalian defence mechanism. Lactoferrin has been reported to have antimicrobial and anti-inflammatory properties. It has also been reported to help establish a healthy gut microflora and aid in the intestinal immune system. Lactoferrin supplementation has been reported to decrease sepsis in preterm infants. There may be a role for lactoferrin to reduce the incidence of sepsis, thus reducing morbidity and mortality and improving enteral feeding in postoperative term neonates. OBJECTIVES The primary objective of this review was to evaluate the efficacy of administering lactoferrin on the incidence of sepsis and mortality in term neonates after gastrointestinal surgery. The secondary objective was to assess the impact of administering lactoferrin on time to full enteral feeds, the intestinal microflora, duration of hospital stay, and mortality before discharge in the same population. SEARCH METHODS The Cochrane Neonatal Information Specialist searched the Cochrane Central Register of Controlled Trials (CENTRAL), Ovid MEDLINE, Embase Ovid, CINAHL, the WHO ICTRP and ClinicalTrials.gov trials registries. The date of the last search was February 2023. There were no restrictions to language, publication year or publication type. We checked references of potentially relevant studies and systematic reviews. SELECTION CRITERIA We planned to include randomised controlled trials that studied infants born at 37 or more weeks of gestation who had one or more episodes of gastrointestinal surgery within 28 days of birth, and compared administration of lactoferrin with a placebo. DATA COLLECTION AND ANALYSIS We used standard Cochrane methodological procedures. We planned to use the GRADE approach to assess the certainty of evidence for each outcome. MAIN RESULTS We identified no published randomised controlled studies that assessed the efficacy of lactoferrin for the postoperative management of term neonates following gastrointestinal surgery. AUTHORS' CONCLUSIONS There is currently no evidence available from randomised controlled trials to show whether lactoferrin is effective or ineffective for the postoperative management of term neonates after gastrointestinal surgery. There is a need for randomised controlled trials to be performed to assess the role of lactoferrin in this setting.
Collapse
Affiliation(s)
- Amit Trivedi
- Grace Centre for Newborn Care, The Children's Hospital at Westmead, Westmead, Australia
| | | | | | | |
Collapse
|
15
|
Li G, Lai Z, Shan A. Advances of Antimicrobial Peptide-Based Biomaterials for the Treatment of Bacterial Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206602. [PMID: 36722732 PMCID: PMC10104676 DOI: 10.1002/advs.202206602] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/12/2023] [Indexed: 05/10/2023]
Abstract
Owing to the increase in multidrug-resistant bacterial isolates in hospitals globally and the lack of truly effective antimicrobial agents, antibiotic resistant bacterial infections have increased substantially. There is thus an urgent need to develop new antimicrobial drugs and their related formulations. In recent years, natural antimicrobial peptides (AMPs), AMP optimization, self-assembled AMPs, AMP hydrogels, and biomaterial-assisted delivery of AMPs have shown great potential in the treatment of bacterial infections. In this review, it is focused on the development prospects and shortcomings of various AMP-based biomaterials for treating animal model infections, such as abdominal, skin, and eye infections. It is hoped that this review will inspire further innovations in the design of AMP-based biomaterials for the treatment of bacterial infections and accelerate their commercialization.
Collapse
Affiliation(s)
- Guoyu Li
- The Institute of Animal NutritionNortheast Agricultural UniversityHarbin150030P. R. China
| | - Zhenheng Lai
- The Institute of Animal NutritionNortheast Agricultural UniversityHarbin150030P. R. China
| | - Anshan Shan
- The Institute of Animal NutritionNortheast Agricultural UniversityHarbin150030P. R. China
| |
Collapse
|
16
|
Alegrete N, Sousa SR, Peleteiro B, Monteiro FJ, Gutierres M. Local Antibiotic Delivery Ceramic Bone Substitutes for the Treatment of Infected Bone Cavities and Bone Regeneration: A Systematic Review on What We Have Learned from Animal Models. MATERIALS (BASEL, SWITZERLAND) 2023; 16:2387. [PMID: 36984267 PMCID: PMC10056339 DOI: 10.3390/ma16062387] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 06/18/2023]
Abstract
AIMS the focus of this study is to evaluate if the combination of an antibiotic with a ceramic biomaterial is effective in treating osteomyelitis in an infected animal model and to define which model and protocol are best suited for in vivo experiments of local bone infection treatment. METHODS a systematic review was carried out based on PRISMA statement guidelines. A PubMed search was conducted to find original papers on animal models of bone infections using local antibiotic delivery systems with the characteristics of bone substitutes. Articles without a control group, differing from the experimental group only by the addition of antibiotics to the bone substitute, were excluded. RESULTS a total of 1185 records were retrieved, and after a three-step selection, 34 papers were included. Six manuscripts studied the effect of antibiotic-loaded biomaterials on bone infection prevention. Five articles studied infection in the presence of foreign bodies. In all but one, the combination of an antibiotic with bioceramic bone substitutes tended to prevent or cure bone infection while promoting biomaterial osteointegration. CONCLUSIONS this systematic review shows that the combination of antibiotics with bioceramic bone substitutes may be appropriate to treat bone infection when applied locally. The variability of the animal models, time to develop an infection, antibiotic used, way of carrying and releasing antibiotics, type of ceramic material, and endpoints limits the conclusions on the ideal therapy, enhancing the need for consistent models and guidelines to develop an adequate combination of material and antimicrobial agent leading to an effective human application.
Collapse
Affiliation(s)
- Nuno Alegrete
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- FMUP-Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Susana R. Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- ISEP-Instituto Superior de Engenharia do Porto, IPP - Instituto Politécnico do Porto, R. Dr. António Bernardino de Almeida 431, 4200-072 Porto, Portugal
| | - Bárbara Peleteiro
- EPIUnit-Instituto de Saúde Pública, Universidade do Porto, Rua das Taipas 135, 4050-600 Porto, Portugal
- Departamento de Ciências da Saúde Pública e Forenses e Educação Médica, Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- ITR-Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional, Rua das Taipas 135, 4050-600 Porto, Portugal
| | - Fernando J. Monteiro
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- FEUP-Faculdade de Engenharia, Universidade do Porto, R. Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Manuel Gutierres
- FMUP-Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- CHUSJ-Centro Hospitalar Universitário S. João, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
17
|
Yu C, Zhang F, Zhang L, Li J, Tang S, Li X, Peng M, Zhao Q, Zhu X. A bioinformatics approach to identifying the biomarkers and pathogenesis of major depressive disorder combined with acute myocardial infarction. Am J Transl Res 2023; 15:932-948. [PMID: 36915729 PMCID: PMC10006793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/17/2023] [Indexed: 03/16/2023]
Abstract
This study investigated the pathogenesis of major depressive disorder (MDD) and acute myocardial infarction (AMI) using bioinformatics. We analyzed MDD and AMI (MDD-AMI) datasets provided by the Gene Expression Omnibus (GEO) database for genes common to MDD and AMI using GEO2R and weighted gene co-expression network analysis (WGCNA). We also performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, and we used Disease Ontology (DO) analysis to identify a) the pathways through which genes function and b) comorbidities. We also created a protein-protein interaction (PPI) network using the STRING database to identify the hub genes and biomarkers. NetworkAnalyst 3.0 was used to construct a transcription factor (TF) gene regulatory network. We also identified relevant complications and potential drug candidates. The 27 genes common to MDD and AMI were enriched in the pathways regulating TFs and mediating immunity and inflammation. The hub genes in the PPI network included TLR2, HP, ICAM1, LCN2, LTF, VCAN, S100A9 and NFKBIA. Key TFs were KLF9, KLF11, ZNF24, and ZNF580. Cardiovascular, pancreatic, and skeletal diseases were common complications. Hydrocortisone, simvastatin, and estradiol were candidate treatment drugs. Identification of these genes and their pathways may provide new targets for further research on the pathogenesis, biomarkers, and treatment of MDD-AMI. Together our results suggested that TLR2 and VCAN might be the key genes associated with MDD complicated by AMI.
Collapse
Affiliation(s)
- Cheng Yu
- Department of Traditional Chinese Medicine Classics, Shandong University of Traditional Chinese Medicine Affiliated HospitalJinan, Shandong, China
| | - Fengjun Zhang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese MedicineJinan, Shandong, China
| | - Lili Zhang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese MedicineJinan, Shandong, China
| | - Jiajing Li
- Department of Traditional Chinese Medicine Classics, Shandong University of Traditional Chinese Medicine Affiliated HospitalJinan, Shandong, China
| | - Saixue Tang
- First Clinical School of Medicine, Shandong University of Traditional Chinese MedicineJinan, Shandong, China
| | - Xuejun Li
- Department of Traditional Chinese Medicine Classics, Shandong University of Traditional Chinese Medicine Affiliated HospitalJinan, Shandong, China
| | - Min Peng
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan, Shandong, China
| | - Qiong Zhao
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan, Shandong, China
| | - Xiuli Zhu
- Department of Radiation Oncology and Shandong Province Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinan, Shandong, China
| |
Collapse
|
18
|
Morici P, Rizzato C, Ghelardi E, Rossolini GM, Lupetti A. Sensitization of KPC and NDM Klebsiella pneumoniae To Rifampicin by the Human Lactoferrin-Derived Peptide hLF1-11. Microbiol Spectr 2023; 11:e0276722. [PMID: 36537823 PMCID: PMC9927577 DOI: 10.1128/spectrum.02767-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
A synergistic effect of non-bactericidal concentrations of the human lactoferrin (hLF)-derived peptide hLF1-11 and rifampicin against multidrug-resistant KPC (Klebsiella pneumoniae carbapenemase)-producing K. pneumoniae has been previously shown. The present study focuses on the mechanism(s) underlying this synergistic effect. The contribution of hLF1-11 and rifampicin to the synergistic effect was evaluated by killing assays with KPC K. pneumoniae cells incubated with hLF1-11 and, after washing, with rifampicin, or vice versa. Cell membrane permeability and polarization upon exposure to hLF1-11 and/or rifampicin were evaluated by ethidium bromide (EtBr) and DiBAC4(3) (bis-1,3-dibutylbarbituric acid trimethine oxonol) permeability, respectively. The effect of carbonyl cyanide m-chlorophenyl hydrazone (CCCP), an uncoupler of oxidative phosphorylation, was also evaluated. KPC K. pneumoniae cells were effectively killed after prior exposure to rifampicin for 30 to 60 min followed by treatment with hLF1-11, while no antibacterial activity was observed when cells were incubated with hLF1-11 first and then with rifampicin. EtBr accumulation increased upon exposure to hLF1-11 or the combination of hLF1-11 and rifampicin, but not upon exposure to rifampicin alone. Moreover, hLF1-11 induced a dose-dependent membrane depolarization. As expected, the antibacterial activity of hLF1-11 alone or combined with rifampicin was significantly reduced in the presence of CCCP. Furthermore, hLF1-11 and rifampicin were synergistic also against a colistin-resistant NDM (New Delhi metallo-β-lactamase)-producing K. pneumoniae strain. The results suggest that rifampicin was accumulated by KPC cells during the 30-to-60-min incubation and that the addition of hLF1-11 sensitized bacterial cells to rifampicin by inducing a transient loss of membrane potential and increased cell membrane permeability, thus facilitating the entrance and retention of rifampicin into the cytoplasm. IMPORTANCE The present study describes a synergistic effect between rifampicin, an impermeable hydrophobic antibiotic with an intracellular target, and an hLF1-11, an antimicrobial peptide derived from human lactoferrin, against multidrug-resistant Klebsiella pneumoniae. Carbapenem-resistant K. pneumoniae has recently caused an outbreak in Tuscany, Italy, thus pressing the need for the development of new treatment options. The mechanisms underlying such a synergistic effect have been studied. The results suggest that the synergistic effect was due to the transient loss of membrane potential induced by hLF1-11 and the subsequent increase in cell membrane permeability which allowed rifampicin to enter the bacterial cell. Therefore, it is likely that a sub-inhibitory concentration of hLF1-11 can efficiently permeabilize K. pneumoniae cells to rifampicin, allowing the antibiotic to reach its intracellular target. These results encourage further exploration of possible applications of this synergistic combination in the treatment of K. pneumoniae infections.
Collapse
Affiliation(s)
- Paola Morici
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Cosmeri Rizzato
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Emilia Ghelardi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Clinical Microbiology and Virology Unit, Florence Careggi University Hospital, Florence, Italy
| | - Antonella Lupetti
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| |
Collapse
|
19
|
Singh A, Duche RT, Wandhare AG, Sian JK, Singh BP, Sihag MK, Singh KS, Sangwan V, Talan S, Panwar H. Milk-Derived Antimicrobial Peptides: Overview, Applications, and Future Perspectives. Probiotics Antimicrob Proteins 2023; 15:44-62. [PMID: 36357656 PMCID: PMC9649404 DOI: 10.1007/s12602-022-10004-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2022] [Indexed: 11/13/2022]
Abstract
The growing consumer awareness towards healthy and safe food has reformed food processing strategies. Nowadays, food processors are aiming at natural, effective, safe, and low-cost substitutes for enhancing the shelf life of food products. Milk, besides being a rich source of nutrition for infants and adults, serves as a readily available source of precious functional peptides. Due to the existence of high genetic variability in milk proteins, there is a great possibility to get bioactive peptides with varied properties. Among other bioactive agents, milk-originated antimicrobial peptides (AMPs) are gaining interest as attractive and safe additive conferring extended shelf life to minimally processed foods. These peptides display broad-spectrum antagonistic activity against bacteria, fungi, viruses, and protozoans. Microbial proteolytic activity, extracellular peptidases, food-grade enzymes, and recombinant DNA technology application are among few strategies to tailor specific peptides from milk and enhance their production. These bioprotective agents have a promising future in addressing the global concern of food safety along with the possibility to be incorporated into the food matrix without compromising overall consumer acceptance. Additionally, in conformity to the current consumer demands, these AMPs also possess functional properties needed for value addition. This review attempts to present the basic properties, synthesis approaches, action mechanism, current status, and prospects of antimicrobial peptide application in food, dairy, and pharma industry along with their role in ensuring the safety and health of consumers.
Collapse
Affiliation(s)
- Anamika Singh
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, 141001 Punjab India
| | - Rachael Terumbur Duche
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, 141001 Punjab India ,Department of Microbiology, Federal University of Agriculture, Makurdi, Nigeria
| | - Arundhati Ganesh Wandhare
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, 141001 Punjab India
| | - Jaspreet Kaur Sian
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, 141001 Punjab India ,Department of Microbiology, Punjab Agricultural University (PAU), Ludhiana, 141001 Punjab India
| | - Brij Pal Singh
- Department of Microbiology, Central University of Haryana, Mahendergarh, 123031 Haryana India
| | - Manvesh Kumar Sihag
- Department of Dairy Chemistry, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, 141001 Punjab India
| | - Kumar Siddharth Singh
- Institute for Microbiology, Gottfried Wilhelm Leibniz University, Herrenhäuser Str. 2, 30419 Hanover, Germany
| | - Vikas Sangwan
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, 141001 Punjab India
| | - Shreya Talan
- Dairy Microbiology Division, ICAR-National Dairy Research Institute (ICAR-NDRI), Karnal, Haryana India
| | - Harsh Panwar
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, 141001, Punjab, India.
| |
Collapse
|
20
|
Gonçalves S, Martins IC, Santos NC. Nanoparticle‐peptide conjugates for bacterial detection and neutralization: Potential applications in diagnostics and therapy. WIRES NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1819. [DOI: 10.1002/wnan.1819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 11/24/2022]
Affiliation(s)
- Sónia Gonçalves
- Instituto de Medicina Molecular, Faculdade de Medicina Universidade de Lisboa Lisbon Portugal
| | - Ivo C. Martins
- Instituto de Medicina Molecular, Faculdade de Medicina Universidade de Lisboa Lisbon Portugal
| | - Nuno C. Santos
- Instituto de Medicina Molecular, Faculdade de Medicina Universidade de Lisboa Lisbon Portugal
| |
Collapse
|
21
|
Sawale M, Ozadali F, Valentine CJ, Benyathiar P, Drolia R, Mishra DK. Impact of bovine lactoferrin fortification on pathogenic organisms to attenuate the risk of infection for infants. Food Control 2022. [DOI: 10.1016/j.foodcont.2022.109078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
22
|
Salivary Proteome, Inflammatory, and NETosis Biomarkers in Older Adult Practitioners and Nonpractitioners of Physical Exercise. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3725056. [PMID: 35502212 PMCID: PMC9056209 DOI: 10.1155/2022/3725056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/22/2022] [Indexed: 11/18/2022]
Abstract
Since aging has been associated with increased production of inflammatory biomarkers, the ability to monitor older adults repeatedly is highly desirable, and saliva is an interesting biofluid for the search of biomarkers, as it is easily accessible in a noninvasive manner. However, given the incipient knowledge of salivary biomarkers in aging and its relationship to physical exercise, the present study is aimed at evaluating the protein expression and the levels of inflammatory and NETosis biomarkers in the saliva of practitioners (PE) and nonpractitioners (NPE) of physical exercise older adults. Six (6) practitioner and 4 nonpractitioner older adults were enrolled in this study. Unstimulated whole saliva was collected for analysis of the proteome by label-free mass spectrometry, as well as of the inflammatory status by evaluation of C-reactive protein (CRP), vascular endothelial growth factor (VEGF), and cytokines (TNF-α, interleukin- (IL-) 1β, and IL-8), while NETosis was assessed by myeloperoxidase (MPO) and neutrophil elastase. Regarding oral health, the decayed, missing, and filled teeth (DMF-T) index, bleeding on probing, suppuration, and probing depth measurement (mm) were evaluated. In addition, functional capacity was investigated using the General Physical Fitness Index (GPFI). In relation to the proteome analysis, 93 and 143 proteins were found exclusively in the PE and NPE groups, respectively; 224 proteins were common to both groups. Among these proteins, 10 proteins showed statistical difference (
) between the groups: alpha-2-macroglobulin, component 3 of the complement, serotransferrin, and protein soluble in brain acid 1 were less expressed, while lactotransferrin, alpha-amylase 1, S100-A8, S100-A9, lactoperoxidase, and galectin-3 binding protein were more expressed in the PE group. No differences between groups were observed in the analysis of inflammatory and NETosis biomarkers. This study shows the potential utility of saliva for detecting protein biomarkers in a noninvasive biological sample of the elderly population.
Collapse
|
23
|
Nakamura M, Tsuda N, Miyata T, Ikenaga M. Antimicrobial effect and mechanism of bovine lactoferrin against the potato common scab pathogen Streptomyces scabiei. PLoS One 2022; 17:e0264094. [PMID: 35213576 PMCID: PMC8880714 DOI: 10.1371/journal.pone.0264094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/02/2022] [Indexed: 12/17/2022] Open
Abstract
Lactoferrin (LF) is a multifunctional protein with a broad spectrum of antimicrobial activities. In this study, we investigated the antimicrobial activity of LF against the potato common scab pathogen Streptomyces scabiei, which causes severe damage to potato tubers. LF derived from bovine (bLF) had much higher activity against S. scabiei than human LF. The minimal inhibitory concentration of bLF was 3.9 μM. The effects of both apo-bLF (iron-free) and holo-bLF (iron-saturated) on S. scabiei were not different. Bovine lactoferricin (LFcinB), a short peptide with a length of 25 amino acid residues located in the N-terminal region of bLF, showed antimicrobial activity against S. scabiei, similar to that of bLF. These results indicated that the antimicrobial activity of bLF against S. scabiei cannot be attributed to its iron-chelating effect but to the bioactivity of its peptides. When S. scabiei was treated with the fusion protein of mCherry-LFcinB (red fluorescent protein) expressed in Escherichia coli, the pseudohyphal cells instantly glowed, indicating that the peptide electrostatically binds to the surface of S. scabiei. An assay of synthetic peptides, with modified number of arginine (Arg) and tryptophan (Trp) residues based on the antimicrobial center (RRWQWR) of LFcinB showed that Trp residues are implicated in the antimicrobial activity against S. scabiei; however, Arg residues are also necessary to carry Trp residues to the cell surface to fully exert its activity. Although the single amino acid effect of Trp had low activity, Trp derivatives showed much higher activity against S. scabiei, suggesting that the derivatives effectively bind to the cell surface (cell membrane) by themselves without a carrier. Thus, amino acid derivatives might be considered effective and alternative antimicrobial substances.
Collapse
Affiliation(s)
- Masayuki Nakamura
- Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
- * E-mail:
| | - Naoaki Tsuda
- Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
| | - Takeshi Miyata
- Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
| | - Makoto Ikenaga
- Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
24
|
Escobar‐Salom M, Torrens G, Jordana‐Lluch E, Oliver A, Juan C. Mammals' humoral immune proteins and peptides targeting the bacterial envelope: from natural protection to therapeutic applications against multidrug‐resistant
Gram
‐negatives. Biol Rev Camb Philos Soc 2022; 97:1005-1037. [PMID: 35043558 PMCID: PMC9304279 DOI: 10.1111/brv.12830] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022]
Abstract
Mammalian innate immunity employs several humoral ‘weapons’ that target the bacterial envelope. The threats posed by the multidrug‐resistant ‘ESKAPE’ Gram‐negative pathogens (Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.) are forcing researchers to explore new therapeutic options, including the use of these immune elements. Here we review bacterial envelope‐targeting (peptidoglycan and/or membrane‐targeting) proteins/peptides of the mammalian immune system that are most likely to have therapeutic applications. Firstly we discuss their general features and protective activity against ESKAPE Gram‐negatives in the host. We then gather, integrate, and discuss recent research on experimental therapeutics harnessing their bactericidal power, based on their exogenous administration and also on the discovery of bacterial and/or host targets that improve the performance of this endogenous immunity, as a novel therapeutic concept. We identify weak points and knowledge gaps in current research in this field and suggest areas for future work to obtain successful envelope‐targeting therapeutic options to tackle the challenge of antimicrobial resistance.
Collapse
Affiliation(s)
- María Escobar‐Salom
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Gabriel Torrens
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Elena Jordana‐Lluch
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Antonio Oliver
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Carlos Juan
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| |
Collapse
|
25
|
Silva ARP, Guimarães M, Rabelo J, Belen L, Perecin C, Farias J, Picado Madalena Santos JH, Rangel-Yagui CO. Recent advances in the design of antimicrobial peptide conjugates. J Mater Chem B 2022; 10:3587-3600. [DOI: 10.1039/d1tb02757c] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Antimicrobial peptides (AMPs) are ubiquitous host defense peptides characterized by antibiotic activity and lower propensity for developing resistance compared to classic antibiotics. While several AMPs have shown activity against antibiotic-sensitive...
Collapse
|
26
|
Kadam S, Madhusoodhanan V, Dhekane R, Bhide D, Ugale R, Tikhole U, Kaushik KS. Milieu matters: An in vitro wound milieu to recapitulate key features of, and probe new insights into, mixed-species bacterial biofilms. Biofilm 2021; 3:100047. [PMID: 33912828 PMCID: PMC8065265 DOI: 10.1016/j.bioflm.2021.100047] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/29/2021] [Accepted: 03/29/2021] [Indexed: 12/20/2022] Open
Abstract
Bacterial biofilms are a major cause of delayed wound healing. Consequently, the study of wound biofilms, particularly in host-relevant conditions, has gained importance. Most in vitro studies employ refined laboratory media to study biofilms, representing conditions that are not relevant to the infection state. To mimic the wound milieu, in vitro biofilm studies often incorporate serum or plasma in growth conditions, or employ clot or matrix-based biofilm models. While incorporating serum or plasma alone is a minimalistic approach, the more complex in vitro wound models are technically demanding, and poorly compatible with standard biofilm assays. Based on previous reports of clinical wound fluid composition, we have developed an in vitro wound milieu (IVWM) that includes, in addition to serum (to recapitulate wound fluid), matrix elements and biochemical factors. With Luria-Bertani broth and Fetal Bovine Serum (FBS) for comparison, the IVWM was used to study planktonic growth, biofilm features, and interspecies interactions, of common wound pathogens, Staphylococcus aureus and Pseudomonas aeruginosa. We demonstrate that the IVWM recapitulates widely reported in vivo biofilm features such as biomass formation, metabolic activity, increased antibiotic tolerance, 3D structure, and interspecies interactions for monospecies and mixed-species biofilms. Further, the IVWM is simple to formulate, uses laboratory-grade components, and is compatible with standard biofilm assays. Given this, it holds potential as a tractable approach to study wound biofilms under host-relevant conditions.
Collapse
Affiliation(s)
- Snehal Kadam
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Vandana Madhusoodhanan
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Radhika Dhekane
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Devyani Bhide
- MES Abasaheb Garware College of Arts and Science, Pune, India
| | - Rutuja Ugale
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Utkarsha Tikhole
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Karishma S. Kaushik
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| |
Collapse
|
27
|
Nayak A, Tiozzo C, Lin X, Mejia C, Gurzenda E, Kim M, Hanna N. Is Lactoferrin Supplementation Beneficial for All Preterm Infants? Am J Perinatol 2021; 40:680-687. [PMID: 34058763 DOI: 10.1055/s-0041-1730433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Human milk (HM) has antibacterial properties due to the presence of immune-modulators, including lactoferrin (LF). This study will determine effect(s) of HM maturation, fortification, and storage conditions on LF levels and its antibacterial properties. STUDY DESIGN HM samples (n = 30) were obtained from preterm and term mothers. The LF levels were analyzed by ELISA, and the antibacterial activity was measured after inoculation with Escherichia coli. RESULTS The highest level of LF in preterm HM was observed in the first week of lactation. However, storage of preterm HM at 4°C decreased LF levels significantly. Both LF levels and antibacterial activity in preterm HM was lower compared with term HM, but significantly higher than donor HM even after HM-based fortification. LF supplementation of donor HM improved its antibacterial activity. CONCLUSION Preterm infants fed donor HM, formula, or stored HM at 4°C may benefits from LF supplementation to improve HM antibacterial properties. KEY POINTS · Milk LF levels vary with storage and maturity.. · Donor milk is deficient in LF even after adding HM-based fortification.. · Donor HM and formula fed infants may benefit from LF..
Collapse
Affiliation(s)
- Amrita Nayak
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York
| | - Caterina Tiozzo
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York.,Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Xinhua Lin
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Claudia Mejia
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Ellen Gurzenda
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Maureen Kim
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York
| | - Nazeeh Hanna
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York.,Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| |
Collapse
|
28
|
Muñoz-Prieto A, Escribano D, Contreras-Aguilar MD, Horvatić A, Guillemin N, Jacobsen S, Cerón JJ, Mrljak V. Tandem Mass Tag (TMT) Proteomic Analysis of Saliva in Horses with Acute Abdominal Disease. Animals (Basel) 2021; 11:ani11051304. [PMID: 33946607 PMCID: PMC8147179 DOI: 10.3390/ani11051304] [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: 02/05/2021] [Revised: 04/13/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary This study shows for the first time the variation of the salivary proteome in horses with acute abdominal disease (AAD) compared with healthy horses through a high-throughput proteomic approach. A total of 118 proteins were identified, and 17 showed significant changes between the two groups. The changes observed in proteins were closely related to an impaired primary immune defense and antimicrobial capacity in the mucosa, and one salivary protein (lactoferrin) was successfully verified. These results may increase the background and knowledge of saliva composition in horses with AAD and further understanding of the physiopathological changes occurring in the organism in this disease. Abstract The aim of this study was to investigate the changes in the salivary proteome in horses with acute abdominal disease (AAD) using a tandem mass tags (TMT)-based proteomic approach. The saliva samples from eight horses with AAD were compared with six healthy horses in the proteomic study. Additionally, saliva samples from eight horses with AAD and eight controls were used to validate lactoferrin (LF) in saliva. The TMT analysis quantified 118 proteins. Of these, 17 differed significantly between horses with AAD and the healthy controls, 11 being downregulated and 6 upregulated. Our results showed the downregulation of gamma-enteric smooth muscle actin (ACTA2), latherin isoform X1, and LF. These proteins could be closely related to an impaired primary immune defense and antimicrobial capacity in the mucosa. In addition, there was an upregulation of mucin 19 (MUC19) and the serine protease inhibitor Kazal-type 5 (SPINK5) associated with a protective effect during inflammation. The proteins identified in our study could have the potential to be novel biomarkers for diagnosis or monitoring the physiopathology of the disease, especially LF, which decreased in the saliva of horses with AAD and was successfully measured using a commercially available immunoassay.
Collapse
Affiliation(s)
- Alberto Muñoz-Prieto
- Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia; (A.M.-P.); (N.G.); (V.M.)
| | - Damián Escribano
- Interdisciplinary Laboratory of Clinical Analysis, Interlab-UMU, Regional Campus of International Excellence ‘Campus Mare Nostrum’, University of Murcia, 30100 Murcia, Spain; (D.E.); (M.D.C.-A.)
| | - María Dolores Contreras-Aguilar
- Interdisciplinary Laboratory of Clinical Analysis, Interlab-UMU, Regional Campus of International Excellence ‘Campus Mare Nostrum’, University of Murcia, 30100 Murcia, Spain; (D.E.); (M.D.C.-A.)
| | - Anita Horvatić
- Department of Chemistry and Biochemistry, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10 000 Zagreb, Croatia;
| | - Nicolas Guillemin
- Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia; (A.M.-P.); (N.G.); (V.M.)
| | - Stine Jacobsen
- Department of Veterinary Clinical Sciences, Veterinary School of Medicine, Sektion Medicine and Surgery, University of Copenhagen, Hoejbakkegaard Allé 5, DK-2630 Taastrup, Denmark;
| | - José Joaquín Cerón
- Interdisciplinary Laboratory of Clinical Analysis, Interlab-UMU, Regional Campus of International Excellence ‘Campus Mare Nostrum’, University of Murcia, 30100 Murcia, Spain; (D.E.); (M.D.C.-A.)
- Correspondence:
| | - Vladimir Mrljak
- Clinic for Internal Diseases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia; (A.M.-P.); (N.G.); (V.M.)
| |
Collapse
|
29
|
El-Baky NA, Elkhawaga MA, Abdelkhalek ES, Sharaf MM, Redwan EM, Kholef HR. De novo expression and antibacterial potential of four lactoferricin peptides in cell-free protein synthesis system. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2021; 29:e00583. [PMID: 33425692 PMCID: PMC7779732 DOI: 10.1016/j.btre.2020.e00583] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 11/20/2022]
Abstract
For the first time, we produced four lactoferricin (LFcin) peptides by a cell-free (in vitro) method. These short antimicrobial peptides were expressed in an E. coli cell-free protein synthesis (CFPS) system and the bioactivity of the produced peptides was demonstrated. Additionally, we designed a novel synthetic consensus peptide (ConLFcin). The genes of bovine Lfcin (bLFcin), human Lfcin (hLFcin), camel Lfcin (cLFcin), and ConLFcin were cloned into pET101/D-TOPO vector then peptides were synthesized in vitro by E. coli CFPS system. The antibacterial activity of these synthesized peptides was evaluated against Escherichia coli, Salmonella typhi, Pseudomonas aeruginosa, Staphylococcus aureus, and methicillin-resistant Staphylococcus aureus (MRSA). The four cell-free synthesized peptides showed significant antibacterial potency at minimum inhibitory concentration (MIC) values between 1.25 and 10 μg/mL. cLFcin and ConLFcin showed higher antibacterial effects than bLFcin and hLFcin. Thus, cell-free expression system is an ideal system for rapid expression of functionally active short bioactive peptides.
Collapse
Key Words
- 3D, three dimensional structures
- Antimicrobial
- Bioactive peptides
- CAMH, cation-adjusted Mueller-Hinton broth
- CFPS, cell-free protein synthesis
- ConLFcin, consensus lactoferricin
- ELISA, enzyme-linked immunosorbent assay
- HSV, herpes simplex virus
- In vitro protein synthesis
- LC50, concentration lethal to 50 % of the cells
- LFcin, lactoferricin
- Lactoferricin
- Lactoferrin
- Lf, lactoferrin
- MIC, minimum inhibitory concentration
- MICs, minimum inhibitory concentrations
- MRSA, methicillin-resistant Staphylococcus aureus
- PBMCs, peripheral blood mononuclear cells
- SD, Shine-Dalgarno sequence
- SDS-PAGE, sodium dodecyl sulfate polyacrylamide gel electrophoresis
- bLFcin, bovine lactoferricin
- cLFcin, camel lactoferricin
- cLf, camel lactoferrin
- hLFcin, human lactoferricin
- hLf, human lactoferrin
- p-NPP, p-Nitrophenyl phosphate
Collapse
Affiliation(s)
- Nawal Abd El-Baky
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, New Borg EL-Arab, 21934, Alexandria, Egypt
| | - Maie Ahmed Elkhawaga
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | | | - Mona Mohammed Sharaf
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, New Borg EL-Arab, 21934, Alexandria, Egypt
| | - Elrashdy Mustafa Redwan
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, New Borg EL-Arab, 21934, Alexandria, Egypt
| | - Hoda Reda Kholef
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, New Borg EL-Arab, 21934, Alexandria, Egypt
| |
Collapse
|
30
|
Beverly RL, Woonnimani P, Scottoline BP, Lueangsakulthai J, Dallas DC. Peptides from the Intestinal Tract of Breast Milk-Fed Infants Have Antimicrobial and Bifidogenic Activity. Int J Mol Sci 2021; 22:ijms22052377. [PMID: 33673498 PMCID: PMC7956819 DOI: 10.3390/ijms22052377] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/17/2022] Open
Abstract
For bioactive milk peptides to be relevant to infant health, they must be released by gastrointestinal proteolysis and resist further proteolysis until they reach their site of activity. The intestinal tract is the likeliest site for most bioactivities, but it is currently unknown whether bioactive milk peptides are present therein. The purpose of the present study was to identify antimicrobial and bifidogenic peptides in the infant intestinal tract. Milk peptides were extracted from infant intestinal samples, and the activities of the bulk peptide extracts were determined by measuring growth of Escherichia coli, Staphylococcus aureus, and Bifidobacterium longum spp. infantis after incubation with serial dilutions. The peptide profiles of active and inactive samples were determined by peptidomics analysis and compared to identify candidate peptides for bioactivity testing. We extracted peptides from 29 intestinal samples collected from 16 infants. Five samples had antimicrobial activity against S. aureus and six samples had bifidogenic activity for B. infantis. We narrowed down a list of 6645 milk peptides to 11 candidate peptides for synthesis, of which 6 fully inhibited E. coli and S. aureus growth at concentrations of 2500 and 3000 µg/mL. This study provides evidence for the potential bioactivity of milk peptides in the infant intestinal tract.
Collapse
Affiliation(s)
- Robert L. Beverly
- College of Public Health and Human Sciences, Oregon State University, Corvallis, OR 97331, USA; (J.L.); (D.C.D.)
- Correspondence: ; Tel.: +1-217-2278-2672
| | - Prajna Woonnimani
- College of Agricultural Sciences, Oregon State University, Corvallis, OR 97331, USA;
| | - Brian P. Scottoline
- Division of Neonatology, School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA;
| | - Jiraporn Lueangsakulthai
- College of Public Health and Human Sciences, Oregon State University, Corvallis, OR 97331, USA; (J.L.); (D.C.D.)
| | - David C. Dallas
- College of Public Health and Human Sciences, Oregon State University, Corvallis, OR 97331, USA; (J.L.); (D.C.D.)
| |
Collapse
|
31
|
Dijksteel GS, Ulrich MMW, Middelkoop E, Boekema BKHL. Review: Lessons Learned From Clinical Trials Using Antimicrobial Peptides (AMPs). Front Microbiol 2021; 12:616979. [PMID: 33692766 PMCID: PMC7937881 DOI: 10.3389/fmicb.2021.616979] [Citation(s) in RCA: 227] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/29/2021] [Indexed: 12/15/2022] Open
Abstract
Antimicrobial peptides (AMPs) or host defense peptides protect the host against various pathogens such as yeast, fungi, viruses and bacteria. AMPs also display immunomodulatory properties ranging from the modulation of inflammatory responses to the promotion of wound healing. More interestingly, AMPs cause cell disruption through non-specific interactions with the membrane surface of pathogens. This is most likely responsible for the low or limited emergence of bacterial resistance against many AMPs. Despite the increasing number of antibiotic-resistant bacteria and the potency of novel AMPs to combat such pathogens, only a few AMPs are in clinical use. Therefore, the current review describes (i) the potential of AMPs as alternatives to antibiotics, (ii) the challenges toward clinical implementation of AMPs and (iii) strategies to improve the success rate of AMPs in clinical trials, emphasizing the lessons we could learn from these trials.
Collapse
Affiliation(s)
- Gabrielle S Dijksteel
- Association of Dutch Burn Centres, Beverwijk, Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Magda M W Ulrich
- Association of Dutch Burn Centres, Beverwijk, Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Esther Middelkoop
- Association of Dutch Burn Centres, Beverwijk, Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | |
Collapse
|
32
|
Ryu M, Park J, Yeom JH, Joo M, Lee K. Rediscovery of antimicrobial peptides as therapeutic agents. J Microbiol 2021; 59:113-123. [PMID: 33527313 DOI: 10.1007/s12275-021-0649-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023]
Abstract
In recent years, the occurrence of antibiotic-resistant pathogens is increasing rapidly. There is growing concern as the development of antibiotics is slower than the increase in the resistance of pathogenic bacteria. Antimicrobial peptides (AMPs) are promising alternatives to antibiotics. Despite their name, which implies their antimicrobial activity, AMPs have recently been rediscovered as compounds having antifungal, antiviral, anticancer, antioxidant, and insecticidal effects. Moreover, many AMPs are relatively safe from toxic side effects and the generation of resistant microorganisms due to their target specificity and complexity of the mechanisms underlying their action. In this review, we summarize the history, classification, and mechanisms of action of AMPs, and provide descriptions of AMPs undergoing clinical trials. We also discuss the obstacles associated with the development of AMPs as therapeutic agents and recent strategies formulated to circumvent these obstacles.
Collapse
Affiliation(s)
- Minkyung Ryu
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jaeyeong Park
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ji-Hyun Yeom
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Minju Joo
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Kangseok Lee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
33
|
de Oliveira Costa B, Franco OL. Cryptic Host Defense Peptides: Multifaceted Activity and Prospects for Medicinal Chemistry. Curr Top Med Chem 2021; 20:1274-1290. [PMID: 32209042 DOI: 10.2174/1568026620666200325112425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/03/2020] [Accepted: 02/24/2020] [Indexed: 01/22/2023]
Abstract
Host defense peptides (HDPs) comprise a heterogeneous group of evolutionarily conserved and biologically active small molecules that are produced by different organisms. HDPs are widely researched because they often have multiple biological activities, for example antimicrobial, immunomodulatory and anticancer activity. In this context, in this review we focus on cryptic HDPs, molecules derived specifically from proteolytic processing of endogenous precursor proteins. Here, we explore the biological activity of such molecules and we further discuss the development of optimized sequences based on these natural cryptic HDPs. In addition, we present clinical-phase studies of cryptic HDPs (natural or optimized), and point out the possible applicability of these molecules in medicinal chemistry.
Collapse
Affiliation(s)
- Bruna de Oliveira Costa
- S-inova Biotech, Graduate Program in Biotechnology, Universidade Catolica Dom Bosco, Campo Grande, MS, Brazil
| | - Octávio Luiz Franco
- S-inova Biotech, Graduate Program in Biotechnology, Universidade Catolica Dom Bosco, Campo Grande, MS, Brazil.,Department of Genomic Sciences and Biotechnology, Center for Analysis of Proteomics and Biochemistry, Catholic University of Brasília, Brasília, DF, Brazil.,Department of Molecular Pathology, Faculty of Medicine, University of Brasília, Brasília-DF, Brazil
| |
Collapse
|
34
|
Iron in immune cell function and host defense. Semin Cell Dev Biol 2020; 115:27-36. [PMID: 33386235 DOI: 10.1016/j.semcdb.2020.12.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022]
Abstract
The control over iron availability is crucial under homeostatic conditions and even more in the case of an infection. This results from diverse properties of iron: first, iron is an important trace element for the host as well as for the pathogen for various cellular and metabolic processes, second, free iron catalyzes Fenton reaction and is therefore producing reactive oxygen species as a part of the host defense machinery, third, iron exhibits important effects on immune cell function and differentiation and fourth almost every immune activation in turn impacts on iron metabolism and spatio-temporal iron distribution. The central importance of iron in the host and microbe interplay and thus for the course of infections led to diverse strategies to restrict iron for invading pathogens. In this review, we focus on how iron restriction to the pathogen is a powerful innate immune defense mechanism of the host called "nutritional immunity". Important proteins in the iron-host-pathogen interplay will be discussed as well as the influence of iron on the efficacy of innate and adaptive immunity. Recently described processes like ferritinophagy and ferroptosis are further covered in respect to their impact on inflammation and infection control and how they impact on our understanding of the interaction of host and pathogen.
Collapse
|
35
|
Gunzer M, Thornton CR, Beziere N. Advances in the In Vivo Molecular Imaging of Invasive Aspergillosis. J Fungi (Basel) 2020; 6:jof6040338. [PMID: 33291706 PMCID: PMC7761943 DOI: 10.3390/jof6040338] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Invasive pulmonary aspergillosis (IPA) is a life-threatening infection of immunocompromised patients with Aspergillus fumigatus, a ubiquitous environmental mould. While there are numerous functioning antifungal therapies, their high cost, substantial side effects and fear of overt resistance development preclude permanent prophylactic medication of risk-patients. Hence, a fast and definitive diagnosis of IPA is desirable, to quickly identify those patients that really require aggressive antimycotic treatment and to follow the course of the therapeutic intervention. However, despite decades of research into this issue, such a diagnostic procedure is still not available. Here, we discuss the array of currently available methods for IPA detection and their limits. We then show that molecular imaging using positron emission tomography (PET) combined with morphological computed tomography or magnetic imaging is highly promising to become a future non-invasive approach for IPA diagnosis and therapy monitoring, albeit still requiring thorough validation and relying on further acceptance and dissemination of the approach. Thereby, our approach using the A. fumigatus-specific humanized monoclonal antibody hJF5 labelled with 64Cu as PET-tracer has proven highly effective in pre-clinical models and hence bears high potential for human application.
Collapse
Affiliation(s)
- Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, 45147 Essen, Germany
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany
- Correspondence: (M.G.); (N.B.); Tel.: +49-201-183-6640 (M.G.); +49-7071-29-87511 (N.B.)
| | - Christopher R. Thornton
- ISCA Diagnostics Ltd. and Biosciences, College of Life & Environmental Sciences, University of Exeter, Exeter EX4 4PY, UK;
| | - Nicolas Beziere
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Correspondence: (M.G.); (N.B.); Tel.: +49-201-183-6640 (M.G.); +49-7071-29-87511 (N.B.)
| |
Collapse
|
36
|
Lactoferrin Metal Saturation-Which Form Is the Best for Neonatal Nutrition? Nutrients 2020; 12:nu12113340. [PMID: 33143055 PMCID: PMC7692973 DOI: 10.3390/nu12113340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 10/28/2020] [Indexed: 01/04/2023] Open
Abstract
We evaluated the impact of metal saturation of lactoferrin (with iron and manganese) on population numbers of pathogenic species relevant for neonatal sepsis that commonly originates from the gut due to bacterial translocation. Little attention has been paid to how metal ions bound to the protein affect its activity. Several reference and clinical strains as well as probiotic strains were incubated with different forms of lactoferrin: metal-depleted (apolactoferrin), iron-saturated (hololactoferrin) and manganese-saturated lactoferrin. We also attempted to confirm the observed effects of lactoferrin forms in vivo using rat pups. The observed decrease in population numbers of Gram-negative rods could not be confirmed by quantitative plating—lactoferrin may regulate these populations diversely (e.g., by anti-biofilm activity) and contribute to the inhibition of inflammatory response. We did not see any effect of lactoferrin forms on staphylococci and bifidobacteria. However, we have noted a significant increase of population numbers of Lactobacillus strains upon incubation with manganese-saturated lactoferrin. These results were confirmed in vivo in a rat model. Metal saturation is an underestimated factor regulating lactoferrin activity. Some forms are more potent in the inhibition of pathogenic species while others, such as manganese-saturated lactoferrin, could contribute to the restoration of gut homeostasis.
Collapse
|
37
|
Mercer DK, O'Neil DA. Innate Inspiration: Antifungal Peptides and Other Immunotherapeutics From the Host Immune Response. Front Immunol 2020; 11:2177. [PMID: 33072081 PMCID: PMC7533533 DOI: 10.3389/fimmu.2020.02177] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/10/2020] [Indexed: 12/17/2022] Open
Abstract
The purpose of this review is to describe antifungal therapeutic candidates in preclinical and clinical development derived from, or directly influenced by, the immune system, with a specific focus on antimicrobial peptides (AMP). Although the focus of this review is AMP with direct antimicrobial effects on fungi, we will also discuss compounds with direct antifungal activity, including monoclonal antibodies (mAb), as well as immunomodulatory molecules that can enhance the immune response to fungal infection, including immunomodulatory AMP, vaccines, checkpoint inhibitors, interferon and colony stimulating factors as well as immune cell therapies. The focus of this manuscript will be a non-exhaustive review of antifungal compounds in preclinical and clinical development that are based on the principles of immunology and the authors acknowledge the incredible amount of in vitro and in vivo work that has been conducted to develop such therapeutic candidates.
Collapse
|
38
|
Ramamourthy G, Vogel HJ. Antibiofilm activity of lactoferrin-derived synthetic peptides against Pseudomonas aeruginosa PAO1. Biochem Cell Biol 2020; 99:138-148. [PMID: 32871093 DOI: 10.1139/bcb-2020-0253] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Many pathogenic bacteria can protect themselves from the effects of antibiotics and the host immune response system by forming biofilms. Biofilms are polymer-entrapped bacterial cells, which adhere to each other and are often attached to a surface. Eradication of bacterial biofilms typically requires much higher concentrations of antibiotics than are normally needed to kill cultured planktonic cells, raising serious clinical concerns. In an attempt to prevent the formation of biofilms or to break up existing biofilms of pathogenic bacteria, herein we have used the standard crystal violet assay as well as the Calgary biofilm device to test several lactoferrin- and lactoferricin-derived antimicrobial peptides for their antibiofilm activity against Pseudomonas aeruginosa PAO1. Our results revealed that the short bovine lactoferricin-derived RRWQWR-NH2 (20-25) hexapeptide has no activity against P. aeruginosa PAO1. Moreover, the longer human lactoferricin-derived peptide GRRRRSVQWCA (1-11) and the bovine lactoferrampin (268-284) peptide were also almost devoid of activity. However, several different "mix-and-match" dimeric versions of the two lactoferricin-derived peptides proved quite effective in preventing the formation of biofilms at low concentrations, and in some cases, could even eradicate an existing biofilm. Moreover, the full-length bovine lactoferricinB (17-41) peptide also displayed considerable antimicrobial activity. Some of the longer lactoferricin-derived dimeric peptides acted through a bactericidal mechanism, whereas others seemed to interfere in cell-signalling processes. Taken together, our results indicate that synthetic dimeric peptides comprising short naturally occurring human and bovine lactoferricin constructs could be further developed as antibiofilm agents.
Collapse
Affiliation(s)
- Gopal Ramamourthy
- Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada.,Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Hans J Vogel
- Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada.,Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
39
|
Tarnow-Mordi WO, Abdel-Latif ME, Martin A, Pammi M, Robledo K, Manzoni P, Osborn D, Lui K, Keech A, Hague W, Ghadge A, Travadi J, Brown R, Darlow BA, Liley H, Pritchard M, Kochar A, Isaacs D, Gordon A, Askie L, Cruz M, Schindler T, Dixon K, Deshpande G, Tracy M, Schofield D, Austin N, Sinn J, Simes RJ. The effect of lactoferrin supplementation on death or major morbidity in very low birthweight infants (LIFT): a multicentre, double-blind, randomised controlled trial. THE LANCET CHILD & ADOLESCENT HEALTH 2020; 4:444-454. [PMID: 32407710 DOI: 10.1016/s2352-4642(20)30093-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/06/2020] [Accepted: 03/23/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Very low birthweight or preterm infants are at increased risk of adverse outcomes including sepsis, necrotising enterocolitis, and death. We assessed whether supplementing the enteral diet of very low-birthweight infants with lactoferrin, an antimicrobial protein, reduces all-cause mortality or major morbidity. METHODS We did a multicentre, double-blind, pragmatic, randomised superiority trial in 14 Australian and two New Zealand neonatal intensive care units. Infants born weighing less than 1500 g and aged less than 8 days, were eligible and randomly assigned (1:1) using minimising web-based randomisation to receive once daily 200 mg/kg pasteurised bovine lactoferrin supplements or no lactoferrin supplement added to breast or formula milk until 34 weeks' post-menstrual age (or for 2 weeks, if longer), or until discharge from the study hospital if that occurred first. Designated nurses preparing the daily feeds were not masked to group assignment, but other nurses, doctors, parents, caregivers, and investigators were unaware. The primary outcome was survival to hospital discharge or major morbidity (defined as brain injury, necrotising enterocolitis, late-onset sepsis at 36 weeks' post-menstrual age, or retinopathy treated before discharge) assessed in the intention-to-treat population. Safety analyses were by treatment received. We also did a prespecified, PRISMA-compliant meta-analysis, which included this study and other relevant randomised controlled trials, to estimate more precisely the effects of lactoferrin supplementation on late-onset sepsis, necrotising enterocolitis, and survival. This trial is registered with the Australian and New Zealand Clinical Trials Registry, ACTRN12611000247976. FINDINGS Between June 27, 2014, and Sept 1, 2017, we recruited 1542 infants; 771 were assigned to the intervention group and 771 to the control group. One infant who had consent withdrawn before beginning lactoferrin treatment was excluded from analysis. In-hospital death or major morbidity occurred in 162 (21%) of 770 infants in the intervention group and in 170 (22%) of 771 infants in the control group (relative risk [RR] 0·95, 95% CI 0·79-1·14; p=0·60). Three suspected unexpected serious adverse reactions occurred; two in the lactoferrin group, namely unexplained late jaundice and inspissated milk syndrome, but were not attributed to the intervention and one in the control group had fatal inspissated milk syndrome. Our meta-analysis identified 13 trials completed before Feb 18, 2020, including this Article, in 5609 preterm infants. Lactoferrin supplements significantly reduced late-onset sepsis (RR 0·79, 95% CI 0·71-0·88; p<0·0001; I2=58%), but not necrotising enterocolitis or all-cause mortality. INTERPRETATION Lactoferrin supplementation did not improve death or major morbidity in this trial, but might reduce late-onset sepsis, as found in our meta-analysis of over 5000 infants. Future collaborative studies should use products with demonstrated biological activity, be large enough to detect moderate and clinically important effects reliably, and assess greater doses of lactoferrin in infants at increased risk, such as those not exclusively receiving breastmilk or infants of extremely low birthweight. FUNDING Australian National Health and Medical Research Council.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kei Lui
- University of New South Wales, Kensington, NSW, Australia
| | | | - Wendy Hague
- University of Sydney, Sydney, NSW, Australia
| | | | | | | | | | - Helen Liley
- University of Queensland, Brisbane, QLD, Australia
| | | | - Anu Kochar
- University of Adelaide, Adelaide, SA, Australia
| | | | | | - Lisa Askie
- University of Sydney, Sydney, NSW, Australia
| | - Melinda Cruz
- Miracle Babies Foundation, Chipping Norton, NSW, Australia
| | - Tim Schindler
- University of New South Wales, Kensington, NSW, Australia
| | - Kelly Dixon
- University of Queensland, Brisbane, QLD, Australia
| | | | - Mark Tracy
- University of Sydney, Sydney, NSW, Australia
| | | | | | - John Sinn
- University of Sydney, Sydney, NSW, Australia
| | | | | |
Collapse
|
40
|
Development and Characterization of a Novel Peptide-Loaded Antimicrobial Ocular Insert. Biomolecules 2020; 10:biom10050664. [PMID: 32344824 PMCID: PMC7277359 DOI: 10.3390/biom10050664] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/08/2020] [Accepted: 04/21/2020] [Indexed: 02/03/2023] Open
Abstract
Infectious ocular keratitis is the leading cause of blindness worldwide. Bacterial resistance to classical pharmacological treatments raised the interest of researchers towards antimicrobial peptide (AMP)-based therapy. hLF 1-11, a synthetic antimicrobial peptide derived from the N-terminus of human lactoferrin, proved effective against different bacteria and yeast but, like all proteinaceous materials, it is unstable from chemical, physical, and biological points of view. In this study, new freeze-dried solid matrices containing mucoadhesive polymers were prepared and characterized in terms of rheology, hydration time, bioadhesion, drug content, and in vitro release. The formulation HPMC/T2/HA/hLF 1-11fd was selected for the delivery of hLF 1-11, since it showed good drug recovery and no chemical degradation up to at least 6 months (long-term stability). Furthermore, the HPMC/T2/HA/hLF 1-11fd matrix allowed for the release of the drug in a simulated physiological environment, linked to an optimal hydration time, and the peptide antimicrobial activity was preserved for up to 15 months of storage, a very promising result considering the chemical liability of proteinaceous material. For its properties, the freeze-dried matrix developed in this study could be a good platform for the delivery of antimicrobial peptides in the precorneal area to treat infectious phenomena of the ocular surface.
Collapse
|
41
|
Lactoferrin Is Broadly Active against Yeasts and Highly Synergistic with Amphotericin B. Antimicrob Agents Chemother 2020; 64:AAC.02284-19. [PMID: 32094132 PMCID: PMC7179636 DOI: 10.1128/aac.02284-19] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/15/2020] [Indexed: 12/23/2022] Open
Abstract
Lactoferrin (LF) is a multifunctional milk protein with antimicrobial activity against a range of pathogens. While numerous studies report that LF is active against fungi, there are considerable differences in the level of antifungal activity and the capacity of LF to interact with other drugs. Here we undertook a comprehensive evaluation of the antifungal spectrum of activity of three defined sources of LF across 22 yeast and 24 mold species and assessed its interactions with six widely used antifungal drugs. LF was broadly and consistently active against all yeast species tested (MICs, 8 to 64 μg/ml), with the extent of activity being strongly affected by iron saturation. LF was synergistic with amphotericin B (AMB) against 19 out of 22 yeast species tested, and synergy was unaffected by iron saturation but was affected by the extent of LF digestion. LF-AMB combination therapy significantly prolonged the survival of Galleria mellonella wax moth larvae infected with Candida albicans or Cryptococcus neoformans and decreased the fungal burden 12- to 25-fold. Evidence that LF directly interacts with the fungal cell surface was seen via scanning electron microscopy, which showed pore formation, hyphal thinning, and major cell collapse in response to LF-AMB synergy. Important virulence mechanisms were disrupted by LF-AMB treatment, which significantly prevented biofilms in C. albicans and C. glabrata, inhibited hyphal development in C. albicans, and reduced cell and capsule size and phenotypic diversity in Cryptococcus Our results demonstrate the potential of LF-AMB as an antifungal treatment that is broadly synergistic against important yeast pathogens, with the synergy being attributed to the presence of one or more LF peptides.
Collapse
|
42
|
Pammi M, Suresh G. Enteral lactoferrin supplementation for prevention of sepsis and necrotizing enterocolitis in preterm infants. Cochrane Database Syst Rev 2020; 3:CD007137. [PMID: 32232984 PMCID: PMC7106972 DOI: 10.1002/14651858.cd007137.pub6] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Lactoferrin, a normal component of human colostrum and milk, can enhance host defenses and may be effective for prevention of sepsis and necrotizing enterocolitis (NEC) in preterm neonates. OBJECTIVES To assess the safety and effectiveness of lactoferrin supplementation to enteral feeds for prevention of sepsis and NEC in preterm neonates. Secondarily, we assessed the effects of lactoferrin supplementation to enteral feeds on the duration of positive-pressure ventilation, development of chronic lung disease (CLD) or periventricular leukomalacia (PVL), length of hospital stay to discharge among survivors, and adverse neurological outcomes at two years of age or later. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to update our search. We searched the Cochrane Central Register of Controlled Trials (CENTRAL 2019, Issue 9), MEDLINE via PubMed (1966 to 20 January 2020), PREMEDLINE (1996 to 20 January 2020), Embase (1980 to 20 January 2020), and CINAHL (1982 to 20 January 2020). We also searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomized controlled trials and quasi-randomized trials. SELECTION CRITERIA In our search, we included randomized controlled trials (RCTs) evaluating enteral lactoferrin supplementation at any dose or duration to prevent sepsis or NEC in preterm neonates. DATA COLLECTION AND ANALYSIS We used the standard methods of Cochrane Neonatal and the GRADE approach to assess the certainty of evidence. MAIN RESULTS Meta-analysis of data from twelve randomized controlled trials showed that lactoferrin supplementation to enteral feeds decreased late-onset sepsis (typical RR 0.82, 95% CI 0.74 to 0.91; typical RD -0.04, 95% CI, -0.06, -0.02; NNTB 25, 95% CI 17 to 50; 12 studies, 5425 participants, low-certainty evidence) and decreased length of hospital stay (MD -2.38, 95% CI, -4.67, -0.09; 3 studies, 1079 participants, low-certainty evidence). Sensitivity analysis including only good methodological certainty studies suggested a decrease in late-onset sepsis with enteral lactoferrin supplementation (typical RR 0.87, 95% CI, 0.78, 0.97; typical RD -0.03, 95% CI, -0.05, -0.0; 9 studies, 4702 participants, low-certainty evidence). There were no differences in NEC stage II or III (typical RR 1.10, 95% CI, 0.86, 1.41; typical RD -0.00, 95% CI, -0.02, 0.01; 7 studies, 4874 participants; low-certainty evidence) or 'all-cause mortality' (typical RR 0.90, 95% CI 0.69, 1.17; typical RD -0.00, 95% CI, -0.01, 0.01; 11 studies, 5510 participants; moderate-certainty evidence). One study reported no differences in neurodevelopmental testing by Mullen's or Bayley III at 24 months of age after enteral lactoferrin supplementation (one study, 292 participants, low-certainty evidence). Lactoferrin supplementation to enteral feeds with probiotics decreased late-onset sepsis (RR 0.25, 95% CI 0.14 to 0.46; RD -0.13, 95% CI -0.18 to -0.08; NNTB 8, 95% CI 6 to 13; 3 studies, 564 participants; low-certainty evidence) and NEC stage II or III (RR 0.04, 95% CI 0.00 to 0.62; RD -0.05, 95% CI -0.08 to -0.03; NNTB 20, 95% CI 12.5 to 33.3; 1 study, 496 participants; very low-certainty evidence), but not 'all-cause mortality' (very low-certainty evidence). Lactoferrin supplementation to enteral feeds with or without probiotics had no effect on CLD, duration of mechanical ventilation or threshold retinopathy of prematurity (low-certainty evidence). Investigators reported no adverse effects in the included studies. AUTHORS' CONCLUSIONS We found low-certainty evidence from studies of good methodological quality that lactoferrin supplementation of enteral feeds decreases late-onset sepsis but not NEC ≥ stage II or 'all cause mortality' or neurodevelopmental outcomes at 24 months of age in preterm infants without adverse effects. Low- to very low-certainty evidence suggests that lactoferrin supplementation of enteral feeds in combination with probiotics decreases late-onset sepsis and NEC ≥ stage II in preterm infants without adverse effects, however, there were few included studies of poor methodological quality. The presence of publication bias and small studies of poor methodology that may inflate the effect size make recommendations for clinical practice difficult.
Collapse
Key Words
- humans
- infant, newborn
- administration, oral
- bacterial infections
- bacterial infections/epidemiology
- bacterial infections/prevention & control
- cause of death
- chronic disease
- enteral nutrition
- enterocolitis, necrotizing
- enterocolitis, necrotizing/epidemiology
- enterocolitis, necrotizing/prevention & control
- infant, premature
- infant, premature, diseases
- infant, premature, diseases/prevention & control
- lacticaseibacillus rhamnosus
- lactoferrin
- lactoferrin/administration & dosage
- lung diseases
- lung diseases/epidemiology
- mycoses
- mycoses/epidemiology
- mycoses/prevention & control
- numbers needed to treat
- probiotics
- probiotics/administration & dosage
- randomized controlled trials as topic
- retinopathy of prematurity
- retinopathy of prematurity/epidemiology
- sepsis
- sepsis/prevention & control
Collapse
Affiliation(s)
- Mohan Pammi
- Baylor College of Medicine, Section of Neonatology, Department of Pediatrics, 6621, Fannin, MC.WT 6-104, Houston, Texas, USA, 77030
| | - Gautham Suresh
- Baylor College of Medicine, Section of Neonatology, Department of Pediatrics, 6621, Fannin, MC.WT 6-104, Houston, Texas, USA, 77030
| |
Collapse
|
43
|
Pandit G, Biswas K, Ghosh S, Debnath S, Bidkar AP, Satpati P, Bhunia A, Chatterjee S. Rationally designed antimicrobial peptides: Insight into the mechanism of eleven residue peptides against microbial infections. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183177. [PMID: 31954105 DOI: 10.1016/j.bbamem.2020.183177] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/23/2019] [Accepted: 01/07/2020] [Indexed: 01/08/2023]
Abstract
The widespread abuse of antibiotics has led to the use of antimicrobial peptides (AMPs) as a replacement for the existing conventional therapeutic agents for combating microbial infections. The broad-spectrum activity and the resilient nature of AMPs has mainly aggrandized their utilization. Here, we report the design of non-toxic, non-hemolytic and salt tolerant undecapeptides (AMP21-24), derived by modification of a peptide P5 (NH2-LRWLRRLCONH2) reported earlier by our group. Our results depict that the designed peptides show potency against several bacterial as well as fungal strains. Circular dichroism (CD) spectroscopy in combination with molecular dynamic (MD) simulations confirm that the peptides are unstructured. Intrinsic tryptophan fluorescence quenching as well as interaction studies using isothermal calorimetry (ITC) of these peptides in the presence of biological microbial membrane mimics establish the strong microbial membrane affinity of these AMPs. Membrane permeabilization assay and cytoplasmic membrane depolarization studies of Pseudomonas aeruginosa and Candida albicans in the presence of AMPs also hint towards the AMP-membrane interactions. Leakage of calcein dye from membrane mimic liposomes, live cell NMR and field emission scanning electron microscopy (FESEM) studies suggest that the AMPs may be primarily involved in membrane perturbation leading to release of intracellular substances resulting in subsequent microbial cell death. Confocal laser scanning microscopy (CLSM) shows localization of the peptides throughout the cell, indicating the possibility of secondary mode of actions. Electrostatic interactions seem to govern the preferential binding of the AMPs to the microbial membranes in comparison to the mammalian membranes as seen from the MD simulations.
Collapse
Affiliation(s)
- Gopal Pandit
- Department of Chemistry, Indian Institute of Technology, Guwahati, Guwahati, India
| | - Karishma Biswas
- Department of Biophysics, Bose Institute, P-1/12 CIT scheme, VII (M), Kolkata, India
| | - Suvankar Ghosh
- Department of Biosciences and bioengineering, IIT Guwahati, Guwahati, India
| | - Swapna Debnath
- Department of Chemistry, Indian Institute of Technology, Guwahati, Guwahati, India
| | - Anil P Bidkar
- Department of Biosciences and bioengineering, IIT Guwahati, Guwahati, India
| | - Priyadarshi Satpati
- Department of Biosciences and bioengineering, IIT Guwahati, Guwahati, India.
| | - Anirban Bhunia
- Department of Biophysics, Bose Institute, P-1/12 CIT scheme, VII (M), Kolkata, India.
| | - Sunanda Chatterjee
- Department of Chemistry, Indian Institute of Technology, Guwahati, Guwahati, India.
| |
Collapse
|
44
|
In Vivo Microbial Targeting of 99mTc-Labeled Human β-Defensin-3 in a Rat Model of Infection. Clin Nucl Med 2019; 44:e602-e606. [PMID: 31306193 DOI: 10.1097/rlu.0000000000002713] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Differentiation of infection from aseptic inflammation represents a major clinical issue. None of the commercially available compounds (labeled granulocytes, antigranulocyte antibodies, Ga-citrate, labeled immunoglobulin G, F-FDG) is capable of this differentiation, producing a nonnegligible false-positive rate. Recently, our group reported on a reliable labeling procedure of the antimicrobial peptide human β-defensin 3 (HBD-3) with Tc. The aim of this study was to evaluate in vivo Tc-HBD-3 uptake in a rat model of infection. METHODS Recombinant HBD-3 was radiolabeled with Tc. Radiolabeling yield and specific activity of the compound were calculated. Chromatographic behavior and biological activity of Tc-HBD-3 were also assessed. An experimental model involving Staphylococcus aureus-induced infection and carrageenan-induced aseptic inflammation was performed in 5 Wistar rats. Serial planar scintigraphic acquisitions were performed from 15 to 180 minutes after Tc-HBD-3 intravenous administration. Radiotracer uptake was evaluated qualitatively and semiquantitatively as a target-to-nontarget ratio. RESULTS Radiolabeling yield of Tc-HBD-3 was 70% with a specific activity of 6 to 8 MBq/μg. A significant and progressive Tc-HBD-3 uptake was observed in the site of S. aureus-induced infection, with a maximum average target-to-nontarget ratio of 5.7-fold higher in the infection site compared with an inflammation site observed at 140 minutes. CONCLUSIONS In vivo imaging with Tc-HBD-3 in a rat model of S. aureus-induced infection demonstrated favorable uptake in the infection site compared with sterile inflammation and background. These promising results, together with previous ex vivo uptake and toxicity assessment, suggest the potential of Tc-HBD-3 as a novel agent for specific infection imaging.
Collapse
|
45
|
Prevention of Nosocomial Infections in Critically Ill Patients With Lactoferrin: A Randomized, Double-Blind, Placebo-Controlled Study. Crit Care Med 2019; 46:1450-1456. [PMID: 30015668 DOI: 10.1097/ccm.0000000000003294] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To obtain preliminary evidence for the efficacy of lactoferrin as a preventative measure for nosocomial infections and inform the conduct of a definitive study. DESIGN Phase 2, multicenter, randomized, double-blind, placebo-controlled study. SETTING Medical-surgical ICUs. PATIENTS Adult, critically ill patients receiving invasive mechanical ventilation. INTERVENTIONS Randomized, eligible, consenting patients expected to require invasive mechanical ventilation more than 48 hours received lactoferrin both enterally and via an oral swab or a placebo of sterile water for up to 28 days. MEASUREMENTS AND MAIN RESULTS Of the 214 patients who were randomized, 212 received at least one dose of the intervention and were analyzed (107 lactoferrin and 105 placebo). Protocol adherence was 87.5%. Patients receiving lactoferrin were older (mean [SD], 66.3 [13.5] vs 62.5 [16.2] yr), had a higher Acute Physiology and Chronic Health Evaluation II score (26.8 [7.8] vs 23.5 [7.9]), and need for vasopressors (79% vs 70%). Antibiotic-free days (17.3 [9.0] vs 18.5 [7.1]; p = 0.91) and nosocomial infections (0.3 [0.7] vs 0.4 [0.6] per patient; p = 0.48) did not differ between lactoferrin and placebo groups, respectively. Clinical outcomes for lactoferrin versus placebo were as follows: ICU length of stay (14.5 [18.0] vs 15.0 [37.3] d; p = 0.82), hospital length of stay (25.0 [25.9] vs 28.1 [44.6] d; p = 0.57), hospital mortality (41.1% vs 30.5%; p = 0.11), and 90-day mortality (44.9% vs 32.4%; p = 0.06). Biomarker levels did not differ between the groups. CONCLUSIONS Lactoferrin did not improve the primary outcome of antibiotic-free days, nor any of the secondary outcomes. Our data do not support the conduct of a larger phase 3 trial.
Collapse
|
46
|
Nibbering PH, Göblyös A, Adriaans AE, Cordfunke RA, Ravensbergen B, Rietveld MH, Zwart S, Commandeur S, van Leeuwen R, Haisma EM, Schimmel KJM, den Hartigh J, Drijfhout JW, Ghalbzouri AE. Eradication of meticillin-resistant Staphylococcus aureus from human skin by the novel LL-37-derived peptide P10 in four pharmaceutical ointments. Int J Antimicrob Agents 2019; 54:610-618. [PMID: 31356860 DOI: 10.1016/j.ijantimicag.2019.07.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 07/17/2019] [Accepted: 07/21/2019] [Indexed: 11/20/2022]
Abstract
Skin bacterial colonization/infection is a frequent cause of morbidity in patients with chronic wounds and allergic/inflammatory skin diseases. This study aimed to develop a novel approach to eradicate meticillin-resistant Staphylococcus aureus (MRSA) from human skin. To achieve this, the stability and antibacterial activity of the novel LL-37-derived peptide P10 in four ointments was compared. Results indicate that P10 is chemically stable and antibacterial in hypromellose gel and Softisan-containing cream, but not in Cetomacrogol cream (with or without Vaseline), at 4 °C for 16 months. Reduction in MRSA counts on Leiden human epidermal models (LEMs) by P10 in hypromellose gel was greater than that of the peptide in Cetomacrogol cream or phosphate buffered saline. P10 did not show adverse effects on LEMs irrespective of the ointment used, while Cetomacrogol with Vaseline and Softisan cream, but not hypromellose gel or Cetomacrogol cream, destroyed MRSA-colonized LEMs. Taking all this into account, P10 in hypromellose gel dose-dependently reduced MRSA colonizing the stratum corneum of the epidermis as well as biofilms of this bacterial strain on LEMs. Moreover, P10 dose-dependently reduced MRSA counts on ex-vivo human skin, with P10 in hypromellose gel being more effective than P10 in Cetomacrogol and Softisan creams. P10 in hypromellose gel is a strong candidate for eradication of MRSA from human skin.
Collapse
Affiliation(s)
- Peter H Nibbering
- Department of Infectious Diseases, Leiden University Medical Centre, Leiden, The Netherlands.
| | - Anikó Göblyös
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Alwin E Adriaans
- Department of Infectious Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | - Robert A Cordfunke
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands
| | - Bep Ravensbergen
- Department of Infectious Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | - Marion H Rietveld
- Department of Dermatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Sarah Zwart
- Biomimiq-Aeon Astron Europe BV, Leiden, The Netherlands
| | | | | | - Elisabeth M Haisma
- Department of Infectious Diseases, Leiden University Medical Centre, Leiden, The Netherlands; Department of Dermatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Kirsten J M Schimmel
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Jan den Hartigh
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Jan Wouter Drijfhout
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands
| | | |
Collapse
|
47
|
Moussa DG, Kirihara JA, Ye Z, Fischer NG, Khot J, Witthuhn BA, Aparicio C. Dentin Priming with Amphipathic Antimicrobial Peptides. J Dent Res 2019; 98:1112-1121. [PMID: 31313946 DOI: 10.1177/0022034519863772] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The evolution of bonded restorations has undergone great progress over several decades. Nonetheless, life spans of bonded restorations are limited mainly because of the eventual incidence of recurrent caries. Over time, water and waterborne agents (acids, enzymes) degrade the components of the dentin/restoration interface, allowing bacterial colonization and dentin reinfection at the margins of the restoration. We developed a 2-tier protective technology consisting of priming/coating dentin with amphipathic and antimicrobial peptides (AAMPs) to obtain hydrophobic/water-repellent and antibiofilm dentin-resisting recurrent caries around bonded restorations. We tested a series of AAMPs to assess their structure-function relationships as well as the effects of different dentin-conditioning methods on the structural features of AAMP-coated dentin. We found relation between the secondary structure of AAMPs (high portion of β-sheet), the antimicrobial potency of AAMPs, and the AAMPs' ability to form hydrophobic coatings on dentin. We also determined that AAMPs had preferential adsorption on the mineral phase of dentin, which suggested that peptides arrange their cationic and hydrophilic motifs in direct contact with the negatively charged minerals in the hydrophilic dentin. These results led us to explore different dentin-conditioning methods that would increase the mineral/collagen ratio and their effect on AAMP immobilization. We innovatively imaged the spatial distribution of the AAMPs in relation to the dentinal tubules and collagen network using a minimally invasive multimodal imaging technique: multiphoton-second harmonic generation. Using multiphoton-second harmonic generation imaging, we determined that partial deproteinization of dentin increased the amount of immobilized AAMPs as compared with the total etched dentin at the dentin surface and extended deeply around dentinal tubules. Last, we analyzed the release rate of AAMPs from dentin coatings in artificial saliva to predict their stability in the clinical setting. In conclusion, priming dentin with AAMPs is a versatile new approach with potential to fortify the otherwise vulnerable adhesive-based interfaces.
Collapse
Affiliation(s)
- D G Moussa
- 1 MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, Department of Restorative Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - J A Kirihara
- 2 Center for Mass Spectrometry and Proteomics, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Z Ye
- 1 MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, Department of Restorative Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - N G Fischer
- 1 MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, Department of Restorative Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - J Khot
- 1 MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, Department of Restorative Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - B A Witthuhn
- 2 Center for Mass Spectrometry and Proteomics, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - C Aparicio
- 1 MDRCBB-Minnesota Dental Research Center for Biomaterials and Biomechanics, Department of Restorative Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
48
|
Anderson JR, Smagul A, Simpson D, Clegg PD, Rubio-Martinez LM, Peffers MJ. The synovial fluid proteome differentiates between septic and nonseptic articular pathologies. J Proteomics 2019; 202:103370. [PMID: 31028944 PMCID: PMC6549134 DOI: 10.1016/j.jprot.2019.04.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/28/2019] [Accepted: 04/22/2019] [Indexed: 12/28/2022]
Abstract
Articular conditions are common in horses and can result in loss of function, chronic pain and/or inability to work. Common conditions include osteoarthritis, osteochondrosis and synovial sepsis, which can be life-threatening, but despite the high clinical prevalence of these conditions, rapid and specific diagnosis, monitoring and prognostication remains a challenge for practicing veterinarians. Synovial fluid from a range of arthropathies was enriched for low abundance proteins using combinatorial peptide ligand ProteoMiner™ beads and analysed via liquid chromatography-tandem mass spectrometry. Changes in protein abundances were analysed using label-free quantification. Principle component analysis of differentially expressed proteins identified groupings associated with joint pathology. Findings were validated using ELISA. Lactotransferrin (LTF) abundance was increased in sepsis compared to all other groups and insulin-like growth factor-binding protein 6 (IGFBP6) abundance decreased in sepsis compared to other disease groups. Pathway analysis identified upregulation of the complement system in synovial joint sepsis and the downregulation of eukaryotic translation initiation factors and mTOR signalling pathways in both OA and OC compared to the healthy group. Overall, we have identified a catalogue of proteins which we propose to be involved in osteoarthritis, osteochondrosis and synovial sepsis pathogenesis. SIGNIFICANCE: Osteoarthritis, osteochondrosis and synovial sepsis, which can be life-threatening, are common articular conditions in which rapid and specific diagnosis, monitoring and prognostication remains a challenge for practicing veterinarians. This study has identified that the equine synovial fluid proteome exhibits distinctive profile changes between osteoarthritis, osteochondrosis, synovial sepsis and healthy joints. Elevated synovial abundance of lactotransferrin and decreased insulin-like growth factor-binding protein 6 were both found to distinguish synovial sepsis from all other study groups. Thus, these protein markers may have a future role in clinical practice to enable an earlier and reliable diagnosis of synovial sepsis.
Collapse
Affiliation(s)
- James R Anderson
- Institute of Ageing and Chronic Disease, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK.
| | - Aibek Smagul
- Institute of Ageing and Chronic Disease, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK.
| | - Deborah Simpson
- Centre for Proteome Research, Institute of Integrative Biology, Biosciences Building, Crown Street, University of Liverpool, Liverpool L69 7ZB, UK.
| | - Peter D Clegg
- Institute of Ageing and Chronic Disease, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK.
| | - Luis M Rubio-Martinez
- Department of Equine Clinical Studies, Institute of Veterinary Science, Chester High Road, Neston CH64 7TE, UK.
| | - Mandy J Peffers
- Institute of Ageing and Chronic Disease, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK.
| |
Collapse
|
49
|
Pammi M, Abrams SA, Cochrane Neonatal Group. Enteral lactoferrin for the treatment of sepsis and necrotizing enterocolitis in neonates. Cochrane Database Syst Rev 2019; 5:CD007138. [PMID: 31077334 PMCID: PMC6511233 DOI: 10.1002/14651858.cd007138.pub4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Neonatal sepsis and necrotizing enterocolitis (NEC) cause significant neonatal mortality and morbidity despite appropriate antibiotic therapy. Enhancing host defense and modulating inflammation by using lactoferrin as an adjunct to antibiotics in the treatment of sepsis, NEC, or both, may improve clinical outcomes. OBJECTIVES The primary objective was to assess safety and efficacy of oral lactoferrin as an adjunct to antibiotics in the treatment of neonates with suspected or confirmed sepsis, NEC, or both. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL 2018, Issue 9), MEDLINE via PubMed, PREMEDLINE, (1966 to 20 September 2018) Embase (1980 to 20 September 2018), and CINAHL (1982 to 20 September 2018). We also searched clinical trial databases, conference proceedings, the reference lists of retried articles and clinical trials, and the authors' personal files. SELECTION CRITERIA We included randomized or quasi-randomized controlled trials evaluating enteral lactoferrin (at any dose or duration), used as an adjunct to antibiotic therapy, compared with antibiotic therapy alone (with or without placebo) or other adjuncts to antibiotic therapy to treat neonates at any gestational age up to 44 weeks' postmenstrual age with confirmed or suspected sepsis or necrotizing enterocolitis (Bell's Stage II or III). DATA COLLECTION AND ANALYSIS We used the standardized methods of Cochrane Neonatal for conducting a systematic review and for assessing the methodological quality of studies (neonatal.cochrane.org/en/index.html). The titles and the abstracts of studies identified by the search strategy were independently assessed by the two review authors and full text versions were obtained for assessment if necessary. Forms were designed to record trial inclusion/exclusion and data extraction. We used the GRADE approach to assess the quality of evidence. MAIN RESULTS We did not identify any eligible trials evaluating lactoferrin for the treatment of neonatal sepsis or NEC. AUTHORS' CONCLUSIONS Implications for practice: currently there is no evidence to support or refute the use of enteral lactoferrin, as an adjunct to antibiotic therapy, for the treatment of neonatal sepsis or necrotizing enterocolitis. IMPLICATIONS FOR RESEARCH given the lack of efficacy of enteral lactoferrin for preventing late-onset sepsis and necrotizing enterocolitis, evaluation of enteral lactoferrin as an adjunctive agent for treatment of sepsis or necrotizing enterocolitis does not appear to be a research priority.
Collapse
Affiliation(s)
- Mohan Pammi
- Baylor College of MedicineSection of Neonatology, Department of Pediatrics6621, Fannin, MC.WT 6‐104HoustonTexasUSA77030
| | - Steven A Abrams
- Dell Medical School, The University of Texas at AustinDepartment of PediatricsAustinTexasUSA
| | | |
Collapse
|
50
|
Pires S, Parker D. Innate Immune Responses to Acinetobacter baumannii in the Airway. J Interferon Cytokine Res 2019; 39:441-449. [PMID: 31013462 DOI: 10.1089/jir.2019.0008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Acinetobacter baumannii is an emerging opportunistic pathogen that has risen to become a serious global threat, prevalent in health care settings and the community, which results in high morbidity and mortality rates. Its alarming expansion of antibiotic resistance is one of the most problematic traits of A. baumannii and as so, this bacterium has been classified as a serious threat and high priority target by the CDC. The most common types of infections induced by this pathogen include pneumonia (both hospital and community acquired), bacteremia, skin and soft tissue, urinary tract infections, endocarditis, and meningitis. Nosocomial pneumonia is the most prevalent of these. This review summarizes the current state of the signaling and innate immune components activated in response to A. baumannii infection in the airway.
Collapse
Affiliation(s)
- Sílvia Pires
- Department of Pathology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Dane Parker
- Department of Pathology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey
| |
Collapse
|