1
|
Adak A, Castelletto V, de Mello L, Mendes B, Barrett G, Seitsonen J, Hamley IW. Effect of Chirality and Amphiphilicity on the Antimicrobial Activity of Tripodal Lysine-Based Peptides. ACS APPLIED BIO MATERIALS 2025. [PMID: 39792083 DOI: 10.1021/acsabm.4c01635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
A series of tripodal (three-arm) lysine-based peptides were designed and synthesized and their self-assembly properties in aqueous solution and antimicrobial activity were investigated. We compare the behaviors of homochiral tripodal peptides (KKY)3K and a homologue containing the bulky aromatic fluorenylmethoxycarbonyl (Fmoc) group Fmoc-(KKY)3K, and heterochiral analogues containing k (d-Lys), (kkY)3K and Fmoc-(kkY)3K. The molecular conformation and self-assembly in aqueous solutions were probed using various spectroscopic techniques, along with small-angle X-ray scattering (SAXS) and cryogenic-transmission electron microscopy (cryo-TEM). In cell viability assays using fibroblast cell lines, the tripodal peptides without Fmoc were observed to be noncytotoxic over the concentration range studied, and the Fmoc functionalized tripodal peptides were only cytotoxic at the highest concentrations (above the critical aggregation concentration of the lipopeptides). The molecules also show good hemocompatibility at sufficiently low concentration, and antimicrobial activity was assessed via MIC (minimum inhibitory concentration) and MBC (minimum bactericidal concentration) assays. These revealed that the Fmoc-functionalized tripodal peptides had significant activity against both Gram-negative and Gram-positive bacteria, and in the case of Gram-positive Staphylococcus aureus, the antimicrobial activity for Fmoc-(kkY)3K was improved compared to polymyxin B. The mechanism of the antimicrobial assay was found to involve rupture of the bacterial membrane as evident from fluorescence microscopy live/dead cell assays, and scanning electron microscopy images.
Collapse
Affiliation(s)
- Anindyasundar Adak
- School of Chemistry, Pharmacy and Food Biosciences, University of Reading, Whiteknights, Reading RG6 6AD, U.K
| | - Valeria Castelletto
- School of Chemistry, Pharmacy and Food Biosciences, University of Reading, Whiteknights, Reading RG6 6AD, U.K
| | - Lucas de Mello
- School of Chemistry, Pharmacy and Food Biosciences, University of Reading, Whiteknights, Reading RG6 6AD, U.K
| | - Bruno Mendes
- School of Biological Sciences, University of Reading, Reading RG6 6AS, U.K
| | - Glyn Barrett
- School of Biological Sciences, University of Reading, Reading RG6 6AS, U.K
| | - Jani Seitsonen
- Nanomicroscopy Center, Aalto University, FIN-02150 Espoo, Finland
| | - Ian W Hamley
- School of Chemistry, Pharmacy and Food Biosciences, University of Reading, Whiteknights, Reading RG6 6AD, U.K
| |
Collapse
|
2
|
Parsad R, Ahlawat S, Bagiyal M, Gera R, Chhabra P, Sharma U, Arora R, Sharma R. Cathelicidins in farm animals: Structural diversity, mechanisms of action, and therapeutic potential in the face of antimicrobial resistance. Vet Immunol Immunopathol 2025; 279:110866. [PMID: 39708585 DOI: 10.1016/j.vetimm.2024.110866] [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: 08/22/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Cathelicidins are a diverse family of antimicrobial peptides found across many vertebrate species, playing a pivotal role in the innate immune system. These peptides exhibit a variety of structural motifs, including α-helices, β-hairpins, and random coils, contributing to their broad-spectrum antimicrobial activity. The structural diversity of cathelicidins allows them to interact with a wide range of microbial targets, thereby enhancing their antimicrobial efficacy. Distinct species produce specific cathelicidins, each adapted to meet their unique immune requirements. Cathelicidins primarily function by disrupting microbial membranes, leading to cell lysis. Beyond their direct antimicrobial action, they possess immunomodulatory properties that bolster host defense mechanisms. These properties include promoting chemotaxis, enhancing phagocytosis, and inducing cytokine production, thereby modulating the host immune response. The therapeutic potential of cathelicidins is significant, especially in light of the growing challenge of antimicrobial resistance (AMR). As conventional antibiotics lose efficacy, cathelicidins emerge as promising alternatives due to their unique mechanisms of action and reduced likelihood of inducing resistance. Recent research underscores their potential in treating infections, inflammatory diseases, and even cancer. Advances in synthetic biology offer promising prospects for effective cathelicidin-based therapies in the future. This review summarizes the diversity, modes of action, and clinical prospects of cathelicidins specific to farm animals.
Collapse
Affiliation(s)
- Ram Parsad
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - Sonika Ahlawat
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India.
| | - Meena Bagiyal
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - Ritika Gera
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - Pooja Chhabra
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - Upasna Sharma
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - Reena Arora
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| | - Rekha Sharma
- ICAR-National Bureau of Animal Genetic Resources, Karnal, India
| |
Collapse
|
3
|
Caselli L, Malmsten M. Combining functionalities-nanoarchitectonics for combatting bacterial infection. Adv Colloid Interface Sci 2024; 337:103385. [PMID: 39721197 DOI: 10.1016/j.cis.2024.103385] [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: 09/01/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
New antimicrobial and anti-inflammatory therapeutics are needed because of antibiotic resistance development and resulting complications such as inflammation, ultimately leading to septic shock. The antimicrobial effects of various nanoparticles (NPs) are currently attracting intensive research interest. Although various NPs display potent antimicrobial effects against strains resistant to conventional antibiotics, the therapeutic use of such materials is restricted by poor selectivity between bacteria and human cells, leading to adverse side effects. As a result, increasing research efforts during the last few years have focused on targeting NPs against bacteria and other components in the infection micro-environment. Examples of approaches explored include peptide-, protein- and nucleic acid-based NP coatings for bacterial membrane recognition, as well as NP conjugation with enzyme substrates or other moieties that respond to bacterial or other enzymes present in the infection micro-environment. In general, this study aims to add to the literature on the antimicrobial effects of nanomaterials by discussing surface modification strategies for targeting bacterial membranes and membrane components, as well as how such surface modifications can improve the antimicrobial effects of nanomaterials and simultaneously decrease toxicity towards human cells and tissues. In doing so, the biological effects observed are related throughout to the physico-chemical modes of action underlying such effects.
Collapse
Affiliation(s)
| | - Martin Malmsten
- Physical Chemistry 1, University of Lund, S-221 00 Lund, Sweden; Department of Pharmacy, University of Copenhagen, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
4
|
Liu L, Li L, Wang T, Li Z, Yan B, Tan R, Zeng A, Ma W, Zhu X, Yin Z, Ma C. Recent nanoengineered therapeutic advancements in sepsis management. Front Bioeng Biotechnol 2024; 12:1495277. [PMID: 39703795 PMCID: PMC11655211 DOI: 10.3389/fbioe.2024.1495277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Sepsis (defined as sepsis 3.0) is a life-threatening organ dysfunction caused by a dysregulated host response to a variety of pathogenic microorganisms. Characterized by high morbidity and mortality, sepsis has become a global public health problem. However, there is a lack of appropriate diagnostic and therapeutic strategies for sepsis and current management rely on the limited treatment strategies. Recently, nanomedicines targeting and controlling the release of bio-active agents have shown excellent potency in sepsis management, with improved therapeutic efficacy and reduced adverse effects. In this review, we have summarized the advantages of nanomaterials. Also, the preparation and efficacy of the main categories of anti-sepsis nanomedicines applied in sepsis management are described in detail, including antibiotic-coated nanomaterials, antimicrobial peptides-coated nanomaterials, biomimetic nanomaterials, nanomaterials targeting macrophages and natural products loaded nanomaterials. These advances in nanomedicines establish the huge potential for nanomaterials-based sepsis management, especially in the improved pharmaceutical and pharmacological properties, enhanced therapeutic efficacy, controllable drug-targeting and reduced side effects. To further facilitate clinical translation of anti-sepsis nanomedicines, we propose that the issues involving safety, regulatory laws and cost-effectiveness should receive much more attention in the future.
Collapse
Affiliation(s)
- Li Liu
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Li Li
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Ting Wang
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Zheyu Li
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Bingpeng Yan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Wenbo Ma
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Xin Zhu
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Zhujun Yin
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, The “Double-First Class” Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Chunhua Ma
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
5
|
Kumar SD, Kim EY, Radhakrishnan NK, Bang JK, Yang S, Shin SY. Enhanced Antibacterial, Anti-Inflammatory, and Antibiofilm Activities of Tryptophan-Substituted Peptides Derived from Cecropin A-Melittin Hybrid Peptide BP100. Molecules 2024; 29:5231. [PMID: 39598621 PMCID: PMC11596392 DOI: 10.3390/molecules29225231] [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: 10/04/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
The emergence of multidrug-resistant pathogens necessitates the development of novel antimicrobial agents. BP100, a short α-helical antimicrobial peptide (AMP) derived from cecropin A and melittin, has shown promise as a potential therapeutic. To enhance its efficacy, we designed and synthesized 16 tryptophan-substituted BP100 analogs based on helical wheel projections. Among these, BP5, BP6, BP8, BP11, and BP13 exhibited 1.5- to 5.5-fold higher antibacterial activity and improved cell selectivity compared to BP100. These analogs demonstrated superior efficacy in suppressing pro-inflammatory cytokine release in LPS-stimulated RAW 264.7 cells and eradicating preformed biofilms of multidrug-resistant Pseudomonas aeruginosa (MDRPA). Additionally, these analogs showed greater resistance to physiological salts and serum compared to BP100. Mechanistic studies revealed that BP100 and its analogs exert their antibacterial effects through membrane disruption, depolarization, and permeabilization. Notably, these analogs showed synergistic antimicrobial activity with ciprofloxacin against MDRPA. Our findings suggest that these tryptophan-substituted BP100 analogs represent promising candidates for combating multidrug-resistant bacterial infections, offering a multifaceted approach through their antibacterial, anti-inflammatory, and antibiofilm activities.
Collapse
Affiliation(s)
- Sukumar Dinesh Kumar
- Department of Cellular & Molecular Medicine, School of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (S.D.K.); (E.Y.K.)
| | - Eun Young Kim
- Department of Cellular & Molecular Medicine, School of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (S.D.K.); (E.Y.K.)
| | | | - Jeong Kyu Bang
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang 28119, Republic of Korea;
| | - Sungtae Yang
- Department of Microbiology, School of Medicine, Chosun University, Gwangju 61452, Republic of Korea
- Institute of Well-Aging Medicare & CSU G-LAMP Project Group, Chosun University, Gwangju 61452, Republic of Korea
| | - Song Yub Shin
- Department of Cellular & Molecular Medicine, School of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (S.D.K.); (E.Y.K.)
- Graduate School of Biomedical Science, Chosun University, Gwangju 61452, Republic of Korea;
| |
Collapse
|
6
|
Caselli L, Parra-Ortiz E, Micciulla S, Skoda MWA, Häffner SM, Nielsen EM, van der Plas MJA, Malmsten M. Boosting Membrane Interactions and Antimicrobial Effects of Photocatalytic Titanium Dioxide Nanoparticles by Peptide Coating. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309496. [PMID: 38402437 DOI: 10.1002/smll.202309496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/30/2024] [Indexed: 02/26/2024]
Abstract
Photocatalytic nanoparticles offer antimicrobial effects under illumination due to the formation of reactive oxygen species (ROS), capable of degrading bacterial membranes. ROS may, however, also degrade human cell membranes and trigger toxicity. Since antimicrobial peptides (AMPs) may display excellent selectivity between human cells and bacteria, these may offer opportunities to effectively "target" nanoparticles to bacterial membranes for increased selectivity. Investigating this, photocatalytic TiO2 nanoparticles (NPs) are coated with the AMP LL-37, and ROS generation is found by C11-BODIPY to be essentially unaffected after AMP coating. Furthermore, peptide-coated TiO2 NPs retain their positive ζ-potential also after 1-2 h of UV illumination, showing peptide degradation to be sufficiently limited to allow peptide-mediated targeting. In line with this, quartz crystal microbalance measurements show peptide coating to promote membrane binding of TiO2 NPs, particularly so for bacteria-like anionic and cholesterol-void membranes. As a result, membrane degradation during illumination is strongly promoted for such membranes, but not so for mammalian-like membranes. The mechanisms of these effects are elucidated by neutron reflectometry. Analogously, LL-37 coating promoted membrane rupture by TiO2 NPs for Gram-negative and Gram-positive bacteria, but not for human monocytes. These findings demonstrate that AMP coating may selectively boost the antimicrobial effects of photocatalytic NPs.
Collapse
Affiliation(s)
- Lucrezia Caselli
- Department of Pharmacy, University of Copenhagen, Copenhagen, DK-2100, Denmark
- Department of Physical Chemistry 1, Lund University, Lund, SE-22100, Sweden
| | - Elisa Parra-Ortiz
- Department of Pharmacy, University of Copenhagen, Copenhagen, DK-2100, Denmark
- Novonesis, Biologiens Vej 2, Lyngby, DK-2800 Kgs, Denmark
| | - Samantha Micciulla
- Institut Laue-Langevin, CS 20156, Grenoble Cedex 9, 38042, France
- Laboratoire Interdisciplinaire de Physique (LIPhy), Saint Martin d'Hères, 38402, France
- Centre National de la Recherche Scientifique (CNRS), Saint-Martin-d'Hères, Auvergne-Rhône-Alpes, France
| | - Maximilian W A Skoda
- ISIS Pulsed Neutron and Muon Source, Rutherford Appleton Laboratory, Harwell, OX11 0QX, UK
| | - Sara Malekkhaiat Häffner
- Department of Pharmacy, University of Copenhagen, Copenhagen, DK-2100, Denmark
- RISE Research Institutes of Sweden, Malvinas väg 3, Stockholm, 114 86, Sweden
| | | | | | - Martin Malmsten
- Department of Pharmacy, University of Copenhagen, Copenhagen, DK-2100, Denmark
- Department of Physical Chemistry 1, Lund University, Lund, SE-22100, Sweden
| |
Collapse
|
7
|
Gafar MA, Omolo CA, Elhassan E, Ibrahim UH, Govender T. Applications of peptides in nanosystems for diagnosing and managing bacterial sepsis. J Biomed Sci 2024; 31:40. [PMID: 38637839 PMCID: PMC11027418 DOI: 10.1186/s12929-024-01029-2] [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: 02/25/2024] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
Sepsis represents a critical medical condition stemming from an imbalanced host immune response to infections, which is linked to a significant burden of disease. Despite substantial efforts in laboratory and clinical research, sepsis remains a prominent contributor to mortality worldwide. Nanotechnology presents innovative opportunities for the advancement of sepsis diagnosis and treatment. Due to their unique properties, including diversity, ease of synthesis, biocompatibility, high specificity, and excellent pharmacological efficacy, peptides hold great potential as part of nanotechnology approaches against sepsis. Herein, we present a comprehensive and up-to-date review of the applications of peptides in nanosystems for combating sepsis, with the potential to expedite diagnosis and enhance management outcomes. Firstly, sepsis pathophysiology, antisepsis drug targets, current modalities in management and diagnosis with their limitations, and the potential of peptides to advance the diagnosis and management of sepsis have been adequately addressed. The applications have been organized into diagnostic or managing applications, with the last one being further sub-organized into nano-delivered bioactive peptides with antimicrobial or anti-inflammatory activity, peptides as targeting moieties on the surface of nanosystems against sepsis, and peptides as nanocarriers for antisepsis agents. The studies have been grouped thematically and discussed, emphasizing the constructed nanosystem, physicochemical properties, and peptide-imparted enhancement in diagnostic and therapeutic efficacy. The strengths, limitations, and research gaps in each section have been elaborated. Finally, current challenges and potential future paths to enhance the use of peptides in nanosystems for combating sepsis have been deliberately spotlighted. This review reaffirms peptides' potential as promising biomaterials within nanotechnology strategies aimed at improving sepsis diagnosis and management.
Collapse
Affiliation(s)
- Mohammed A Gafar
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
- Department of Pharmaceutics, Faculty of Pharmacy, University of Khartoum, P.O. Box 1996, Khartoum, Sudan
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa.
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, P. O. Box 14634-00800, Nairobi, Kenya.
| | - Eman Elhassan
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Usri H Ibrahim
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa.
| |
Collapse
|
8
|
Cafaro V, Bosso A, Di Nardo I, D’Amato A, Izzo I, De Riccardis F, Siepi M, Culurciello R, D’Urzo N, Chiarot E, Torre A, Pizzo E, Merola M, Notomista E. The Antimicrobial, Antibiofilm and Anti-Inflammatory Activities of P13#1, a Cathelicidin-like Achiral Peptoid. Pharmaceuticals (Basel) 2023; 16:1386. [PMID: 37895857 PMCID: PMC10610514 DOI: 10.3390/ph16101386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Cationic antimicrobial peptides (CAMPs) are powerful molecules with antimicrobial, antibiofilm and endotoxin-scavenging activities. These properties make CAMPs very attractive drugs in the face of the rapid increase in multidrug-resistant (MDR) pathogens, but they are limited by their susceptibility to proteolytic degradation. An intriguing solution to this issue could be the development of functional mimics of CAMPs with structures that enable the evasion of proteases. Peptoids (N-substituted glycine oligomers) are an important class of peptidomimetics with interesting benefits: easy synthetic access, intrinsic proteolytic stability and promising bioactivities. Here, we report the characterization of P13#1, a 13-residue peptoid specifically designed to mimic cathelicidins, the best-known and most widespread family of CAMPs. P13#1 showed all the biological activities typically associated with cathelicidins: bactericidal activity over a wide spectrum of strains, including several ESKAPE pathogens; the ability to act in combination with different classes of conventional antibiotics; antibiofilm activity against preformed biofilms of Pseudomonas aeruginosa, comparable to that of human cathelicidin LL-37; limited toxicity; and an ability to inhibit LPS-induced proinflammatory effects which is comparable to that of "the last resource" antibiotic colistin. We further studied the interaction of P13#1 with SDS, LPSs and bacterial cells by using a fluorescent version of P13#1. Finally, in a subcutaneous infection mouse model, it showed antimicrobial and anti-inflammatory activities comparable to ampicillin and gentamicin without apparent toxicity. The collected data indicate that P13#1 is an excellent candidate for the formulation of new antimicrobial therapies.
Collapse
Affiliation(s)
- Valeria Cafaro
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Andrea Bosso
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Ilaria Di Nardo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Assunta D’Amato
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, 84084 Fisciano, Italy; (A.D.); (I.I.); (F.D.R.)
| | - Irene Izzo
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, 84084 Fisciano, Italy; (A.D.); (I.I.); (F.D.R.)
| | - Francesco De Riccardis
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, 84084 Fisciano, Italy; (A.D.); (I.I.); (F.D.R.)
| | - Marialuisa Siepi
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Rosanna Culurciello
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Nunzia D’Urzo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | | | | | - Elio Pizzo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Marcello Merola
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Eugenio Notomista
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| |
Collapse
|
9
|
Safi R, Sánchez-Álvarez M, Bosch M, Demangel C, Parton RG, Pol A. Defensive-lipid droplets: Cellular organelles designed for antimicrobial immunity. Immunol Rev 2023; 317:113-136. [PMID: 36960679 DOI: 10.1111/imr.13199] [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] [Indexed: 03/25/2023]
Abstract
Microbes have developed many strategies to subvert host organisms, which, in turn, evolved several innate immune responses. As major lipid storage organelles of eukaryotes, lipid droplets (LDs) are an attractive source of nutrients for invaders. Intracellular viruses, bacteria, and protozoan parasites induce and physically interact with LDs, and the current view is that they "hijack" LDs to draw on substrates for host colonization. This dogma has been challenged by the recent demonstration that LDs are endowed with a protein-mediated antibiotic activity, which is upregulated in response to danger signals and sepsis. Dependence on host nutrients could be a generic "Achilles' heel" of intracellular pathogens and LDs a suitable chokepoint harnessed by innate immunity to organize a front-line defense. Here, we will provide a brief overview of the state of the conflict and discuss potential mechanisms driving the formation of the 'defensive-LDs' functioning as hubs of innate immunity.
Collapse
Affiliation(s)
- Rémi Safi
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Miguel Sánchez-Álvarez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols (IIB), Madrid, Spain
| | - Marta Bosch
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Caroline Demangel
- Immunobiology and Therapy Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Robert G Parton
- Institute for Molecular Bioscience (IMB), Brisbane, Queensland, Australia
- Centre for Microscopy and Microanalysis (CMM), University of Queensland, Brisbane, Queensland, Australia
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
10
|
Bajorat R, Danckert L, Ebert F, Bancken T, Bergt S, Klawitter F, Vollmar B, Reuter DA, Schürholz T, Ehler J. The Effect of Early Application of Synthetic Peptides 19-2.5 and 19-4LF to Improve Survival and Neurological Outcome in a Mouse Model of Cardiac Arrest and Resuscitation. Biomedicines 2023; 11:biomedicines11030855. [PMID: 36979834 PMCID: PMC10045145 DOI: 10.3390/biomedicines11030855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/16/2023] Open
Abstract
The synthetic antimicrobial peptides (sAMPs) Pep19-2.5 and Pep19-4LF have been shown in vitro and in vivo to reduce the release of pro-inflammatory cytokines, leading to the suppression of inflammation and immunomodulation. We hypothesized that intervention with Pep19-2.5 and Pep19-4LF immediately after cardiac arrest and resuscitation (CA-CPR) might attenuate immediate systemic inflammation, survival, and long-term outcomes in a standardized mouse model of CA-CPR. Long-term outcomes up to 28 days were assessed between a control group (saline) and two peptide intervention groups. Primarily, survival as well as neurological and cognitive parameters were assessed. In addition, systemic inflammatory molecules and specific biomarkers were analyzed in plasma as well as in brain tissue. Treatment with sAMPs did not provide any short- or long-term benefits for either survival or neurological outcomes, and no significant benefit on inflammation in the CA-CPR animal model. While no difference was found in the plasma analysis of early cytokines between the intervention groups four hours after resuscitation, a significant increase in UCH-L1, a biomarker of neuronal damage and blood–brain barrier rupture, was measured in the Pep19-4LF-treated group. The theoretical benefit of both sAMPs tested here for the treatment of post-cardiac arrest syndrome could not be proven.
Collapse
Affiliation(s)
- Rika Bajorat
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany
- Correspondence:
| | - Lena Danckert
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Florian Ebert
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Theresa Bancken
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Stefan Bergt
- Department of Anesthesiology and Intensive Care Medicine, MEDICLIN Müritz-Klinikum, 17192 Waren, Germany
| | - Felix Klawitter
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Brigitte Vollmar
- Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Daniel A. Reuter
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Tobias Schürholz
- Department of Intensive and Intermediate Care, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Johannes Ehler
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
11
|
Gabriel C, Marks DC, Henschler R, Schallmoser K, Burnouf T, Koh MBC. Eye drops of human origin-Current status and future needs: Report on the workshop organized by the ISBT Working Party for Cellular Therapies. Vox Sang 2023; 118:301-309. [PMID: 36847186 DOI: 10.1111/vox.13413] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 03/01/2023]
Abstract
BACKGROUND AND OBJECTIVES Serum eye drops (SEDs) are used to treat ocular surface disease (OSD) and to promote ocular surface renewal. However, their use and production are not standardized, and several new forms of human eye drops have been developed. MATERIALS AND METHODS The International Society for Blood Transfusion Working Party (ISBT WP) for Cellular Therapies held a workshop to review the current types of eye drops of human origin (EDHO) status and provide guidance. RESULTS The ISBT WP for Cellular Therapies introduced the new terminology 'EDHO' to emphasize that these products are analogous to 'medical products of human origin'. This concept encompasses their source (serum, platelet lysate, and cord blood) and the increasingly diverse spectrum of clinical usage in ophthalmology and the need for traceability. The workshop identified the wide variability in EDHO manufacturing, lack of harmonized quality and production standards, distribution issues, reimbursement schemes and regulations. EDHO use and efficacy is established for the treatment of OSD, especially for those refractory to conventional treatments. CONCLUSION Production and distribution of single-donor donations are cumbersome and complex. The workshop participants agreed that allogeneic EDHO have advantages over autologous EDHO although more data on clinical efficacy and safety are needed. Allogeneic EDHOs enable more efficient production and, when pooled, can provide enhanced standardization for clinical consistency, provided optimal margin of virus safety is ensured. Newer products, including platelet-lysate- and cord-blood-derived EDHO, show promise and benefits over SED, but their safety and efficacy are yet to be fully established. This workshop highlighted the need for harmonization of EDHO standards and guidelines.
Collapse
Affiliation(s)
- Christian Gabriel
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria.,Ludwig Boltzmann Institute for Clinical and Experimental Traumatology, Vienna, Austria
| | - Denese C Marks
- Research and Development, The Australian Red Cross Lifeblood, Sydney, Australia.,Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Reinhard Henschler
- Institute of Transfusion Medicine, University Hospital and Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Katharina Schallmoser
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), Salzburg, Austria.,Department of Blood Group Serology and Transfusion Medicine, Universitätsklinikum, Salzburger Landeskliniken GesmbH (SALK), Salzburg, Austria
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Mickey B C Koh
- Institute for Infection and Immunity, St. George's University of London, Cranmer Terrace, Jenner Wing, London, UK
| |
Collapse
|
12
|
Huang C, Lou C, Zheng X, Pang L, Wang G, Zhu M, Dai X, Wang J, Tu M, Xu W, Chen Z, Gao H, Xu L. Plasma human neutrophil peptides as biomarkers of disease severity and mortality in patients with decompensated cirrhosis. Liver Int 2023; 43:1096-1106. [PMID: 36648384 DOI: 10.1111/liv.15520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/03/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023]
Abstract
BACKGROUND & AIMS Human neutrophil peptides (HNP)-1, -2 and -3 are the most abundant proteins in neutrophil azurophilic granules and are rapidly released via neutrophil degranulation upon activation. The aims of our study were to assess the role of HNP1-3 as biomarkers of disease severity in patients with decompensated cirrhosis and their value in predicting short-term mortality. METHODS In this study, 451 patients with acutely decompensated cirrhosis (AD) were enrolled at the two medical centres. Overall, 281 patients were enrolled as the training cohort from October 2015 to April 2019, and 170 patients were enrolled as the validation cohort from June 2020 to February 2021. Plasma HNP1-3 levels were measured using enzyme-linked immunosorbent assay (ELISA). RESULTS Plasma HNP1-3 increased stepwise with disease severity (compensated cirrhosis: 0.3 (0.2-0.4); AD without acute-on-chronic liver failure (ACLF): 1.9 (1.3-4.8); ACLF-1: 2.3 (1.8-6.1); ACLF-2: 5.6 (2.9-12.3); ACLF-3: 10.3 (5.7-17.2) ng/ml). From the multivariate Cox regression analysis, HNP1-3 emerged as independent predictors of mortality at 30 and 90 days. Similar results were observed in the subgroup analysis. On ROC analysis, plasma HNP1-3 showed better predictive accuracy for 30- and 90-day mortality (area under the receiver operating characteristic (AUROC) of 0.850 and 0.885, respectively) than the neutrophil-to-lymphocyte ratio (NLR) and similar accuracy as end-stage liver disease (MELD: 0.881 and 0.874) and chronic liver failure-sequential organ failure (CLIF-SOFA: 0.887 and 0.878). CONCLUSIONS Plasma HNP1-3 levels were closely associated with disease severity and might be used to identify patients with AD at high risk of short-term mortality.
Collapse
Affiliation(s)
- Chunhong Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Congcong Lou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoping Zheng
- Department of Pathology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| | - Lantian Pang
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Gang Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengfei Zhu
- Department of Infectious Diseases, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| | - Xiahong Dai
- Department of Infectious Diseases, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| | - Jie Wang
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingmin Tu
- Department of Clinical Laboratory, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| | - Wei Xu
- Department of Clinical Laboratory, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hainv Gao
- Department of Infectious Diseases, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| | - Lichen Xu
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
13
|
Soni M, Handa M, Singh KK, Shukla R. Recent nanoengineered diagnostic and therapeutic advancements in management of Sepsis. J Control Release 2022; 352:931-945. [PMID: 36273527 PMCID: PMC9665001 DOI: 10.1016/j.jconrel.2022.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022]
Abstract
COVID-19 acquired symptoms have affected the worldwide population and increased the load of Intensive care unit (ICU) patient admissions. A large number of patients admitted to ICU end with a deadly fate of mortality. A high mortality rate of patients was reported with hospital-acquired septic shock that leads to multiple organ failures and ultimately ends with death. The patients who overcome this septic shock suffer from morbidity that also affects their caretakers. To overcome these situations, scientists are exploring progressive theragnostic techniques with advanced techniques based on biosensors, biomarkers, biozymes, vesicles, and others. These advanced techniques pave the novel way for early detection of sepsis-associated symptoms and timely treatment with appropriate antibiotics and immunomodulators and prevent the undue effect on other parts of the body. There are other techniques like externally modulated electric-based devices working on the principle of piezoelectric mechanism that not only sense the endotoxin levels but also target them with a loaded antibiotic to neutralize the onset of inflammatory response. Recently researchers have developed a lipopolysaccharide (LPS) neutralizing cartridge that not only senses the LPS but also appropriately neutralizes with dual mechanistic insights of antibiotic and anti-inflammatory effects. This review will highlight recent developments in the new nanotechnology-based approaches for the diagnosis and therapeutics of sepsis that is responsible for the high number of deaths of patients suffering from this critical disease.
Collapse
Affiliation(s)
- Mukesh Soni
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, U.P., India
| | - Mayank Handa
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, U.P., India
| | - Kamalinder K. Singh
- School of Pharmacy and Biomedical Sciences, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK,Correspondence to: Prof. Kamalinder K. Singh, School of Pharmacy and Biomedical Sciences, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, United Kingdom
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, U.P., India,School of Pharmacy and Biomedical Sciences, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK,Correspondence to: Dr. Rahul Shukla (M. Pharm. PhD), National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow 226002, UP, India
| |
Collapse
|
14
|
Louvrier C, Awad F, Cosnes A, El Khouri E, Assrawi E, Daskalopoulou A, Copin B, Bocquet H, Chantot Bastaraud S, Arenas Garcia A, Dastot Le Moal F, De La Grange P, Duquesnoy P, Guerrera CI, Piterboth W, Ortonne N, Chosidow O, Karabina SA, Amselem S, Giurgea I. RNF213-associated urticarial lesions with hypercytokinemia. J Allergy Clin Immunol 2022; 150:1545-1555. [PMID: 35780935 DOI: 10.1016/j.jaci.2022.06.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/24/2022] [Accepted: 06/02/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Urticarial lesions are observed in both cutaneous and systemic disorders. Familial forms of urticarial syndromes are rare and can be encountered in systemic autoinflammatory diseases. OBJECTIVE We sought to investigate a large family with dominantly inherited chronic urticarial lesions associated with hypercytokinemia. METHODS We performed a genetic linkage analysis in 14 patients from a 5-generation family, as well as whole-exome sequencing, cytokine profiling, and transcriptomic analyses on samples from 2 patients. The identified candidate protein was studied after in vitro expression of the corresponding normal and mutated recombinant proteins. An unsupervised proteomic approach was used to unveil the associated protein network. RESULTS The disease phenotype of the most affected family members is characterized by chronic urticarial flares associated with extremely high plasma levels of proinflammatory (IL-1β, IL-6, and TNF-α) and anti-inflammatory (IL-10 and IL-1 receptor antagonist [IL-1RA]) cytokines, with no secondary organ dysfunction, no susceptibility to infections, no fever, and normal C-reactive protein levels. Monocyte transcriptomic analyses identified an immunotolerant profile in the most affected patient. The affected family members carried a loss-of-function mutation in RNF213 that encodes mysterin, a protein with a poorly known physiologic role. We identified the deubiquitinase CYLD, a major regulator of inflammation, as an RNF213 partner and showed that CYLD expression is inhibited by wild-type but not mutant RNF213. CONCLUSION We identified a new entity characterized by chronic urticarial lesions associated with a clinically blunted hypercytokinemia. This disease, which is due to loss of function of RNF213, reveals mysterin's key role in the complex molecular network of innate immunity.
Collapse
Affiliation(s)
- Camille Louvrier
- Sorbonne Université, Inserm, Childhood Genetic Disorders, Hôpital Armand-Trousseau, Paris, France; Département de Génétique Médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Armand-Trousseau, Paris, France
| | - Fawaz Awad
- Sorbonne Université, Inserm, Childhood Genetic Disorders, Hôpital Armand-Trousseau, Paris, France
| | - Anne Cosnes
- Faculté de Santé de Créteil and Service de Dermatologie, APHP, Hôpital Henri-Mondor, Université Paris-Est, Créteil, France
| | - Elma El Khouri
- Sorbonne Université, Inserm, Childhood Genetic Disorders, Hôpital Armand-Trousseau, Paris, France
| | - Eman Assrawi
- Sorbonne Université, Inserm, Childhood Genetic Disorders, Hôpital Armand-Trousseau, Paris, France
| | - Aphrodite Daskalopoulou
- Sorbonne Université, Inserm, Childhood Genetic Disorders, Hôpital Armand-Trousseau, Paris, France
| | - Bruno Copin
- Sorbonne Université, Inserm, Childhood Genetic Disorders, Hôpital Armand-Trousseau, Paris, France; Département de Génétique Médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Armand-Trousseau, Paris, France
| | - Hélène Bocquet
- Faculté de Santé de Créteil and Service de Dermatologie, APHP, Hôpital Henri-Mondor, Université Paris-Est, Créteil, France
| | - Sandra Chantot Bastaraud
- Département de Génétique Médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Armand-Trousseau, Paris, France
| | - Angela Arenas Garcia
- Sorbonne Université, Inserm, Childhood Genetic Disorders, Hôpital Armand-Trousseau, Paris, France
| | - Florence Dastot Le Moal
- Département de Génétique Médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Armand-Trousseau, Paris, France
| | | | - Philippe Duquesnoy
- Sorbonne Université, Inserm, Childhood Genetic Disorders, Hôpital Armand-Trousseau, Paris, France
| | - Chiara I Guerrera
- Plateforme protéomique Necker, Université de Paris, Structure Fédérative de Recherche Necker, Inserm US24/CNRS UMS3633, Paris, France
| | - William Piterboth
- Département de Génétique Médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Armand-Trousseau, Paris, France
| | - Nicolas Ortonne
- Département d'Anatomo-Pathologie, APHP, Hôpital Henri-Mondor, Créteil, France
| | - Olivier Chosidow
- Faculté de Santé de Créteil and Service de Dermatologie, APHP, Hôpital Henri-Mondor, Université Paris-Est, Créteil, France; Research Group Dynamic, EA7380, Faculté de Santé de Créteil, Ecole Nationale Vétérinaire d'Alfort, USC ANSES, Université Paris-Est Créteil, Créteil, France
| | - Sonia A Karabina
- Sorbonne Université, Inserm, Childhood Genetic Disorders, Hôpital Armand-Trousseau, Paris, France
| | - Serge Amselem
- Sorbonne Université, Inserm, Childhood Genetic Disorders, Hôpital Armand-Trousseau, Paris, France; Département de Génétique Médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Armand-Trousseau, Paris, France.
| | - Irina Giurgea
- Sorbonne Université, Inserm, Childhood Genetic Disorders, Hôpital Armand-Trousseau, Paris, France; Département de Génétique Médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Armand-Trousseau, Paris, France.
| |
Collapse
|
15
|
Zhang B, Yin S, Guo C, Gao Z, Li T, Lee W, Shen J, Yang X. Fragments of bombinakinin M exist in lipopolysaccharide-stimulated skin secretions of Bombina maxima and show lipopolysaccharide-neutralizing activity. J Immunol Methods 2022; 509:113343. [PMID: 36029800 DOI: 10.1016/j.jim.2022.113343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022]
Abstract
Lipopolysaccharide (LPS) is a major pathogen-associated pattern molecule that can initiate lethal sepsis. Bioactive peptides in amphibian skin secretions, especially antimicrobial peptides, are essential components of the host immune system and help fight the microbial invasion. In this study, two peptides: peptide 1 (KINRKGPRPPG) and peptide 2 (INRKGPRPPG) were isolated, from skin secretions of the Chinese red belly frog (Bombina maxima). After stimulation with LPS, peptide 1 showed direct LPS-binding activity, low cytotoxicity, immunoregulatory functions in vitro, and neutralizing LPS effects in animal models. Thus, natural peptide 1 exhibits potential as an ideal candidate against LPS.
Collapse
Affiliation(s)
- Baiyu Zhang
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Saige Yin
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Caifen Guo
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Zhenhua Gao
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Tonghai Li
- Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Wenhui Lee
- Key Laboratory of Animal Models and Human Disease Mechanisms, Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| | - Jihong Shen
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China.
| | - Xinwang Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650500, China.
| |
Collapse
|
16
|
Human Neutrophil Defensins Disrupt Liver Interendothelial Junctions and Aggravate Sepsis. Mediators Inflamm 2022; 2022:7659282. [PMID: 35935811 PMCID: PMC9355784 DOI: 10.1155/2022/7659282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 06/30/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Human neutrophil peptides 1-3 (HNP1-3), also known as human α-defensins, are the most abundant neutrophil granule proteins. The genes that encode HNP1-3, DEFA1/DEFA3, exhibit extensive copy number variations, which correlate well with their protein levels. Human and mouse studies have shown that increased copy numbers of DEFA1/DEFA3 worsen sepsis outcomes. Additionally, high concentrations of HNP1-3 in body fluids have been reported in patients with sepsis. However, direct evidence for the pathogenic role of HNP1-3 proteins during sepsis progression is lacking. In current study, sepsis was induced by means of cecal puncture and ligation. Various doses of HNP-1 (low dose with 0.5 mg/kg body weight and high dose with 10 mg/kg body weight) or phosphate buffer saline were intraperitoneally administered to mice at six hours after sepsis onset. Survival rate was monitored, and vascular permeability, endothelial cell pyroptosis, and immunofluorescence of endothelial adherens junction protein vascular endothelial-cadherin were evaluated. The administration of a high dose of HNP-1 after sepsis onset led to increased mortality, more severe liver injury, and increased vascular permeability in the liver and mesentery. The injection of high dose of HNP-1 did not directly induce liver endothelial cell death but destroyed interendothelial junctions in the liver. Moreover, genetic deficiency of nucleotide-binding oligomerization domain-like receptor protein-3 or caspase-1 abrogated the high mortality and disrupted liver interendothelial junctions caused by high dose of HNP-1 during sepsis. This study directly demonstrates that neutrophil defensins play a key role in regulating endothelial stability during sepsis development.
Collapse
|
17
|
Lee CG, Park C, Hwang S, Hong JE, Jo M, Eom M, Lee Y, Rhee KJ. Pulsed Electromagnetic Field (PEMF) Treatment Reduces Lipopolysaccharide-Induced Septic Shock in Mice. Int J Mol Sci 2022; 23:ijms23105661. [PMID: 35628471 PMCID: PMC9147061 DOI: 10.3390/ijms23105661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 12/04/2022] Open
Abstract
Despite advances in medicine, mortality due to sepsis has not decreased. Pulsed electromagnetic field (PEMF) therapy is emerging as an alternative treatment in many inflammation-related diseases. However, there are few studies on the application of PEMF therapy to sepsis. In the current study, we examined the effect of PEMF therapy on a mouse model of lipopolysaccharide (LPS)-induced septic shock. Mice injected with LPS and treated with PEMF showed higher survival rates compared with the LPS group. The increased survival was correlated with decreased levels of pro-inflammatory cytokine mRNA expression and lower serum nitric oxide levels and nitric oxide synthase 2 mRNA expression in the liver compared with the LPS group. In the PEMF + LPS group, there was less organ damage in the liver, lungs, spleen, and kidneys compared to the LPS group. To identify potential gene targets of PEMF treatment, microarray analysis was performed, and the results showed that 136 genes were up-regulated, and 267 genes were down-regulated in the PEMF + LPS group compared to the LPS group. These results suggest that PEMF treatment can dramatically decrease septic shock through the reduction of pro-inflammatory cytokine gene expression. In a clinical setting, PEMF may provide a beneficial effect for patients with bacteria-induced sepsis and reduce septic shock-induced mortality.
Collapse
Affiliation(s)
- Chang-Gun Lee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea; (C.-G.L.); (C.P.); (S.H.); (J.-E.H.); (M.J.)
- Department of Medical Genetics, School of Medicine, Ajou University, Suwon 16499, Korea
| | - Chanoh Park
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea; (C.-G.L.); (C.P.); (S.H.); (J.-E.H.); (M.J.)
| | - Soonjae Hwang
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea; (C.-G.L.); (C.P.); (S.H.); (J.-E.H.); (M.J.)
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, GAIST, College of Medicine, Gachon University, Incheon 21999, Korea
| | - Ju-Eun Hong
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea; (C.-G.L.); (C.P.); (S.H.); (J.-E.H.); (M.J.)
| | - Minjeong Jo
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea; (C.-G.L.); (C.P.); (S.H.); (J.-E.H.); (M.J.)
| | - Minseob Eom
- Department of Pathology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
| | - Yongheum Lee
- Department of Biomedical Engineering, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea
- Correspondence: (Y.L.); (K.-J.R.); Tel.: +82-33-760-2863 (Y.L.); +82-33-760-2445 (K.-J.R.); Fax: +82-33-760-2561 (Y.L.); +82-33-760-2195 (K.-J.R.)
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea; (C.-G.L.); (C.P.); (S.H.); (J.-E.H.); (M.J.)
- Correspondence: (Y.L.); (K.-J.R.); Tel.: +82-33-760-2863 (Y.L.); +82-33-760-2445 (K.-J.R.); Fax: +82-33-760-2561 (Y.L.); +82-33-760-2195 (K.-J.R.)
| |
Collapse
|
18
|
Antimicrobial and Immunomodulatory Effects of Selected Chemokine and Antimicrobial Peptide on Cytokine Profile during Salmonella Typhimurium Infection in Mouse. Antibiotics (Basel) 2022; 11:antibiotics11050607. [PMID: 35625251 PMCID: PMC9137564 DOI: 10.3390/antibiotics11050607] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/05/2023] Open
Abstract
The antimicrobial and immunomodulatory capacities of the peptide Css54 and the chemokine MCP-1 were tested. The first, a peptide isolated from the venom of the scorpion Centruroides suffusus suffusus was synthesized chemically. In contrast, the second is a monocyte chemoattractant expressed as a recombinant protein in our lab. It was observed in vitro that Css54 inhibited the growth of Salmonella enterica serovar Typhimurium (6.2 µg/mL). At high concentrations, it was toxic to macrophages (25 µg/mL), activated macrophage phagocytosis (1.5 µg/mL), and bound Salmonella LPS (3 µg/mL). On the other hand, the recombinant MCP-1 neither inhibited the growth of Salmonella Typhimurium nor was it toxic to macrophages (up to 25 µg/mL), nor activated macrophage phagocytosis or bound Salmonella LPS (up to 3 µg/mL). Although it was observed in vivo in mice Balb/C that both Css54 and MCP-1 did not resolve the intraperitoneal infection by S. Typhimurium, Css54 decreased the expression of IL-6 and increased IL-10, IL-12p70, and TNF-α levels; meanwhile, MCP-1 decreased the expression of IFN-γ and increased IL-12p70 and TNF-α. It was also observed that the combination of both molecules Css54 and MCP-1 increased the expression of IL-10 and TNF-α.
Collapse
|
19
|
Immunomodulatory and Allergenic Properties of Antimicrobial Peptides. Int J Mol Sci 2022; 23:ijms23052499. [PMID: 35269641 PMCID: PMC8910669 DOI: 10.3390/ijms23052499] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
With the growing problem of the emergence of antibiotic-resistant bacteria, the search for alternative ways to combat bacterial infections is extremely urgent. While analyzing the effect of antimicrobial peptides (AMPs) on immunocompetent cells, their effect on all parts of the immune system, and on humoral and cellular immunity, is revealed. AMPs have direct effects on neutrophils, monocytes, dendritic cells, T-lymphocytes, and mast cells, participating in innate immunity. They act on B-lymphocytes indirectly, enhancing the induction of antigen-specific immunity, which ultimately leads to the activation of adaptive immunity. The adjuvant activity of AMPs in relation to bacterial and viral antigens was the reason for their inclusion in vaccines and made it possible to formulate the concept of a “defensin vaccine” as an innovative basis for constructing vaccines. The immunomodulatory function of AMPs involves their influence on cells in the nearest microenvironment, recruitment and activation of other cells, supporting the response to pathogenic microorganisms and completing the inflammatory process, thus exhibiting a systemic effect. For the successful use of AMPs in medical practice, it is necessary to study their immunomodulatory activity in detail, taking into account their pleiotropy. The degree of maturity of the immune system and microenvironment can contribute to the prevention of complications and increase the effectiveness of therapy, since AMPs can suppress inflammation in some circumstances, but aggravate the response and damage of organism in others. It should also be taken into account that the real functions of one or another AMP depend on the types of total regulatory effects on the target cell, and not only on properties of an individual peptide. A wide spectrum of biological activity, including direct effects on pathogens, inactivation of bacterial toxins and influence on immunocompetent cells, has attracted the attention of researchers, however, the cytostatic activity of AMPs against normal cells, as well as their allergenic properties and low stability to host proteases, are serious limitations for the medical use of AMPs. In this connection, the tasks of searching for compounds that selectively affect the target and development of an appropriate method of application become critically important. The scope of this review is to summarize the current concepts and newest advances in research of the immunomodulatory activity of natural and synthetic AMPs, and to examine the prospects and limitations of their medical use.
Collapse
|
20
|
Zhu Y, Hao W, Wang X, Ouyang J, Deng X, Yu H, Wang Y. Antimicrobial peptides, conventional antibiotics, and their synergistic utility for the treatment of drug-resistant infections. Med Res Rev 2022; 42:1377-1422. [PMID: 34984699 DOI: 10.1002/med.21879] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/09/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022]
Abstract
Antimicrobial peptides (AMPs), also known as host defense peptides (HDPs), are important effector immune defense molecules in multicellular organisms. AMPs exert their antimicrobial activities through several mechanisms; thus far, induction of drug resistance through AMPs has been regarded as unlikely. Therefore, they have great potential as new generation antimicrobial agents. To date, more than 30 AMP-related drugs are in the clinical trial phase. In recent years, studies show that some AMPs and conventional antibiotics have synergistic effects. The combined use of AMPs and antibiotics can kill drug-resistant pathogens, prevent drug resistance, and significantly improve the therapeutic effects of antibiotics. In this review, we discuss the progress in synergistic studies on AMPs and conventional antibiotics. An overview of the current understanding of the functional scope of AMPs, ongoing clinical trials, and challenges in the development processes are also presented.
Collapse
Affiliation(s)
- Yiyun Zhu
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Weijing Hao
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xia Wang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Jianhong Ouyang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xinyi Deng
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Haining Yu
- Department of Bioscience and Biotechnology, Dalian University of Technology, Dalian, Liaoning, China
| | - Yipeng Wang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
21
|
Pereshein AV, Kuznetsova SV, Shevantaeva ON. On the Nonspecific Resistance in Burn Injury: Pathophysiological Aspects (Review). Sovrem Tekhnologii Med 2021; 12:84-93. [PMID: 34795984 PMCID: PMC8596251 DOI: 10.17691/stm2020.12.3.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Indexed: 11/14/2022] Open
Abstract
An analysis of nonspecific resistance in burn patients is conducted. The role of subpopulations of neutrophils and monocytes/ macrophages in severe burn injury is discussed. The significance of blood cells for the burn-induced immune dysfunction, susceptibility to sepsis and multiple organ failure is underscored. The involvement of secondary complications in the development of morbidity and mortality in patients with burn injury is shown. New approaches to identifying individuals with a risk of adverse outcome are considered.
Collapse
Affiliation(s)
- A V Pereshein
- Assistant, Department of Pathological Physiology; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - S V Kuznetsova
- Associate Professor, Department of Pathological Physiology; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - O N Shevantaeva
- Professor, Department of Pathological Physiology Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| |
Collapse
|
22
|
Srivastava M, Chandra A, R R, Nigam J, Rajan P, Parmar D, Srivastava RN, Gupta V. Expression of Antimicrobial Peptides and Cytokines in Human Omentum Following Abdominal Surgery. Cureus 2021; 13:e17477. [PMID: 34589365 PMCID: PMC8464651 DOI: 10.7759/cureus.17477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2021] [Indexed: 12/02/2022] Open
Abstract
Introduction Omentum can secrete out biological agents like different growth factors, cytokines, and antimicrobial peptides. The aim of our study was to determine the expression of antimicrobial peptides and cytokines in human omentum tissue and its response to intra-abdominal infection. Methodology Omentum tissue was obtained from 60 patients: control (n=20) and cases (n=40). mRNA expression of antimicrobial peptides (LL-37, HBD-1, HBD-2, HNP1-3) and cytokines (TNF- α, IL-8, IL-10, IL1β) was evaluated using Real-Time PCR. Protein quantification was done by Immunoblotting and ELISA. Results Significantly higher expression of antimicrobial peptides (LL-37, HBD-1, HBD-2, HNP1-3) and cytokines (TNF- α, IL-8, IL-10, IL1β) was observed in cases as compared to control at both the transcriptional and translational level (p<0.0001). Conclusion Omentum governs a population of antimicrobial peptides with potent immunologic functions. The expression of antimicrobial peptides and cytokines is inducible and increases with the severity of infection. Omentum is thus an immunologically active and adaptable organ but its complete regulatory mechanism is still elusive.
Collapse
Affiliation(s)
- Meenu Srivastava
- Surgical Gastroenterology, King George's Medical University, Lucknow, IND
| | - Abhijit Chandra
- Surgical Gastroenterology, King George's Medical University, Lucknow, IND
| | - Rahul R
- Surgical Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, IND
| | - Jaya Nigam
- Surgical Gastroenterology, King George's Medical University, Lucknow, IND
| | - Pritheesh Rajan
- Surgical Gastroenterology, King George's Medical University, Lucknow, IND
| | - Devendra Parmar
- Developmental Toxicology, Indian Institute of Toxicology Research, Lucknow, IND
| | | | - Vivek Gupta
- Surgical Gastroenterology, King George's Medical University, Lucknow, IND
| |
Collapse
|
23
|
Bosch M, Sweet MJ, Parton RG, Pol A. Lipid droplets and the host-pathogen dynamic: FATal attraction? J Cell Biol 2021; 220:e202104005. [PMID: 34165498 PMCID: PMC8240858 DOI: 10.1083/jcb.202104005] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
In the ongoing conflict between eukaryotic cells and pathogens, lipid droplets (LDs) emerge as a choke point in the battle for nutrients. While many pathogens seek the lipids stored in LDs to fuel an expensive lifestyle, innate immunity rewires lipid metabolism and weaponizes LDs to defend cells and animals. Viruses, bacteria, and parasites directly and remotely manipulate LDs to obtain substrates for metabolic energy, replication compartments, assembly platforms, membrane blocks, and tools for host colonization and/or evasion such as anti-inflammatory mediators, lipoviroparticles, and even exosomes. Host LDs counterattack such advances by synthesizing bioactive lipids and toxic nucleotides, organizing immune signaling platforms, and recruiting a plethora of antimicrobial proteins to provide a front-line defense against the invader. Here, we review the current state of this conflict. We will discuss why, when, and how LDs efficiently coordinate and precisely execute a plethora of immune defenses. In the age of antimicrobial resistance and viral pandemics, understanding innate immune strategies developed by eukaryotic cells to fight and defeat dangerous microorganisms may inform future anti-infective strategies.
Collapse
Affiliation(s)
- Marta Bosch
- Lipid Trafficking and Disease Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Matthew J. Sweet
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia
| | - Robert G. Parton
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, Australia
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| |
Collapse
|
24
|
Ilyas H, van der Plas MJA, Agnoletti M, Kumar S, Mandal AK, Atreya HS, Bhunia A, Malmsten M. Effect of PEGylation on Host Defense Peptide Complexation with Bacterial Lipopolysaccharide. Bioconjug Chem 2021; 32:1729-1741. [PMID: 34282895 DOI: 10.1021/acs.bioconjchem.1c00259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Conjugation with poly(ethylene glycol) ("PEGylation") is a widely used approach for improving the therapeutic propensities of peptide and protein drugs through prolonging bloodstream circulation, reducing toxicity and immunogenicity, and improving proteolytic stability. In the present study, we investigate how PEGylation affects the interaction of host defense peptides (HDPs) with bacterial lipopolysaccharide (LPS) as well as HDP suppression of LPS-induced cell activation. In particular, we investigate the effects of PEGylation site for KYE28 (KYEITTIHNLFRKLTHRLFRRNFGYTLR), a peptide displaying potent anti-inflammatory effects, primarily provided by its N-terminal part. PEGylation was performed either in the N-terminus, the C-terminus, or in both termini, keeping the total number of ethylene groups (n = 48) constant. Ellipsometry showed KYE28 to exhibit pronounced affinity to both LPS and its hydrophobic lipid A moiety. The PEGylated peptide variants displayed lower, but comparable, affinity for both LPS and lipid A, irrespective of the PEGylation site. Furthermore, both KYE28 and its PEGylated variants triggered LPS aggregate disruption. To investigate the peptide structure in such LPS complexes, a battery of nuclear magnetic resonance (NMR) methods was employed. From this, it was found that KYE28 formed a well-folded structure after LPS binding, stabilized by hydrophobic domains involving aromatic amino acids as well as by electrostatic interactions. In contrast, the PEGylated peptide variants displayed a less well-defined secondary structure, suggesting weaker LPS interactions in line with the ellipsometry findings. Nevertheless, the N-terminal part of KYE28 retained helix formation after PEGylation, irrespective of the conjugation site. For THP1-Xblue-CD14 reporter cells, KYE28 displayed potent suppression of LPS activation at simultaneously low cell toxicity. Interestingly, the PEGylated KYE28 variants displayed similar or improved suppression of LPS-induced cell activation, implying the underlying key role of the largely retained helical structure close to the N-terminus, irrespective of PEGylation site. Taken together, the results show that PEGylation of HDPs can be done insensitively to the conjugation site without losing anti-inflammatory effects, even for peptides inducing such effects through one of its termini.
Collapse
Affiliation(s)
- Humaira Ilyas
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700054, India
| | - Mariena J A van der Plas
- Department of Pharmacy, University of Copenhagen, DK-2100 Copenhagen, Denmark.,Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-221 84 Lund, Sweden
| | - Monica Agnoletti
- Department of Pharmacy, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Sourav Kumar
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700054, India
| | - Atin Kumar Mandal
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700054, India
| | - Hanudatta S Atreya
- NMR Research Center, Indian Institute of Science, Bangalore 560012, India
| | - Anirban Bhunia
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700054, India
| | - Martin Malmsten
- Department of Pharmacy, University of Copenhagen, DK-2100 Copenhagen, Denmark.,Department of Physical Chemistry 1, University of Lund, SE-22100 Lund, Sweden
| |
Collapse
|
25
|
Wang C, Li Q, Tang C, Zhao X, He Q, Tang X, Ren J. Characterization of the blood and neutrophil-specific microbiomes and exploration of potential bacterial biomarkers for sepsis in surgical patients. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:1343-1357. [PMID: 34288545 PMCID: PMC8589375 DOI: 10.1002/iid3.483] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/12/2021] [Accepted: 06/21/2021] [Indexed: 12/29/2022]
Abstract
Introduction Recent studies have demonstrated the presence of a circulating microbiome in the blood of healthy subjects and chronic inflammatory patients. However, our knowledge regarding the blood microbiome and its potential roles in surgical patients remains very limited. The objective of this study was to determine the blood microbial landscape in surgical patients and to explore its potential associations with postoperative sepsis. Materials and Methods 2825 patients who underwent surgical treatments were screened for enrollment and 204 cases were recruited in this study. The patients were sub‐grouped into noninfected, infected, sepsis, and septic shock according to postoperative clinical manifestations. A total of 222 blood samples were obtained for neutrophil isolation, DNA extraction and high‐throughput sequencing, quantitative proteomics analysis, and flow cytometric analyses. Results Blood and neutrophils in surgical patients and healthy controls contained highly diverse microbiomes, mainly comprising Proteobacteria, Actinobacteria, Firmicutes and Bacteroidetes. The majority (80.7%–91.5%) of the microbiomes were composed of gut‐associated bacteria. The microbiomes in septic patients were significantly distinct from those of healthy controls, and marked differences in microbiome composition were observed between sepsis and septic shock groups. Several specific bacterial genera, including Flavobacterium, Agrococcus, Polynucleobacter, and Acidovorax, could distinguish patients with septic shock from those with sepsis, with higher area under curve values. Moreover, Agrococcus, Polynucleobacter, and Acidovorax were positively associated with the sequential (sepsis‐related) organ failure assessment scores and/or acute physiology and chronic health examination scores in septic shock patients. The proteins involved in bactericidal activities of neutrophils were downregulated in septic patients. Conclusions We present evidence identifying significant changes of blood and neutrophil‐specific microbiomes across various stages of sepsis, which might be associated with the progression of sepsis after surgical treatments. Several certain bacterial genera in blood microbiome could have potential as microbial markers for early detection of sepsis.
Collapse
Affiliation(s)
- Chenyang Wang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qiurong Li
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chun Tang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaofan Zhao
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qin He
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xingming Tang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
26
|
Huang F, Huang S. Active vitamin D3 attenuates the severity of Salmonella colitis in mice by orchestrating innate immunity. Immun Inflamm Dis 2021; 9:481-491. [PMID: 33559391 PMCID: PMC8127544 DOI: 10.1002/iid3.408] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/02/2021] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Salmonella spp. pose major public health problems worldwide. A better understanding of the pathogenesis of these foodborne pathogens is a prerequisite for the design of improved intervention strategies that could reduce the use of antimicrobial agents and drug-resistant Salmonellosis. Accumulating evidence indicates that vitamin D is involved in regulating innate immunity, and may, therefore, play a key role in human responses to infection. Studies have suggested 1,25-dihydroxyvitamin D3 (1,25D3), the active form of vitamin D, effectively ameliorates colitis. These findings have broad implications for the use of vitamin D compounds in colitis. This study investigated the effect of active vitamin D3 on the severity of Salmonella colitis. METHODS A Salmonella colitis model was established with 6-8-week-old male C57BL/6 mice: Streptomycin-pretreated C57BL/6 mice were infected orally with Salmonella enterica serova Typhimurium wild-type strain SL1344 for 48 h. The mice were randomly assigned to control, model, and 1,25(OH)2 D3 -treated groups. After the experiment, the mice were sacrificed, and intestinal, spleen, and liver tissue samples were removed to analyze bacterial colonization, western blot for protein levels, and real-time-polymer chain reaction for messenger RNA (mRNA) expression. RESULTS We observed that 1,25D3 reduced the severity of Salmonella colitis in C57BL/6 mice by reducing cecal mIL-1beta, mIL-6, mTNF-alpha, and mIL-8 mRNA expressions, bacterial colonization (CFU/mg tissue) in the liver and spleen, but increased the human β-defensin-2 mRNA and autophagy protein expression, compared to those of the SL1344 infection only. CONCLUSIONS Our results document that active vitamin D3 reduced Salmonella colitis by decreasing inflammation, and bacterial translocation via induction of killing and autophagic clearance of pathogenic organisms.
Collapse
Affiliation(s)
- Fu‐Chen Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial HospitalChang Gung University College of MedicineKaohsiungTaiwan
| | - Shun‐Chen Huang
- Department of PathologyKaohsiung Chang Gung Memorial HospitalKaohsiungTaiwan
| |
Collapse
|
27
|
Kadir NA, Hatta M, Arif M, Bahrun U, Parwati I, Mubin RH, Minhajat R, Hamid F, Widaningsih Y, Usman AN, Handayani I, Tiro S, Kurniawan LB. Human neutrophil peptide (HNP) in deteriorate sepsis patients. ENFERMERIA CLINICA 2021. [PMID: 32204264 DOI: 10.1016/j.enfcli.2019.07.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE The objective of this study was analyzed HNP1 and HNP3 level of deteriorating sepsis patients. METHODS It was a cross-sectional study that observed HNP and sepsis level, samples size were 40 patients taken by consecutive random sampling. Analysis of HNP serum level use enzyme-linked immunosorbent assay (ELISA), data analyzed used independent T-test and presented in tables with mean, standard deviation, mean difference and probability value. RESULTS Results show that when patients become deteriorate or shock sepsis, HNP1 levels increase dramatically, which is 356.2pg/ml higher than sepsis patients and statistically significant (p=0.016, p≤0.05). It is similar to HNP3 level, which is 2.04ng/ml higher than with sepsis patients and statistically significant (p=0.021, p≤0.05). CONCLUSION This study reveals that HNP1 and HNP3 level increases dramatically in deteriorate sepsis patients. Level of HNP1 and HNP3 of sepsis patient is higher than nonsepsis patient, shock sepsis patient is higher than sepsis patient and both HNP1 and HNP3 level is higher in die patient compare to alive patient.
Collapse
Affiliation(s)
- Nursin Abdul Kadir
- Clinical Pathology Department, Faculty of Medicine, Hasanuddin University, Indonesia
| | - Mochammad Hatta
- Microbiology Department, Faculty of Medicine, Hasanuddin University, Indonesia
| | - Mansyur Arif
- Clinical Pathology Department, Faculty of Medicine, Hasanuddin University, Indonesia
| | - Uleng Bahrun
- Clinical Pathology Department, Faculty of Medicine, Hasanuddin University, Indonesia.
| | - Ida Parwati
- Clinical Pathology Department, Faculty of Medicine, Padjajaran University, Indonesia
| | - Risna Halim Mubin
- Tropical Infection Division of Internal Medicine Department, Faculty of Medicine, Hasanuddin University, Indonesia
| | - Rahmawaty Minhajat
- Internal Medicine Department, Faculty of Medicine, Hasanuddin University, Indonesia
| | - Firdaus Hamid
- Microbiology Department, Faculty of Medicine, Hasanuddin University, Indonesia
| | - Yuyun Widaningsih
- Clinical Pathology Department, Faculty of Medicine, Hasanuddin University, Indonesia
| | | | - Irda Handayani
- Clinical Pathology Department, Faculty of Medicine, Hasanuddin University, Indonesia
| | - Steven Tiro
- Clinical Pathology Department, Faculty of Medicine, Hasanuddin University, Indonesia
| | - Liong Boy Kurniawan
- Clinical Pathology Department, Faculty of Medicine, Hasanuddin University, Indonesia
| |
Collapse
|
28
|
Song M, Liu G, Liu Y, Cheng Z, Lin H, Liu J, Wu Z, Xue J, Hong W, Huang M, Li J, Xu P. Using porphyrins as albumin-binding molecules to enhance antitumor efficacies and reduce systemic toxicities of antimicrobial peptides. Eur J Med Chem 2021; 217:113382. [PMID: 33751980 DOI: 10.1016/j.ejmech.2021.113382] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/24/2021] [Accepted: 03/08/2021] [Indexed: 11/20/2022]
Abstract
Antimicrobial peptides (AMPs) are originally developed for anti-infective treatments. Because of their membrane-lytic property, AMPs have been considered as candidates of antitumor agents for a long time. However, their antitumor applications are mainly hampered by fast renal clearance and high systemic toxicities. This study proposes a strategy aiming at addressing these two issues by conjugating AMPs with porphyrins, which bind to albumin increasing AMPs' resistance against renal clearance and thus enhancing their antitumor efficacies. Porphyrins' photodynamic properties can further augment AMPs' antitumor effects. In addition, circulating with albumin ameliorates AMPs' systemic toxicities, i.e. hemolysis and organ dysfunctions. As an example, we conjugated an AMP, K6L9, with pyropheophorbide-a (PPA) leading to a conjugate of PPA-K6L9. PPA-K6L9 bound to albumin with a KD value at the sub-micromolar range. Combining computational and experimental approaches, we characterized the molecular interaction of PPA-K6L9 with albumin. Furthermore, PPA-conjugation promoted K6L9' antitumor effects by prolonging its in vivo retention time, and reduced the hemolysis and hepatic injuries, which confirmed our design strategy.
Collapse
Affiliation(s)
- Meiru Song
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China; National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, Fuzhou University, Fujian, China
| | - Ge Liu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China; National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, Fuzhou University, Fujian, China
| | - Yichang Liu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China; National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, Fuzhou University, Fujian, China
| | - Ziwei Cheng
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China; National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, Fuzhou University, Fujian, China
| | - Haili Lin
- Department of Pharmacy, The Peoples Hospital of Fujian Province, Fuzhou, China
| | - Jianyong Liu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China; National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, Fuzhou University, Fujian, China
| | - Zaisheng Wu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Jinping Xue
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China; National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, Fuzhou University, Fujian, China
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, A∗STAR (Agency of Science, Technology and Research), 117608, Singapore
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China; National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, Fuzhou University, Fujian, China; Fujian Key Laboratory of Marine Enzyme Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Jinyu Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China; National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, Fuzhou University, Fujian, China.
| | - Peng Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350116, China; National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, Fuzhou University, Fujian, China; Fujian Key Laboratory of Marine Enzyme Engineering, Fuzhou University, Fuzhou, Fujian 350108, China.
| |
Collapse
|
29
|
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: 24] [Impact Index Per Article: 6.0] [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
|
30
|
Krishnan M, Choi J, Choi S, Kim Y. Anti-Endotoxin 9-Meric Peptide with Therapeutic Potential for the Treatment of Endotoxemia. J Microbiol Biotechnol 2021; 31:25-32. [PMID: 33263333 PMCID: PMC9705858 DOI: 10.4014/jmb.2011.11011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022]
Abstract
Inflammatory reactions activated by lipopolysaccharide (LPS) of gram-negative bacteria can lead to severe septic shock. With the recent emergence of multidrug-resistant gram-negative bacteria and a lack of efficient ways to treat resulting infections, there is a need to develop novel anti-endotoxin agents. Antimicrobial peptides have been noticed as potential therapeutic molecules for bacterial infection and as candidates for new antibiotic drugs. We previously designed the 9-meric antimicrobial peptide Pro9-3 and it showed high antimicrobial activity against gram-negative bacteria. Here, to further examine its potency as an anti-endotoxin agent, we examined the antiendotoxin activities of Pro9-3 and elucidated its mechanism of action. We performed a dye-leakage experiment and BODIPY-TR cadaverine and limulus amebocyte lysate assays for Pro9-3 as well as its lysine-substituted analogue and their enantiomers. The results confirmed that Pro9-3 targets the bacterial membrane and the arginine residues play key roles in its antimicrobial activity. Pro9-3 showed excellent LPS-neutralizing activity and LPS-binding properties, which were superior to those of other peptides. Saturation transfer difference-nuclear magnetic resonance experiments to explore the interaction between LPS and Pro9-3 revealed that Trp3 and Tlr7 in Pro9-3 are critical for attracting Pro9-3 to the LPS in the gram-negative bacterial membrane. Moreover, the anti-septic effect of Pro9-3 in vivo was investigated using an LPS-induced endotoxemia mouse model, demonstrating its dual activities: antibacterial activity against gram-negative bacteria and immunosuppressive effect preventing LPS-induced endotoxemia. Collectively, these results confirmed the therapeutic potential of Pro9-3 against infection of gram-negative bacteria.
Collapse
Affiliation(s)
- Manigandan Krishnan
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Joonhyeok Choi
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Sungjae Choi
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Yangmee Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea,Corresponding author Phone: +82-2-450-3421 Fax: +82-2-447-5987 E-mail:
| |
Collapse
|
31
|
Mohammad S, Thiemermann C. Role of Metabolic Endotoxemia in Systemic Inflammation and Potential Interventions. Front Immunol 2021; 11:594150. [PMID: 33505393 PMCID: PMC7829348 DOI: 10.3389/fimmu.2020.594150] [Citation(s) in RCA: 214] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Diet-induced metabolic endotoxemia is an important factor in the development of many chronic diseases in animals and man. The gut epithelium is an efficient barrier that prevents the absorption of liposaccharide (LPS). Structural changes to the intestinal epithelium in response to dietary alterations allow LPS to enter the bloodstream, resulting in an increase in the plasma levels of LPS (termed metabolic endotoxemia). LPS activates Toll-like receptor-4 (TLR4) leading to the production of numerous pro-inflammatory cytokines and, hence, low-grade systemic inflammation. Thus, metabolic endotoxemia can lead to several chronic inflammatory conditions. Obesity, diabetes, and non-alcoholic fatty liver disease (NAFLD) can also cause an increase in gut permeability and potential pharmacological and dietary interventions could be used to reduce the chronic low-grade inflammation associated with endotoxemia.
Collapse
Affiliation(s)
- Shireen Mohammad
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
32
|
Marimuthu SK, Nagarajan K, Perumal SK, Palanisamy S, Subbiah L. Structural stability of antimicrobial peptides rich in tryptophan, proline and arginine: a computational study. J Biomol Struct Dyn 2020; 40:3551-3559. [PMID: 33210568 DOI: 10.1080/07391102.2020.1848631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The host defense peptides or antimicrobial peptides (AMPs) often contain short sequence of amino acids, either positive or negatively charged and express broad-spectrum antibacterial, antiviral and antifungal activity. Many researchers had reported that tryptophan, arginine and proline rich AMPs have a promising source of next-generation antibiotics. Nowadays, AMPs are used as a possible therapeutic source for future antibiotics. In the present study, the amino acid sequences of 2924 AMPs belonging to various sources rich in Tryptophan, Proline and Arginine was chosen for investigation. The AMPs were further categorized according to their source, structure and antimicrobial activities. The AMPs with tryptophan, arginine, proline residues in abundance with maximum sequence length of 20 amino acids alone was obtained. Homology modeling was performed with PEP-FOLD and the modeled structures were evaluated using RAMPAGE to identify the structural information. Further, the stability of peptide in aqueous condition was probed using molecular dynamics simulations.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sathish Kumar Marimuthu
- Department of Pharmaceutical Technology, Centre for Excellence in Nanobio Translational Research, University College of Engineering, Anna University, Bharathidasan Institute of Technology (BIT) Campus, Tiruchirappalli, Tamilnadu, India
| | - Krishnanand Nagarajan
- Department of Pharmaceutical Technology, Centre for Excellence in Nanobio Translational Research, University College of Engineering, Anna University, Bharathidasan Institute of Technology (BIT) Campus, Tiruchirappalli, Tamilnadu, India
| | - Sathish Kumar Perumal
- Department of Pharmaceutical Technology, Centre for Excellence in Nanobio Translational Research, University College of Engineering, Anna University, Bharathidasan Institute of Technology (BIT) Campus, Tiruchirappalli, Tamilnadu, India
| | - Selvamani Palanisamy
- Department of Pharmaceutical Technology, Centre for Excellence in Nanobio Translational Research, University College of Engineering, Anna University, Bharathidasan Institute of Technology (BIT) Campus, Tiruchirappalli, Tamilnadu, India
| | - Latha Subbiah
- Department of Pharmaceutical Technology, Centre for Excellence in Nanobio Translational Research, University College of Engineering, Anna University, Bharathidasan Institute of Technology (BIT) Campus, Tiruchirappalli, Tamilnadu, India
| |
Collapse
|
33
|
Browne K, Chakraborty S, Chen R, Willcox MDP, Black DS, Walsh WR, Kumar N. A New Era of Antibiotics: The Clinical Potential of Antimicrobial Peptides. Int J Mol Sci 2020; 21:E7047. [PMID: 32987946 PMCID: PMC7582481 DOI: 10.3390/ijms21197047] [Citation(s) in RCA: 239] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Antimicrobial resistance is a multifaceted crisis, imposing a serious threat to global health. The traditional antibiotic pipeline has been exhausted, prompting research into alternate antimicrobial strategies. Inspired by nature, antimicrobial peptides are rapidly gaining attention for their clinical potential as they present distinct advantages over traditional antibiotics. Antimicrobial peptides are found in all forms of life and demonstrate a pivotal role in the innate immune system. Many antimicrobial peptides are evolutionarily conserved, with limited propensity for resistance. Additionally, chemical modifications to the peptide backbone can be used to improve biological activity and stability and reduce toxicity. This review details the therapeutic potential of peptide-based antimicrobials, as well as the challenges needed to overcome in order for clinical translation. We explore the proposed mechanisms of activity, design of synthetic biomimics, and how this novel class of antimicrobial compound may address the need for effective antibiotics. Finally, we discuss commercially available peptide-based antimicrobials and antimicrobial peptides in clinical trials.
Collapse
Affiliation(s)
- Katrina Browne
- School of Chemistry, University of New South Wales (UNSW) Sydney, Sydney 2052, Australia; (K.B.); (S.C.); (R.C.)
| | - Sudip Chakraborty
- School of Chemistry, University of New South Wales (UNSW) Sydney, Sydney 2052, Australia; (K.B.); (S.C.); (R.C.)
| | - Renxun Chen
- School of Chemistry, University of New South Wales (UNSW) Sydney, Sydney 2052, Australia; (K.B.); (S.C.); (R.C.)
| | - Mark DP Willcox
- School of Optometry and Vision Science, University of New South Wales (UNSW) Sydney, Sydney 2052, Australia;
| | - David StClair Black
- School of Chemistry, University of New South Wales (UNSW) Sydney, Sydney 2052, Australia; (K.B.); (S.C.); (R.C.)
| | - William R Walsh
- Surgical and Orthopaedic Research Laboratories (SORL), Prince of Wales Clinical School, Prince of Wales Hospital, University of New South Wales (UNSW), Randwick 2031, Australia;
| | - Naresh Kumar
- School of Chemistry, University of New South Wales (UNSW) Sydney, Sydney 2052, Australia; (K.B.); (S.C.); (R.C.)
| |
Collapse
|
34
|
Alford MA, Baquir B, Santana FL, Haney EF, Hancock REW. Cathelicidin Host Defense Peptides and Inflammatory Signaling: Striking a Balance. Front Microbiol 2020; 11:1902. [PMID: 32982998 PMCID: PMC7481365 DOI: 10.3389/fmicb.2020.01902] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/20/2020] [Indexed: 12/28/2022] Open
Abstract
Host-defense peptides (HDPs) are vital components of innate immunity in all vertebrates. While their antibacterial activity toward bacterial cells was the original focus for research, their ability to modulate immune and inflammatory processes has emerged as one of their major functions in the host and as a promising approach from which to develop novel therapeutics targeting inflammation and innate immunity. In this review, with particular emphasis on the cathelicidin family of peptides, the roles of natural HDPs are examined in managing immune activation, cellular recruitment, cytokine responses, and inflammation in response to infection, as well as their contribution(s) to various inflammatory disorders and autoimmune diseases. Furthermore, we discuss current efforts to develop synthetic HDPs as therapeutics aimed at restoring balance to immune responses that are dysregulated and contribute to disease pathologies.
Collapse
Affiliation(s)
- Morgan A. Alford
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Beverlie Baquir
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Felix L. Santana
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Evan F. Haney
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Robert E. W. Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
35
|
Fattahi F, Grailer JJ, Parlett M, Lu H, Malan EA, Abe E, Russell MW, Frydrych LM, Delano MJ, Zetoune FS, Ward PA. Requirement of Complement C6 for Intact Innate Immune Responses in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:251-260. [PMID: 32444389 DOI: 10.4049/jimmunol.1900801] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 04/01/2020] [Indexed: 12/11/2022]
Abstract
Over the first days of polymicrobial sepsis, there is robust activation of the innate immune system, causing the appearance of proinflammatory cytokines and chemokines, along with the appearance of extracellular histones, which are highly proinflammatory and prothrombotic. In the current study, we studied different innate immune responses in mice with knockout (KO) of complement protein 6 (C6). Polymorphonuclear neutrophils (PMNs) from these KO mice had defective innate immune responses, including defective expression of surface adhesion molecules, generation of superoxide anion, and appearance of reactive oxygen species and histone release after activation of PMNs, along with defective phagocytosis. In addition, in C6-/- mice, the NLRP3 inflammasome was defective both in PMNs and in macrophages. When these KO mice were subjected to polymicrobial sepsis, their survival was improved, associated with reduced levels in the plasma of proinflammatory cytokines and chemokines and lower levels of histones in plasma. In addition, sepsis-induced cardiac dysfunction was attenuated in these KO mice. In a model of acute lung injury induced by LPS, C6-/- mice showed reduced PMN buildup and less lung epithelial/endothelial cell dysfunction (edema and hemorrhage). These data indicate that C6-/- mice have reduced innate immune responses that result in less organ injury and improved survival after polymicrobial sepsis.
Collapse
Affiliation(s)
- Fatemeh Fattahi
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Jamison J Grailer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Michella Parlett
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Hope Lu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Elizabeth A Malan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Elizabeth Abe
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Mark W Russell
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Lynn M Frydrych
- Department of Surgery, University of Michigan School Medical School, Ann Arbor, MI 48109
| | - Matthew J Delano
- Department of Surgery, University of Michigan School Medical School, Ann Arbor, MI 48109
| | - Firas S Zetoune
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Peter A Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109;
| |
Collapse
|
36
|
Ahmad B, Li Z, Hanif Q, Hu Q, Wei X, Zhang L, Khan SA, Aihemaiti M, Gulzar H, Shahid M, Si D, Zhang R. A Hybrid Peptide DEFB-TP5 Expressed in Methylotrophic Yeast Neutralizes LPS With Potent Anti-inflammatory Activities. Front Pharmacol 2020; 11:461. [PMID: 32457599 PMCID: PMC7221121 DOI: 10.3389/fphar.2020.00461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/24/2020] [Indexed: 12/31/2022] Open
Abstract
DEFB-TP5 is a novel auspicious health-beneficial peptide derivative from two naturally occurring peptides, β-Defensin (DEFB) and thymopentin (TP5), and shows strong anti-inflammatory activity and binds to LPS without cytotoxicity and hemolytic effect. Furthermore, the application of DEFB-TP5 peptide is inadequate by its high cost. In the current study, we developed a biocompatible mechanism for expression of the DEFB-TP5 peptide in Pichia pastoris. The transgenic strain of hybrid DEFB-TP5 peptide with a molecular weight of 6.7kDa as predictable was obtained. The recombinant DEFB-TP5 peptide was purified by Ni-NTA chromatography, estimated 30.41 mg/L was obtained from the cell culture medium with 98.2% purity. Additionally, The purified DEFB-TP5 peptide significantly (p< 0.05) diminished the release of nitric oxide (NO), TNF-α, IL-6, IL-1β in LPS-stimulated RAW264.7 macrophages in a dose-dependent manner. This study will not only help to understand the molecular mechanism of expression that can potentially be used to develop an anti-endotoxin peptide but also to serve as the basis for the development of antimicrobial and anti-inflammatory agents as well, which also provides a potential source for the production of recombinant bioactive DEFB-TP5 at the industrial level.
Collapse
Affiliation(s)
- Baseer Ahmad
- State Key Laboratory of Animal Nutrition and Feed Sciences, Laboratory of Feed Biotechnology, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhongxuan Li
- State Key Laboratory of Animal Nutrition and Feed Sciences, Laboratory of Feed Biotechnology, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Quratulain Hanif
- Computational Biology Laboratory, Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic Engineering, Faisalabad, Pakistan.,Department of Biotechnology, Pakistan Institute of Engineering and Applied Sciences, Nilore, Islamabad, Pakistan
| | - Qingyong Hu
- State Key Laboratory of Animal Nutrition and Feed Sciences, Laboratory of Feed Biotechnology, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xubiao Wei
- State Key Laboratory of Animal Nutrition and Feed Sciences, Laboratory of Feed Biotechnology, College of Animal Science and Technology, China Agricultural University, Beijing, China.,College of Life Sciences, Peking University, Beijing, China
| | - Lulu Zhang
- State Key Laboratory of Animal Nutrition and Feed Sciences, Laboratory of Feed Biotechnology, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shahzad Akbar Khan
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Maierhaba Aihemaiti
- State Key Laboratory of Animal Nutrition and Feed Sciences, Laboratory of Feed Biotechnology, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Huma Gulzar
- College of Life Sciences, China Agricultural University, Beijing, China
| | - Muhammad Shahid
- State Key Laboratory of Animal Nutrition and Feed Sciences, Laboratory of Feed Biotechnology, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dayong Si
- State Key Laboratory of Animal Nutrition and Feed Sciences, Laboratory of Feed Biotechnology, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Rijun Zhang
- State Key Laboratory of Animal Nutrition and Feed Sciences, Laboratory of Feed Biotechnology, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
37
|
Sasani N, Roghanian R, Emtiazi G, Aghaie A. A Novel Approach on Leukodepletion Filters: Investigation of Synergistic Anticancer Effect of Purified α-Defensins and Nisin. Adv Pharm Bull 2020; 11:378-384. [PMID: 33880361 PMCID: PMC8046393 DOI: 10.34172/apb.2021.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 12/09/2019] [Accepted: 04/15/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose: There are number of reports available regarding defensins activity against mammalian cells besides their antimicrobial and immune regulatory activities. This study aims to investigate anticancer and apoptosis activity of the purified defensins from leukodepletion filters alone or in synergism with bacterial peptide, nisin, on prostate and colorectal cancer. Methods: Leucoflex LCR-5 filters were backflushed by an optimized elution system. Isolated granulocytes were sonicated and the supernatant treated before further purification by high performance liquid chromatography (HPLC). SDS-PAGE and western blot testing verified the fraction. Cell culture on PC-3 (human prostate adenocarcinoma), and HCT-116 (human colorectal carcinoma) were conducted following by MTT assays in addition to annexin flow cytometry for sole and synergistic effects with peptide nisin. Results: Viable and active neutrophils could recover, and α-defensins were extracted and purified. Combinations of an optimal dose of α-defensins and nisin showed a remarkable synergistic effect on cancer cell lines (over 90% and 70% for PC-3 and HCT-116, respectively). Conclusion: It also observed that less than 40% of both cells could survive after co-treatment with optimal dose. Also, apoptosis was increased after treatment by these peptides together. Annexin Vpositive populations significantly increased in percentage in comparison with control.
Collapse
Affiliation(s)
- Niloofar Sasani
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, P. O Box 81746-79441, Isfahan, Iran
| | - Rasoul Roghanian
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, P. O Box 81746-79441, Isfahan, Iran
| | - Giti Emtiazi
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, P. O Box 81746-79441, Isfahan, Iran
| | - Afsaneh Aghaie
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
38
|
Romo1-Derived Antimicrobial Peptide Is a New Antimicrobial Agent against Multidrug-Resistant Bacteria in a Murine Model of Sepsis. mBio 2020; 11:mBio.03258-19. [PMID: 32291307 PMCID: PMC7157825 DOI: 10.1128/mbio.03258-19] [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] [Indexed: 02/06/2023] Open
Abstract
Abuse of antibiotics often leads to increase of multidrug-resistant (MDR) bacteria, which threatens the life of human beings. To overcome threat of antibiotic resistance, scientists are developing a novel class of antibiotics, antimicrobial peptides, that can eradicate MDR bacteria. Unfortunately, these antibiotics have mainly been developed to cure bacterial skin infections rather than others, such as life-threatening sepsis. Major pharmaceutical companies have tried to develop antiseptic drugs; however, they have not been successful. Here, we report that AMPR-11, the antimicrobial peptide (AMP) derived from mitochondrial nonselective channel Romo1, has antimicrobial activity against Gram-positive and Gram-negative bacteria comprising many clinically isolated MDR strains. Moreover, AMPR-11 increased the survival rate in a murine model of sepsis caused by MDR bacteria. We propose that AMPR-11 could be a novel antiseptic drug candidate with a broad antimicrobial spectrum to overcome MDR bacterial infection. To overcome increasing bacterial resistance to conventional antibiotics, many antimicrobial peptides (AMPs) derived from host defense proteins have been developed. However, there are considerable obstacles to their application to systemic infections because of their low bioavailability. In the present study, we developed an AMP derived from Romo1 (AMPR-11) that exhibits a broad spectrum of antimicrobial activity. AMPR-11 showed remarkable efficacy against sepsis-causing bacteria, including multidrug-resistant strains, with low toxicity in a murine model of sepsis after intravenous administration. It seems that AMPR-11 disrupts bacterial membranes by interacting with cardiolipin and lipid A. From the results of this study, we suggest that AMPR-11 is a new class of agent for overcoming low efficacy in the intravenous application of AMPs and is a promising candidate to overcome multidrug resistance.
Collapse
|
39
|
Fernández de Ullivarri M, Arbulu S, Garcia-Gutierrez E, Cotter PD. Antifungal Peptides as Therapeutic Agents. Front Cell Infect Microbiol 2020; 10:105. [PMID: 32257965 PMCID: PMC7089922 DOI: 10.3389/fcimb.2020.00105] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/27/2020] [Indexed: 12/17/2022] Open
Abstract
Fungi have been used since ancient times in food and beverage-making processes and, more recently, have been harnessed for the production of antibiotics and in processes of relevance to the bioeconomy. Moreover, they are starting to gain attention as a key component of the human microbiome. However, fungi are also responsible for human infections. The incidence of community-acquired and nosocomial fungal infections has increased considerably in recent decades. Antibiotic resistance development, the increasing number of immunodeficiency- and/or immunosuppression-related diseases and limited therapeutic options available are triggering the search for novel alternatives. These new antifungals should be less toxic for the host, with targeted or broader antimicrobial spectra (for diseases of known and unknown etiology, respectively) and modes of actions that limit the potential for the emergence of resistance among pathogenic fungi. Given these criteria, antimicrobial peptides with antifungal properties, i.e., antifungal peptides (AFPs), have emerged as powerful candidates due to their efficacy and high selectivity. In this review, we provide an overview of the bioactivity and classification of AFPs (natural and synthetic) as well as their mode of action and advantages over current antifungal drugs. Additionally, natural, heterologous and synthetic production of AFPs with a view to greater levels of exploitation is discussed. Finally, we evaluate the current and potential applications of these peptides, along with the future challenges relating to antifungal treatments.
Collapse
Affiliation(s)
- Miguel Fernández de Ullivarri
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Food Bioscience Department, Teagasc Food Research Centre, Fermoy, Ireland
| | - Sara Arbulu
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Food Bioscience Department, Teagasc Food Research Centre, Fermoy, Ireland
| | - Enriqueta Garcia-Gutierrez
- Food Bioscience Department, Teagasc Food Research Centre, Fermoy, Ireland.,Gut Microbes and Health, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Paul D Cotter
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Food Bioscience Department, Teagasc Food Research Centre, Fermoy, Ireland
| |
Collapse
|
40
|
Shruti SR, Rajasekaran R. Identification of therapeutic peptide scaffold from tritrpticin family for urinary tract infections using in silico techniques. J Biomol Struct Dyn 2019; 38:4407-4417. [DOI: 10.1080/07391102.2019.1680437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- S. R. Shruti
- Department of Biotechnology, School of Biosciences and Technology, VIT (Deemed to Be University), Vellore, India
| | - R. Rajasekaran
- Department of Biotechnology, School of Biosciences and Technology, VIT (Deemed to Be University), Vellore, India
| |
Collapse
|
41
|
Correa W, Brandenburg J, Behrends J, Heinbockel L, Reiling N, Paulowski L, Schwudke D, Stephan K, Martinez-de-Tejada G, Brandenburg K, Gutsmann T. Inactivation of Bacteria by γ-Irradiation to Investigate the Interaction with Antimicrobial Peptides. Biophys J 2019; 117:1805-1819. [PMID: 31676134 DOI: 10.1016/j.bpj.2019.10.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/30/2019] [Accepted: 10/08/2019] [Indexed: 11/27/2022] Open
Abstract
The activity of antimicrobial peptides (AMPs) has been investigated extensively using model membranes composed of phospholipids or lipopolysaccharides in aqueous environments. However, from a biophysical perspective, there is a large scientific interest regarding the direct interaction of membrane-active peptides with whole bacteria. Working with living bacteria limits the usability of experimental setups and the interpretation of the resulting data because of safety risks and the overlap of active and passive effects induced by AMPs. We killed or inactivated metabolic-active bacteria using γ-irradiation or sodium azide, respectively. Microscopy, flow cytometry, and SYTOX green assays showed that the cell envelope remained intact to a high degree at the minimal bactericidal dose. Furthermore, the tumor-necrosis-factor-α-inducing activity of the lipopolysaccharides and the chemical lipid composition was unchanged. Determining the binding capacity of AMPs to the bacterial cell envelope by calorimetry is difficult because of an overlapping of the binding heat and metabolic activities of the bacteria-induced by the AMPs. The inactivation of all active processes helps to decipher the complex thermodynamic information. From the isothermal titration calorimetry (ITC) results, we propose that the bacterial membrane potential (Δψ) is possibly an underestimated modulator of the AMP activity. The negative surface charge of the outer leaflet of the outer membrane of Gram-negative bacteria is already neutralized by peptide concentrations below the minimal inhibitory concentration. This proves that peptide aggregation on the bacterial membrane surface plays a decisive role in the degree of antimicrobial activity. This will not only enable many biophysical approaches for the investigation between bacteria and membrane-active peptides in the future but will also make it possible to compare biophysical parameters of active and inactive bacteria. This opens up new possibilities to better understand the active and passive interaction processes between AMPs and bacteria.
Collapse
Affiliation(s)
- Wilmar Correa
- Division of Biophysics, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany.
| | - Julius Brandenburg
- Microbial Interface Biology, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Jochen Behrends
- Fluorescence Cytometry Department, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | | | - Norbert Reiling
- Microbial Interface Biology, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Laura Paulowski
- Division of Biophysics, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Dominik Schwudke
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Kerstin Stephan
- Division of Biophysics, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | | | - Klaus Brandenburg
- Brandenburg Antiinfektiva GmbH, c/o Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Thomas Gutsmann
- Division of Biophysics, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| |
Collapse
|
42
|
Jang M, Kim J, Choi Y, Bang J, Kim Y. Antiseptic Effect of Ps-K18: Mechanism of Its Antibacterial and Anti-Inflammatory Activities. Int J Mol Sci 2019; 20:E4895. [PMID: 31581682 PMCID: PMC6801626 DOI: 10.3390/ijms20194895] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/29/2019] [Accepted: 09/30/2019] [Indexed: 12/16/2022] Open
Abstract
Recently, bioactive peptides have attracted attention for their therapeutic applications in the pharmaceutical industry. Among them, antimicrobial peptides are candidates for new antibiotic drugs. Since pseudin-2 (Ps), isolated from the skin of the paradoxical frog Pseudis paradoxa, shows broad-spectrum antibacterial activity with high cytotoxicity, we previously designed Ps-K18 with a Lys substitution for Leu18 in Ps, which showed high antibacterial activity and low toxicity. Here, we examined the potency of Ps-K18, aiming to develop antibiotics derived from bioactive peptides for the treatment of Gram-negative sepsis. We first investigated the antibacterial mechanism of Ps-K18 based on confocal micrographs and field emission scanning electron microscopy, confirming that Ps-K18 targets the bacterial membrane. Anti-inflammatory mechanism of Ps-K18 was investigated by secreted alkaline phosphatase reporter gene assays and RT-PCR, which revealed that Ps-K18 activates innate defense via Toll-like receptor 4-mediated nuclear factor-kappa B signaling pathways. Moreover, we investigated the antiseptic effect of Ps-K18 using a lipopolysaccharide or Escherichia coli K1-induced septic shock mouse model. Ps-K18 significantly reduced bacterial growth and inflammatory responses in the septic shock model. Ps-K18 showed low renal and liver toxicity and attenuated lung damage effectively. This study suggests that Ps-K18 is a potent peptide antibiotic that could be applied therapeutically to Gram-negative sepsis.
Collapse
Affiliation(s)
- Mihee Jang
- Department of Bioscience and Biotechnology, Research Institute for Bioactive-Metabolome Network, Konkuk University, Seoul 05029, Korea.
| | - Jieun Kim
- Department of Bioscience and Biotechnology, Research Institute for Bioactive-Metabolome Network, Konkuk University, Seoul 05029, Korea.
| | - Yujin Choi
- Chuncheon Center, Korea Basic Science Institute, Chuncheon 24341, Korea.
| | - JeongKyu Bang
- Protein Structure Group, Korea Basic Science Institute, Ochang, Cheongju, Chung-Buk 28199, Korea.
| | - Yangmee Kim
- Department of Bioscience and Biotechnology, Research Institute for Bioactive-Metabolome Network, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
43
|
Novel Synthetic, Host-defense Peptide Protects Against Organ Injury/Dysfunction in a Rat Model of Severe Hemorrhagic Shock. Ann Surg 2019; 268:348-356. [PMID: 28288070 DOI: 10.1097/sla.0000000000002186] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To evaluate (1) levels of the host-defense/antimicrobial peptide LL-37 in patients with trauma and hemorrhagic shock (HS) and (2) the effects of a synthetic host-defense peptide; Pep19-4LF on multiple organ failure (MOF) associated with HS. BACKGROUND HS is a common cause of death in severely injured patients. There is no specific therapy that reduces HS-associated MOF. METHODS (1) LL-37 was measured in 47 trauma/HS patients admitted to an urban major trauma center. (2) Male Wistar rats were submitted to HS (90 min, target mean arterial pressure: 27-32 mm Hg) or sham operation. Rats were treated with Pep19-4LF [66 (n = 8) or 333 μg/kg · h (n = 8)] or vehicle (n = 12) for 4 hours following resuscitation. RESULTS Plasma LL-37 was 12-fold higher in patients with trauma/HS compared to healthy volunteers. HS rats treated with Pep19-4LF (high dose) had a higher mean arterial pressure at the end of the 4-hour resuscitation period (79 ± 4 vs 54 ± 5 mm Hg) and less renal dysfunction, liver injury, and lung inflammation than HS rats treated with vehicle. Pep19-4LF enhanced (kidney/liver) the phosphorylation of (1) protein kinase B and (2) endothelial nitric oxide synthase. Pep19-4LF attenuated the HS-induced (1) translocation of p65 from cytosol to nucleus, (2) phosphorylation of IκB kinase on Ser, and (3) phosphorylation of IκBα on Ser resulting in inhibition of nuclear factor kappa B and formation of proinflammatory cytokines. Pep19-4LF prevented the release of tumor necrosis factor alpha caused by heparan sulfate in human mononuclear cells by binding to this damage-associated molecular pattern. CONCLUSIONS Trauma-associated HS results in release of LL-37. The synthetic host-defense/antimicrobial peptide Pep19-4LF attenuates the organ injury/dysfunction associated with HS.
Collapse
|
44
|
Sallustio F, Stasi A, Curci C, Divella C, Picerno A, Franzin R, De Palma G, Rutigliano M, Lucarelli G, Battaglia M, Staffieri F, Crovace A, Pertosa GB, Castellano G, Gallone A, Gesualdo L. Renal progenitor cells revert LPS-induced endothelial-to-mesenchymal transition by secreting CXCL6, SAA4, and BPIFA2 antiseptic peptides. FASEB J 2019; 33:10753-10766. [PMID: 31268775 DOI: 10.1096/fj.201900351r] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endothelial dysfunction is a hallmark of LPS-induced acute kidney injury (AKI). Endothelial cells (ECs) acquired a fibroblast-like phenotype and contributed to myofibroblast generation through the endothelial-to-mesenchymal transition (EndMT) process. Of note, human adult renal stem/progenitor cells (ARPCs) enhance the tubular regenerative mechanism during AKI but little is known about their effects on ECs. Following LPS exposure, ECs proliferated, decreased EC markers CD31 and vascular endothelial cadherin, and up-regulated myofibroblast markers, collagen I, and vimentin. The coculture with ARPCs normalized the EC proliferation rate and abrogated the LPS-induced EndMT. The gene expression analysis showed that most of the genes modulated in LPS-stimulated ARPCs belong to cell activation and defense response pathways. We showed that the ARPC-specific antifibrotic effect is exerted by the secretion of CXCL6, SAA4, and BPIFA2 produced after the anaphylatoxin stimulation. Next, we investigated the molecular signaling that underlies the ARPC protective mechanism and found that renal progenitors diverge from differentiated tubular cells and ECs in myeloid differentiation primary response 88-independent pathway activation. Finally, in a swine model of LPS-induced AKI, we observed that activated ARPCs secreted CXCL6, SAA4, and BPIFA2 as a defense response. These data open new perspectives on the treatment of both sepsis- and endotoxemia-induced AKI, suggesting an underestimated role of ARPCs in preventing endothelial dysfunction and novel strategies to protect the endothelial compartment and promote kidney repair.-Sallustio, F., Stasi, A., Curci, C., Divella, C., Picerno, A., Franzin, R., De Palma, G., Rutigliano, M., Lucarelli, G., Battaglia, M., Staffieri, F., Crovace, A., Pertosa, G. B., Castellano, G., Gallone, A., Gesualdo, L. Renal progenitor cells revert LPS-induced endothelial-to-mesenchymal transition by secreting CXCL6, SAA4, and BPIFA2 antiseptic peptides.
Collapse
Affiliation(s)
- Fabio Sallustio
- Department of Basic Medical Sciences, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy.,Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Alessandra Stasi
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Claudia Curci
- Department of Basic Medical Sciences, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy.,Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Chiara Divella
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Angela Picerno
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Rossana Franzin
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe De Palma
- Institutional Biobank, Experimental Oncology and Biobank Management Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori Bari Giovanni Paolo II, Bari, Italy
| | - Monica Rutigliano
- Urology, Andrology, and Renal Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Lucarelli
- Urology, Andrology, and Renal Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Michele Battaglia
- Urology, Andrology, and Renal Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Staffieri
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Crovace
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Battista Pertosa
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Anna Gallone
- Department of Basic Medical Sciences, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis, and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
45
|
Nakamura T, Satoh-Nakamura T, Nakajima A, Kawanami T, Sakai T, Fujita Y, Iwao H, Miki M, Masaki Y, Okazaki T, Ishigaki Y, Kawano M, Yamada K, Matsui S, Saeki T, Kamisawa T, Yamamoto M, Hamano H, Origuchi T, Hirata S, Tanaka Y, Tsuboi H, Sumida T, Okazaki K, Tanaka M, Chiba T, Mimori T, Umehara H. Impaired expression of innate immunity-related genes in IgG4-related disease: A possible mechanism in the pathogenesis of IgG4-RD. Mod Rheumatol 2019; 30:551-557. [PMID: 31116057 DOI: 10.1080/14397595.2019.1621475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background: IgG4-related disease (IgG4-RD) is characterized by elevated serum IgG4 and tissue infiltration by IgG4-positive plasma cells. The pathogenesis of this disease is not clear. Transcriptome analysis was performed to identify genes over- and under-expressed in patients with IgG4-RD.Method: DNA microarray analysis was performed using RNA from peripheral blood mononuclear cells of two patients with IgG4-RD and four healthy individuals. Genes showing a greater than threefold change in expression in IgG4-RD patients following steroid therapy were identified. Four genes related to innate immunity such as transcobalamin I (TCN1), secretory leukocyte peptidase inhibitor (SLPI), bactericidal/permeability-increasing protein (BPI) and lactotransferrin (LTF) were assessed by real-time PCR in 15 IgG4-RD patients and 13 healthy individuals.Result: DNA microarray analysis identified 30 genes showing a greater than threefold change in expression in IgG4-RD patients following steroid therapy. Real-time RT-PCR showed that the levels of mRNAs encoding TCNI and SLPI, except for BPI and LTF, were significantly lower in patients with IgG4-RD than in healthy people. The levels of all four mRNAs in patients with IgG4-RD were significantly increased after steroid treatment.Conclusion: These results indicate that reduction in expression of innate immunity-related genes may participate in the pathogenesis of IgG4-RD that steroid treatment may rectify impaired innate immunity as well as acquired immunity.
Collapse
Affiliation(s)
- Takuji Nakamura
- Department of Rheumatology and Immunology, Nagahama City Hospital, Shiga, Japan.,Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan
| | - Tomomi Satoh-Nakamura
- Department of Rheumatology and Immunology, Nagahama City Hospital, Shiga, Japan.,Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan
| | - Akio Nakajima
- Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan.,Division of Rheumatology, Kudo General Hospital, Ishikawa, Japan
| | - Takafumi Kawanami
- Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan
| | - Tomoyuki Sakai
- Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan
| | - Yoshimasa Fujita
- Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan
| | - Haruka Iwao
- Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan
| | - Miyuki Miki
- Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan
| | - Yasufumi Masaki
- Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan
| | - Toshiro Okazaki
- Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan
| | - Yasuhito Ishigaki
- Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Mitsuhiro Kawano
- Division of Rheumatology, Department of Internal Medicine, Kanazawa University Hospital, Ishikawa, Japan
| | - Kazunori Yamada
- Division of Rheumatology, Department of Internal Medicine, Kanazawa University Hospital, Ishikawa, Japan
| | - Shoko Matsui
- Health Administration Center, University of Toyama, Toyama, Japan
| | - Takako Saeki
- Department of Internal Medicine, Nagaoka Red Cross Hospital, Niigata, Japan
| | - Terumi Kamisawa
- Department of Internal Medicine, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | - Motohisa Yamamoto
- Department of Rheumatology, Sapporo Medical University School of Medicine, Hokkaido, Japan
| | - Hideaki Hamano
- Medical Informatics Division and Department of Internal Medicine, Gastroenterology, Shinshu University School Hospital, Nagano, Japan
| | - Tomoki Origuchi
- First Department of Internal Medicine, Department of Immunology and Rheumatology, Nagasaki Graduate School of Health Sciences, Nagasaki, Japan
| | - Shintaro Hirata
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan.,Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Hiroto Tsuboi
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kazuichi Okazaki
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Osaka, Japan
| | - Masao Tanaka
- Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan.,Department of Clinical Immunology, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Tsutomu Chiba
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tsuneyo Mimori
- Department of Clinical Immunology, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Hisanori Umehara
- Department of Rheumatology and Immunology, Nagahama City Hospital, Shiga, Japan.,Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan
| |
Collapse
|
46
|
Vitamin D binding protein, but not vitamin D or vitamin D-related peptides, is associated with septic shock mortality. Enferm Infecc Microbiol Clin 2019; 37:239-243. [DOI: 10.1016/j.eimc.2018.06.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/13/2018] [Accepted: 06/17/2018] [Indexed: 01/24/2023]
|
47
|
Development of a novel short 12-meric papiliocin-derived peptide that is effective against Gram-negative sepsis. Sci Rep 2019; 9:3817. [PMID: 30846839 PMCID: PMC6405874 DOI: 10.1038/s41598-019-40577-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 02/19/2019] [Indexed: 01/08/2023] Open
Abstract
The development of novel peptide antibiotics with potent activity against multidrug-resistant Gram-negative bacteria and anti-septic activity is urgently needed. In this study, we designed short, 12-meric antimicrobial peptides by substituting amino acids from the N-terminal 12 residues of the papiliocin (Pap12-1) peptide to alter cationicity and amphipathicity and improve antibacterial activity and bacterial membrane interactions. Pap12-6, with an amphipathic α-helical structure and Trp12 at the C-terminus, showed broad-spectrum antibacterial activity, especially against multidrug-resistant Gram-negative bacteria. Dye leakage, membrane depolarization, and electron microscopy data proved that Pap12-6 kills bacteria by permeabilizing the bacterial membrane. Additionally, Pap12-6 significantly reduced the secretion of NO, TNF-α, and IL-6 and secreted alkaline phosphatase reporter gene activity confirmed that Pap12-6 shows anti-inflammatory activity via a TLR4-mediated NF-κB signaling pathway. In a mouse sepsis model, Pap12-6 significantly improved survival, reduced bacterial growth in organs, and reduced LPS and inflammatory cytokine levels in the serum and organs. Pap12-6 showed minimal cytotoxicity towards mammalian cells and controlled liver and kidney damage, proving its high bacterial selectivity. Our results suggest that Pap12-6 is a promising peptide antibiotic for the therapeutic treatment of Gram-negative sepsis via dual bactericidal and immunomodulatory effects on the host.
Collapse
|
48
|
Pimienta G, Heithoff DM, Rosa-Campos A, Tran M, Esko JD, Mahan MJ, Marth JD, Smith JW. Plasma Proteome Signature of Sepsis: a Functionally Connected Protein Network. Proteomics 2019; 19:e1800389. [PMID: 30706660 DOI: 10.1002/pmic.201800389] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/14/2019] [Indexed: 12/29/2022]
Abstract
Sepsis is an extreme host response to infection that leads to loss of organ function and cardiovascular integrity. Mortality from sepsis is on the rise. Despite more than three decades of research and clinical trials, specific diagnostic and therapeutic strategies for sepsis are still absent. The use of LFQ- and TMT-based quantitative proteomics is reported here to study the plasma proteome in five mouse models of sepsis. A knowledge-based interpretation of the data reveals a protein network with extensive connectivity through documented functional or physical interactions. The individual proteins in the network all have a documented role in sepsis and are known to be extracellular. The changes in protein abundance observed in the mouse models of sepsis have for the most part the same directionality (increased or decreased abundance) as reported in the literature for human sepsis. This network has been named the Plasma Proteome Signature of Sepsis (PPSS). The PPSS is a quantifiable molecular readout that can supplant the current symptom-based approach used to diagnose sepsis. This type of molecular interpretation of sepsis, its progression, and its response to therapeutic intervention are an important step in advancing our understanding of sepsis, and for discovering and evaluating new therapeutic strategies.
Collapse
Affiliation(s)
- Genaro Pimienta
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 9207, USA
| | - Douglas M Heithoff
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, 93106, USA.,Center for Nanomedicine, University of California, Santa Barbara, CA, 93106, USA
| | - Alexandre Rosa-Campos
- Proteomics Facility, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Minerva Tran
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 9207, USA
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Michael J Mahan
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, 93106, USA
| | - Jamey D Marth
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 9207, USA.,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, 93106, USA.,Center for Nanomedicine, University of California, Santa Barbara, CA, 93106, USA
| | - Jeffrey W Smith
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 9207, USA.,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, 93106, USA
| |
Collapse
|
49
|
Liquid chromatography-tandem mass spectrometry based method development and validation of S016-1271 (LR8P), a novel cationic antimicrobial peptide for its application to pharmacokinetic studies. J Pharm Biomed Anal 2019; 169:116-126. [PMID: 30851514 DOI: 10.1016/j.jpba.2019.01.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/31/2018] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
Abstract
S016-1271 (LR8P) is a broad spectrum novel cationic antimicrobial peptide. The objective of the present study was to develop a selective liquid chromatography-tandem mass spectrometry (LC-MS/MS) based bioanalytical method of S016-1271 peptide in mice and human plasma in order to uncover its pharmacokinetic aspects. The chromatographic separation of S016-1271 (FR8P as internal standard) was achieved on a Waters™ X select CSH-C18 column (75 × 3.0 mm, 2.5 μ) using mixture of acetonitrile and triple distilled water (TDW) both containing 0.05% formic acid as mobile phase. A seven minute linear gradient method was designed to separate analytes from ion suppression at a flow rate of 0.3 mL/min. The extraction of analytes from mice and human plasma was performed through solid phase extraction technique using mixed mode weak cation exchange cartridge (Thermo SOLA WCX 10 mg 1CC) with an extraction recovery of analytes about 75%. Mass spectrometric detection of S016-1271 and FR8P was performed with optimized multiple reaction monitoring (MRM) transitions (Q1/Q3) at 658.8 [M+3H] 3+/653.2 [M+3H-NH3] 3+ and 443.4 [M+5H]5+ /434.7 [y12-NH3]4+,respectively in positive electrospray ionization (ESI) mode. The linearity in mice and human plasma was established over a concentration range of 7.81-250 ng/mL with regression coefficient (r2 > 0.99). The currently developed method was validated as per US-FDA guidelines and found to be within the acceptable limits. The method was successfully applied to intravenous (IV) pharmacokinetic study in mice wherein the levels were detected upto 24 h. The peptide demonstrated poor distribution characteristics which were demonstrated through volume of distribution at steady state (202.71 ± 47.02 mL/kg less than total body water of mice; 580 mL/kg). The clearance of the peptide predominantly occurred through central compartment (central clearance is 25 fold greater than peripheral clearance). Also, the in vitro pharmacokinetic studies demonstrated the stability of S016-1271 in plasma and high plasma protein binding in mice and humans.
Collapse
|
50
|
Increased gene copy number of DEFA1/DEFA3 worsens sepsis by inducing endothelial pyroptosis. Proc Natl Acad Sci U S A 2019; 116:3161-3170. [PMID: 30718392 PMCID: PMC6386704 DOI: 10.1073/pnas.1812947116] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sepsis claims an estimated 30 million episodes and 6 million deaths per year, and treatment options are rather limited. Human neutrophil peptides 1-3 (HNP1-3) are the most abundant neutrophil granule proteins but their neutrophil content varies because of unusually extensive gene copy number polymorphism. A genetic association study found that increased copy number of the HNP-encoding gene DEFA1/DEFA3 is a risk factor for organ dysfunction during sepsis development. However, direct experimental evidence demonstrating that these risk alleles are pathogenic for sepsis is lacking because the genes are present only in some primates and humans. Here, we generate DEFA1/DEFA3 transgenic mice with neutrophil-specific expression of the peptides. We show that mice with high copy number of DEFA1/DEFA3 genes have more severe sepsis-related vital organ damage and mortality than mice with low copy number of DEFA1/DEFA3 or wild-type mice, resulting from more severe endothelial barrier dysfunction and endothelial cell pyroptosis after sepsis challenge. Mechanistically, HNP-1 induces endothelial cell pyroptosis via P2X7 receptor-mediating canonical caspase-1 activation in a NLRP3 inflammasome-dependent manner. Based on these findings, we engineered a monoclonal antibody against HNP-1 to block the interaction with P2X7 and found that the blocking antibody protected mice carrying high copy number of DEFA1/DEFA3 from lethal sepsis. We thus demonstrate that DEFA1/DEFA3 copy number variation strongly modulates sepsis development in vivo and explore a paradigm for the precision treatment of sepsis tailored by individual genetic information.
Collapse
|