1
|
Acharya Y, Taneja KK, Haldar J. Dual functional therapeutics: mitigating bacterial infection and associated inflammation. RSC Med Chem 2023; 14:1410-1428. [PMID: 37593575 PMCID: PMC10429821 DOI: 10.1039/d3md00166k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 05/21/2023] [Indexed: 08/19/2023] Open
Abstract
The emergence of antimicrobial resistance, coupled with the occurrence of persistent systemic infections, has already complicated clinical therapy efforts. Moreover, infections are also accompanied by strong inflammatory responses, generated by the host's innate and adaptive immune systems. The closely intertwined relationship between bacterial infection and inflammation has multiple implications on the ability of antibacterial therapeutics to tackle infection and inflammation. Particularly, uncontrolled inflammatory responses to infection can lead to sepsis, a life-threatening physiological condition. In this review, we discuss dual-functional antibacterial therapeutics that have potential to be developed for treating inflammation associated with bacterial infections. Immense research is underway that aims to develop new therapeutic agents that, when administered, regulate the excess inflammatory response, i.e. they have immunomodulatory properties along with the desired antibacterial activity. The classes of antibiotics that have immunomodulatory function in addition to antibacterial activity have been reviewed. Host defense peptides and their synthetic mimics are amongst the most sought-after solutions to develop such dual-functional therapeutics. This review also highlights the important classes of peptidomimetics that exhibit both antibacterial and immunomodulatory properties.
Collapse
Affiliation(s)
- Yash Acharya
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur Bengaluru 560064 Karnataka India
| | - Kashish Kumar Taneja
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur Bengaluru 560064 Karnataka India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur Bengaluru 560064 Karnataka India
- School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur Bengaluru 560064 Karnataka India
| |
Collapse
|
2
|
Kontou A, Kourti M, Iosifidis E, Sarafidis K, Roilides E. Use of Newer and Repurposed Antibiotics against Gram-Negative Bacteria in Neonates. Antibiotics (Basel) 2023; 12:1072. [PMID: 37370391 DOI: 10.3390/antibiotics12061072] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Antimicrobial resistance has become a significant public health problem globally with multidrug resistant Gram negative (MDR-GN) bacteria being the main representatives. The emergence of these pathogens in neonatal settings threatens the well-being of the vulnerable neonatal population given the dearth of safe and effective therapeutic options. Evidence from studies mainly in adults is now available for several novel antimicrobial compounds, such as new β-lactam/β-lactamase inhibitors (e.g., ceftazidime-avibactam, meropenem-vaborbactam, imipenem/cilastatin-relebactam), although old antibiotics such as colistin, tigecycline, and fosfomycin are also encompassed in the fight against MDR-GN infections that remain challenging. Data in the neonatal population are scarce, with few clinical trials enrolling neonates for the evaluation of the efficacy, safety, and dosing of new antibiotics, while the majority of old antibiotics are used off-label. In this article we review data about some novel and old antibiotics that are active against MDR-GN bacteria causing sepsis and are of interest to be used in the neonatal population.
Collapse
Affiliation(s)
- Angeliki Kontou
- 1st Department of Neonatology and Neonatal Intensive Care Unit, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki 54642, Greece
| | - Maria Kourti
- Infectious Diseases Unit, 3rd Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki 54642, Greece
| | - Elias Iosifidis
- Infectious Diseases Unit, 3rd Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki 54642, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki 54642, Greece
| | - Kosmas Sarafidis
- 1st Department of Neonatology and Neonatal Intensive Care Unit, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki 54642, Greece
| | - Emmanuel Roilides
- Infectious Diseases Unit, 3rd Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki 54642, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki 54642, Greece
| |
Collapse
|
3
|
Grafeneder J, Derhaschnig U, Eskandary F, Buchtele N, Sus N, Frank J, Jilma B, Schoergenhofer C. Micellar Curcumin: Pharmacokinetics and Effects on Inflammation Markers and PCSK-9 Concentrations in Healthy Subjects in a Double-Blind, Randomized, Active-Controlled, Crossover Trial. Mol Nutr Food Res 2022; 66:e2200139. [PMID: 36101515 PMCID: PMC9787856 DOI: 10.1002/mnfr.202200139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/23/2022] [Indexed: 12/30/2022]
Abstract
SCOPE Preclinical models have demonstrated the anti-inflammatory and lipid-lowering effects of curcumin. Innovative formulations have been developed to overcome the poor bioavailability of native curcumin. The study hypothesizes that the bioavailability of micellar curcumin is superior to native curcumin and investigates the potential anti-inflammatory and proprotein convertase subtilisin/kexin type 9 (PCSK9) concentration lowering effects. METHODS AND RESULTS In this double-blind, randomized, crossover trial, 15 healthy volunteers receive micellar or native curcumin (105 mg day-1 ) for 7 days with a ≥7 days washout period. Curcumin and metabolite concentrations are quantified by high-performance liquid chromatography with fluorescence detection (HPLC-FD), and pharmacokinetics are calculated. To analyze anti-inflammatory effects, blood samples (baseline, 2 h, 7 days) are stimulated with 50 ng mL-1 lipopolysaccharides (LPS). Interleukin (IL)-6, tumor-necrosis factor (TNF-α), and PCSK9 concentrations are quantified. Micellar curcumin demonstrates improved bioavailability (≈39-fold higher maximum concentrations, ≈14-fold higher area-under-the-time-concentration curve, p < 0.001) but does not reduce pro-inflammatory cytokines in the chosen model. Subjects receiving micellar curcumin have significantly lower PCSK9 concentrations (≈10% reduction) after 7 days compared to baseline (p = 0.038). CONCLUSION Micellar curcumin demonstrates an improved oral bioavailability but does not show anti-inflammatory effects in this model. Potential effects on PCSK9 concentrations warrant further investigation.
Collapse
Affiliation(s)
- Juergen Grafeneder
- Department of Emergency MedicineMedical University of ViennaVienna1090Austria
| | - Ulla Derhaschnig
- Department of Clinical PharmacologyMedical University of ViennaVienna1090Austria
| | - Farsad Eskandary
- Division of Nephrology, Department of Medicine IIIMedical University of ViennaVienna1090Austria
| | - Nina Buchtele
- Department of Medicine IMedical University of ViennaVienna1090Austria
| | - Nadine Sus
- Department of Food Biofunctionality (140b)Institute of Nutritional SciencesUniversity of HohenheimStuttgartGermany
| | - Jan Frank
- Department of Food Biofunctionality (140b)Institute of Nutritional SciencesUniversity of HohenheimStuttgartGermany
| | - Bernd Jilma
- Department of Clinical PharmacologyMedical University of ViennaVienna1090Austria
| | | |
Collapse
|
4
|
Tan P, Tang Q, Xu S, Zhang Y, Fu H, Ma X. Designing Self-Assembling Chimeric Peptide Nanoparticles with High Stability for Combating Piglet Bacterial Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105955. [PMID: 35285170 PMCID: PMC9109057 DOI: 10.1002/advs.202105955] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/22/2022] [Indexed: 05/14/2023]
Abstract
As a novel type of antibiotic alternative, peptide-based antibacterial drug shows potential application prospects attributable to their unique mechanism for lysing the membrane of pathogenic bacteria. However, peptide-based antibacterial drugs suffer from a series of problems, most notably their immature stability, which seriously hinders their application. In this study, self-assembling chimeric peptide nanoparticles (which offer excellent stability in the presence of proteases and salts) are constructed and applied to the treatment of bacterial infections. In vitro studies are used to demonstrate that peptide nanoparticles NPs1 and NPs2 offer broad-spectrum antibacterial activity and desirable biocompatibility, and they retain their antibacterial ability in physiological salt environments. Peptide nanoparticles NPs1 and NPs2 can resist degradation under high concentrations of proteases. In vivo studies illustrate that the toxicity caused by peptide nanoparticles NPs1 and NPs2 is negligible, and these nanoparticles can alleviate systemic bacterial infections in mice and piglets. The membrane permeation mechanism and interference with the cell cycle differ from that of antibiotics and mean that the nanoparticles are at a lower risk of inducing drug resistance. Collectively, these advances may accelerate the development of peptide-based antibacterial nanomaterials and can be applied to the construction of supramolecular nanomaterials.
Collapse
Affiliation(s)
- Peng Tan
- State Key Laboratory of Animal NutritionCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Qi Tang
- State Key Laboratory of Animal NutritionCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Shenrui Xu
- State Key Laboratory of Animal NutritionCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Yucheng Zhang
- State Key Laboratory of Animal NutritionCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Huiyang Fu
- State Key Laboratory of Animal NutritionCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Xi Ma
- State Key Laboratory of Animal NutritionCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| |
Collapse
|
5
|
Pharmacodynamic and immunomodulatory effects of polymyxin B in combination with fosfomycin against KPC-2-producing Klebsiella pneumoniae. Int J Antimicrob Agents 2022; 59:106566. [DOI: 10.1016/j.ijantimicag.2022.106566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 02/25/2022] [Accepted: 03/06/2022] [Indexed: 11/23/2022]
|
6
|
OUP accepted manuscript. J Antimicrob Chemother 2022; 77:1424-1431. [DOI: 10.1093/jac/dkac039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/14/2022] [Indexed: 11/13/2022] Open
|
7
|
Boehm T, Karer M, Matzneller P, Buchtele N, Ratzinger F, Petroczi K, Schoergenhofer C, Schwameis M, Burgmann H, Zeitlinger M, Jilma B. Human diamine oxidase is readily released from activated neutrophils ex vivo and in vivo but is rarely elevated in bacteremic patients. Int J Immunopathol Pharmacol 2021; 34:2058738420954945. [PMID: 32997559 PMCID: PMC7533923 DOI: 10.1177/2058738420954945] [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] [Indexed: 11/16/2022] Open
Abstract
During human diamine oxidase (DAO) ELISA development we noticed that in serum DAO concentrations appear to be higher when compared to plasma. Neutrophils contain DAO in the specific granules and we hypothesized that DAO is released from neutrophils during serum coagulation. If activation of neutrophils can release DAO, its concentrations might be elevated in vivo after lipopolysaccharide (LPS) administration and in bacteremic patients. Using blood from healthy volunteers DAO concentrations were measured ex vivo in serum, citrate, EDTA and heparin plasma over several hours and after activation of neutrophils. Lipopolysaccharide and granulocyte-colony stimulating factor (G-CSF) were administered to 15 and 8 healthy volunteers, respectively and DAO concentrations were measured at different timepoints. DAO antigen levels were also determined in three different subcohorts of patients with culture-proven bacteremia and high C-reactive protein (CRP) levels. DAO concentrations were elevated in a time-dependent manner in serum but not in EDTA or citrate plasma (P < 0.01). Neutrophil activation using phorbol myristate acetate (PMA) and zymosan dose-dependently caused DAO concentrations to be elevated more than 10-fold at both 22°C and 37°C (both P-values <0.001). Administration of LPS to healthy volunteers released DAO from neutrophils (P < 0.001). Of the 55 different bacteremic patients selected from three independent cohorts only 3 (5.4%) showed highly elevated DAO concentrations. Serum DAO concentrations do not accurately reflect circulating enzyme levels but coagulation-induced neutrophil activation and consequently DAO release. Only a few bacteremic patients show high DAO concentrations able to degrade histamine rapidly.
Collapse
Affiliation(s)
- Thomas Boehm
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Matthias Karer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Peter Matzneller
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Nina Buchtele
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Franz Ratzinger
- Division of Medical and Chemical Laboratory Diagnostics, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Karin Petroczi
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Michael Schwameis
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Heinz Burgmann
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
8
|
Wheatley R, Diaz Caballero J, Kapel N, de Winter FHR, Jangir P, Quinn A, Del Barrio-Tofiño E, López-Causapé C, Hedge J, Torrens G, Van der Schalk T, Xavier BB, Fernández-Cuenca F, Arenzana A, Recanatini C, Timbermont L, Sifakis F, Ruzin A, Ali O, Lammens C, Goossens H, Kluytmans J, Kumar-Singh S, Oliver A, Malhotra-Kumar S, MacLean C. Rapid evolution and host immunity drive the rise and fall of carbapenem resistance during an acute Pseudomonas aeruginosa infection. Nat Commun 2021; 12:2460. [PMID: 33911082 PMCID: PMC8080559 DOI: 10.1038/s41467-021-22814-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/31/2021] [Indexed: 02/08/2023] Open
Abstract
It is well established that antibiotic treatment selects for resistance, but the dynamics of this process during infections are poorly understood. Here we map the responses of Pseudomonas aeruginosa to treatment in high definition during a lung infection of a single ICU patient. Host immunity and antibiotic therapy with meropenem suppressed P. aeruginosa, but a second wave of infection emerged due to the growth of oprD and wbpM meropenem resistant mutants that evolved in situ. Selection then led to a loss of resistance by decreasing the prevalence of low fitness oprD mutants, increasing the frequency of high fitness mutants lacking the MexAB-OprM efflux pump, and decreasing the copy number of a multidrug resistance plasmid. Ultimately, host immunity suppressed wbpM mutants with high meropenem resistance and fitness. Our study highlights how natural selection and host immunity interact to drive both the rapid rise, and fall, of resistance during infection.
Collapse
Affiliation(s)
| | | | - Natalia Kapel
- University of Oxford, Department of Zoology, Oxford, UK
| | - Fien H R de Winter
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | - Pramod Jangir
- University of Oxford, Department of Zoology, Oxford, UK
| | - Angus Quinn
- University of Oxford, Department of Zoology, Oxford, UK
| | | | | | - Jessica Hedge
- University of Oxford, Department of Zoology, Oxford, UK
| | - Gabriel Torrens
- Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - Thomas Van der Schalk
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | - Basil Britto Xavier
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | | | - Angel Arenzana
- Departamento de Medicina, Universidad de Sevilla, Seville, Spain
| | - Claudia Recanatini
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Leen Timbermont
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | | | - Alexey Ruzin
- Microbial Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Omar Ali
- Microbial Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
- Viela Bio, Gaithersburg, MD, USA
| | - Christine Lammens
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | - Herman Goossens
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | - Jan Kluytmans
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Microvida Laboratory for Medical Microbiology and Department of Infection Control, Amphia Hospital, Breda, The Netherlands
| | - Samir Kumar-Singh
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Molecular Pathology Group, Faculty of Medicine-Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Belgium
| | - Antonio Oliver
- Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | - Craig MacLean
- University of Oxford, Department of Zoology, Oxford, UK.
| |
Collapse
|
9
|
Sheikh Z, Bradbury P, Reekie TA, Pozzoli M, Robinson PD, Kassiou M, Young PM, Ong HX, Traini D. Tobramycin and Colistin display anti-inflammatory properties in CuFi-1 cystic fibrosis cell line. Eur J Pharmacol 2021; 902:174098. [PMID: 33848541 DOI: 10.1016/j.ejphar.2021.174098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/01/2021] [Accepted: 04/07/2021] [Indexed: 10/21/2022]
Abstract
Current cystic fibrosis (CF) treatment strategies are primarily focused on oral/inhaled anti-inflammatories and antibiotics, resulting in a considerable treatment burden for CF patients. Therefore, combination treatments consisting of anti-inflammatories with antibiotics could reduce the CF treatment burden. However, there is an imperative need to understand the potential drug-drug interactions of these combination treatments to determine their efficacy. Thus, this study aimed to determine the interactions of the anti-inflammatory agent Ibuprofen with each of the CF-approved inhaled antibiotics (Tobramycin, Colistin and its prodrug colistimethate sodium/Tadim) and anti-bacterial and anti-inflammatory efficacy. Chemical interactions of the Ibuprofen:antibiotic combinations were elucidated using High-Resolution Mass-Spectrometry (HRMS) and 1H NMR. HRMS showed pairing of Ibuprofen and Tobramycin, further confirmed by 1H NMR whilst no pairing was observed for either Ibuprofen:Colistin or Ibuprofen:Tadim combinations. The anti-bacterial activity of the combinations against Pseudomonas aeruginosa showed that neither paired nor non-paired Ibuprofen:antibiotic therapies altered the anti-bacterial activity. The anti-inflammatory efficacy of the combination therapies was next determined at two different concentrations (Low and High) using in vitro models of NuLi-1 (healthy) and CuFi-1 (CF) cell lines. Differential response in the anti-inflammatory efficacy of Ibuprofen:Tobramycin combination was observed between the two concentrations due to changes in the structural conformation of the paired Ibuprofen:Tobramycin complex at High concentration, confirmed by 1H NMR. In contrast, the non-pairing of the Ibuprofen:Colistin and Ibuprofen:Tadim combinations showed a significant decrease in IL-8 secretion at both the concentrations. Importantly, all antibiotics alone showed anti-inflammatory properties, highlighting the inherent anti-inflammatory properties of these antibiotics.
Collapse
Affiliation(s)
- Zara Sheikh
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Peta Bradbury
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Australia; Discipline of Medicine, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Tristan A Reekie
- Research School of Chemistry, Australian National University, Canberra, Australia
| | - Michele Pozzoli
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Australia
| | - Paul D Robinson
- Department of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, Australia
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, Sydney, Australia
| | - Paul M Young
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Hui Xin Ong
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| | - Daniela Traini
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| |
Collapse
|
10
|
Zou D, Yu H, Li F. The Difference Between Polymyxin B and Polymyxin E in Causing Skin Hyerpigmentation. Front Pharmacol 2021; 12:647564. [PMID: 33935746 PMCID: PMC8087172 DOI: 10.3389/fphar.2021.647564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/24/2021] [Indexed: 11/17/2022] Open
Affiliation(s)
- Dongna Zou
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Haitao Yu
- Department of Pharmacy, Zao Zhuang Municipal Hospital, Zao Zhuang, China
| | - Feifei Li
- Department of Infection, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
11
|
Resl M, Heinzl MW, Klammer C, Egger M, Feldbauer R, Pohlhammer J, Dieplinger B, Clodi M. Storm of Cardiovascular Markers After LPS Administration in Human Volunteers. J Cardiovasc Transl Res 2021; 14:941-947. [PMID: 33751402 PMCID: PMC7983963 DOI: 10.1007/s12265-021-10109-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 02/15/2021] [Indexed: 12/01/2022]
Abstract
Acute infections are associated with an elevated cardiovascular risk. However, little is known about the interactions of acute inflammatory responses and the cardiovascular system. We therefore aimed to evaluate effects of acute inflammatory stimuli mediated by LPS administration on a set of 89 cardiovascular biomarkers. A single-blinded, placebo-controlled cross-over study using the human endotoxin model was performed. Ten healthy men were administered lipopolysaccharide (LPS) or placebo on two different study days after an overnight fast. Eighty-nine different cardiovascular biomarkers were measured repetitively over 48 h. Out of 89 cardiovascular biomarkers, 54 markers were significantly influenced by LPS infusion. The observed biomarker response to inflammation was more pronounced and complex than anticipated. In conclusion, our data show that the cardiovascular system is under enormous distress in response to experimental low-dose inflammation in humans, as demonstrated by a significant effect on 54 of the 89 biomarkers tested.
Collapse
Affiliation(s)
- Michael Resl
- Department of Medicine, St. John of God Hospital Linz, Seilerstaette 2, 4021, Linz, Austria
| | | | - Carmen Klammer
- Department of Medicine, St. John of God Hospital Linz, Seilerstaette 2, 4021, Linz, Austria
| | - Margot Egger
- Department of Laboratory Medicine, St. John of God Hospital Linz, ICMR - Institute for Cardiovascular and Metabolic Research, JKU Linz, Linz, Austria
| | - Roland Feldbauer
- Department of Medicine, St. John of God Hospital Linz, Seilerstaette 2, 4021, Linz, Austria
| | - Johannes Pohlhammer
- Department of Medicine, St. John of God Hospital Linz, Seilerstaette 2, 4021, Linz, Austria
| | - Benjamin Dieplinger
- Department of Laboratory Medicine, St. John of God Hospital Linz, ICMR - Institute for Cardiovascular and Metabolic Research, JKU Linz, Linz, Austria
| | - Martin Clodi
- Department of Medicine, St. John of God Hospital Linz, Seilerstaette 2, 4021, Linz, Austria.
| |
Collapse
|
12
|
Wicha SG, Märtson AG, Nielsen EI, Koch BCP, Friberg LE, Alffenaar JW, Minichmayr IK. From Therapeutic Drug Monitoring to Model-Informed Precision Dosing for Antibiotics. Clin Pharmacol Ther 2021; 109:928-941. [PMID: 33565627 DOI: 10.1002/cpt.2202] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/01/2021] [Indexed: 12/14/2022]
Abstract
Therapeutic drug monitoring (TDM) and model-informed precision dosing (MIPD) have evolved as important tools to inform rational dosing of antibiotics in individual patients with infections. In particular, critically ill patients display altered, highly variable pharmacokinetics and often suffer from infections caused by less susceptible bacteria. Consequently, TDM has been used to individualize dosing in this patient group for many years. More recently, there has been increasing research on the use of MIPD software to streamline the TDM process, which can increase the flexibility and precision of dose individualization but also requires adequate model validation and re-evaluation of existing workflows. In parallel, new minimally invasive and noninvasive technologies such as microneedle-based sensors are being developed, which-together with MIPD software-have the potential to revolutionize how patients are dosed with antibiotics. Nonetheless, carefully designed clinical trials to evaluate the benefit of TDM and MIPD approaches are still sparse, but are critically needed to justify the implementation of TDM and MIPD in clinical practice. The present review summarizes the clinical pharmacology of antibiotics, conventional TDM and MIPD approaches, and evidence of the value of TDM/MIPD for aminoglycosides, beta-lactams, glycopeptides, and linezolid, for which precision dosing approaches have been recommended.
Collapse
Affiliation(s)
- Sebastian G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Anne-Grete Märtson
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Jan-Willem Alffenaar
- Faculty of Medicine and Health, Sydney Pharmacy School, University of Sydney, Camperdown, Australia.,Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia.,Westmead Hospital, Wentworthville, Australia
| | | | | |
Collapse
|
13
|
Shao C, Zhu Y, Jian Q, Lai Z, Tan P, Li G, Shan A. Cross-Strand Interaction, Central Bending, and Sequence Pattern Act as Biomodulators of Simplified β-Hairpin Antimicrobial Amphiphiles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2003899. [PMID: 33354914 DOI: 10.1002/smll.202003899] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/19/2020] [Indexed: 06/12/2023]
Abstract
Novel antimicrobial peptides (AMPs) have revolutionarily evolved into formidable candidates for antibiotic substitute materials against pathogenic infections. However, cost, lability, disorderly sequences, systemic toxicology, and biological profiles have plagued the perennial search. Here, a progressive β-hairpin solution with the simplest formulation is implanted into an AMP-based therapeutic strategy to systematically reveal the complex balance between function and toxicity of structural moieties, including cationicity, hydrophobicity, cross-strand interactions, center bending, and sequence pattern. Comprehensive implementation of structural identification, ten microorganisms, eleven in vitro barriers, four mammalian cells, and a diversified membrane operation setup led to the emergence of β-hairpin prototypes from a 24-member library. Lead amphiphiles, WKF-PG and WRF-NG, can tackle bacterial infection through direct antimicrobial efficacy and potential inflammation-limiting capabilities, such as an Escherichia coli challenge in a mouse peritonitis-sepsis model, without observed toxicity after systemic administration. Their optimal states with dissimilar modulators and the unavailable drug resistance related to membrane lytic mechanisms, also provide an usher for renewed innovation among β-sheet peptide-based antimicrobial biomaterials.
Collapse
Affiliation(s)
- Changxuan Shao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Yongjie Zhu
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Qiao Jian
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Zhenheng Lai
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Peng Tan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Guoyu Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| |
Collapse
|
14
|
Niu H, Yang T, Wang J, Wang R, Cai Y. Immunomodulatory Effect of Colistin and its Protective Role in Rats with Methicillin-Resistant Staphylococcus aureus-induced Pneumonia. Front Pharmacol 2021; 11:602054. [PMID: 33551807 PMCID: PMC7854386 DOI: 10.3389/fphar.2020.602054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/10/2020] [Indexed: 11/13/2022] Open
Abstract
Objectives: Colistin is the last resort of antimicrobials against multi-drug resistant Gram-negative pathogens. Previous studies in Caenorhabditis elegans and macrophages of rats have suggested that colistin possesses the immunomodulatory properties by acting p38/MAPK pathway. Here, we aimed to confirm the immunomodulatory role of colistin in animal models. Methods: Rat model of Methicillin-resistant Staphylococcus aureus (MRSA)-induced pneumonia was established. Plasma concentrations of proinflammatory cytokines, quantitative bacteriology, histology and immunohistochemistry of lungs were assessed to compare the immunomodulatory properties of colistin pre-administration. Results: The numbers of white blood cells and granulocytes were significantly increased in the 9 mg/kg colistin pre-administration group at 72 h after infection. Levels of TNF-α, IL-6 and IL-1β in plasma after colistin pre-administration were lower compared with the infected group without treatment. Colistin pre-treatment resulted in lower bacterial counts, a dramatic decrease of cytokines and improved histopathological injury in infected lung tissues compared with the untreated animals. However, p38/MAPK inhibitor SB203580 did not fully block the above-mentioned effects caused by colistin. Conclusion: Pre-administration of colistin could attenuate an excessive inflammatory reaction and protect the lungs from MRSA-associated damages. However, these effects could not be reversed by blocking the p38/MAPK pathway alone. Collectively, the mechanism underlying the immunoregulatory effects of colistin in mammals needs to be further explored.
Collapse
Affiliation(s)
- Hui Niu
- Department of Pharmacy, Center of Medicine Clinical Research, Medical Supplies Center of PLA General Hospital, Beijing, China
| | - Tianli Yang
- Department of Pharmacy, Center of Medicine Clinical Research, Medical Supplies Center of PLA General Hospital, Beijing, China
| | - Jin Wang
- Department of Pharmacy, Center of Medicine Clinical Research, Medical Supplies Center of PLA General Hospital, Beijing, China
| | - Rui Wang
- Department of Pharmacy, Center of Medicine Clinical Research, Medical Supplies Center of PLA General Hospital, Beijing, China
| | - Yun Cai
- Department of Pharmacy, Center of Medicine Clinical Research, Medical Supplies Center of PLA General Hospital, Beijing, China
| |
Collapse
|
15
|
Dolmatova EV, Wang K, Mandavilli R, Griendling KK. The effects of sepsis on endothelium and clinical implications. Cardiovasc Res 2021; 117:60-73. [PMID: 32215570 PMCID: PMC7810126 DOI: 10.1093/cvr/cvaa070] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/03/2020] [Accepted: 03/20/2020] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT Sepsis accounts for nearly 700 000 deaths in Europe annually and is caused by an overwhelming host response to infection resulting in organ failure. The endothelium is an active contributor to sepsis and as such represents a major target for therapy. During sepsis, endothelial cells amplify the immune response and activate the coagulation system. They are both a target and source of inflammation and serve as a link between local and systemic immune responses. In response to cytokines produced by immune cells, the endothelium expresses adhesion molecules and produces vasoactive compounds, inflammatory cytokines, and chemoattractants, thus switching from an anticoagulant to procoagulant state. These responses contribute to local control of infection, but systemic activation can lead to microvascular thrombosis, capillary permeability, hypotension, tissue hypoxia, and ultimately tissue damage. This review focuses on the role of the endothelium in leucocyte adhesion and transmigration as well as production of reactive oxygen and nitrogen species, microRNAs and cytokines, formation of signalling microparticles, and disseminated intravascular coagulation. We also discuss alterations in endothelial permeability and apoptosis. Finally, we review the diagnostic potential of endothelial markers and endothelial pathways as therapeutic targets for this devastating disease.
Collapse
Affiliation(s)
- Elena V Dolmatova
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Keke Wang
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Rohan Mandavilli
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| |
Collapse
|
16
|
Ubagai T, Sato Y, Kamoshida G, Unno Y, Ono Y. Immunomodulatory gene expression analysis in LPS-stimulated human polymorphonuclear leukocytes treated with antibiotics commonly used for multidrug-resistant strains. Mol Immunol 2020; 129:39-44. [PMID: 33271353 DOI: 10.1016/j.molimm.2020.11.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 10/30/2020] [Accepted: 11/17/2020] [Indexed: 11/17/2022]
Abstract
Conventional antibiotics used for the treatment of severe infections such as sepsis and septic shock confer immunomodulatory benefits. However, the growing problem of multidrug resistant infections has led to an increase in the administration of non-conventional last-resort antibiotics, including quinolones, aminoglycosides, and polypeptides, and the effects of these drugs on immunomodulatory gene expression in activated human polymorphonuclear leukocytes (PMNs) have not been reported. In this study, lipopolysaccharide-stimulated PMNs were incubated with piperacillin, rifampicin, fosfomycin (FOM), levofloxacin (LVFX), minocycline (MINO), colistin, tigecycline, or amikacin, and the mRNA expression levels of pattern recognition receptors (TLR2, TLR4, and CD14), inflammatory cytokines (TNFα and IL6), and chemokine receptors (IL8Rs and ITGAM) in these cells were quantitated using real-time qPCR. Many of the tested antibiotics altered the expression of the investigated cytokines. Notably, FOM, LVFX, and MINO significantly downregulated the expression of IL6, which is associated with pro- and anti-inflammatory defense mechanisms. Treatment of FOM and LVFX reduced IL-6 production as well as observed for IL6 gene expression. These findings indicated transcription and translation cooperation under the used experimental conditions. Therefore, our findings suggest that administration of these antibiotics suppresses the host anti-inflammatory response.
Collapse
Affiliation(s)
- Tsuneyuki Ubagai
- Department of Microbiology & Immunology, Teikyo University School of Medicine Tokyo, 173-8605, Japan.
| | - Yoshinori Sato
- Department of Microbiology & Immunology, Teikyo University School of Medicine Tokyo, 173-8605, Japan
| | - Go Kamoshida
- Department of Microbiology & Immunology, Teikyo University School of Medicine Tokyo, 173-8605, Japan
| | - Yuka Unno
- Department of Microbiology & Immunology, Teikyo University School of Medicine Tokyo, 173-8605, Japan
| | - Yasuo Ono
- Department of Microbiology & Immunology, Teikyo University School of Medicine Tokyo, 173-8605, Japan
| |
Collapse
|
17
|
Immunomodulatory effects of colistin on host responses against carbapenem-resistant Klebsiella pneumoniae biofilms. Int J Antimicrob Agents 2020; 56:106182. [PMID: 33045355 DOI: 10.1016/j.ijantimicag.2020.106182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 07/04/2020] [Accepted: 10/03/2020] [Indexed: 12/17/2022]
Abstract
Colistin (CST) is a last-resort therapeutic option for carbapenem-resistant Klebsiella pneumoniae (CR-Kp) infections in critically ill patients. The effect of subinhibitory CST concentrations (sub-MICs) on biofilm formation is organism-dependent. We investigated the interactions between CST and innate immune cells against CR-Kp biofilms (CR-KpBF) by studying the effect of biofilm sub-MICs of CST on (i) damage induced by human polymorphonuclear neutrophils (PMNs) on CR-KpBF and (ii) the immunomodulatory potential on human mononuclear cells (MNCs) exposed to CR-KpBF. The impact of CST on PMN-induced biofilm damage was assessed by XTT reduction assay. Signal transduction and gene expression profiles in response to CST sub-MICs of MNCs exposed to CR-KpBF were studied by RT-PCR and multiplex ELISA. Pre-exposure of CR-Kp to 0.06 mg/L CST led to subsequent increased PMN-mediated biofilm damage against CR-KpBF in the presence of CST biofilm sub-MICs: there was an additive effect at 2, 4, 8 and 16 mg/L. However, the overall biofilm damage was not >52%. MNCs responded to CR-KpBF through Toll-like receptor 2 (TLR2) by 2.5-fold upregulation and NLRP3 inflammasome activation. CR-KpBF stimulated increased production of interleukin 1-beta (IL-1β), tumour necrosis factor-alpha (TNFα), IL-8 and IL-6. In the combination treatment, 0.5 mg/L CST reduced IL-1β, TNFα and IL-8 levels, whereas at 2 mg/L and 8 mg/L it increased the anti-inflammatory cytokine IL-10 (P < 0.05). Biofilm sub-MICs of CST enhance PMN killing capacity and attenuate production of inflammatory cytokines by MNCs exposed to CR-KpBF, playing a potentially important immunotherapeutic role especially for patients with cytokine deregulation.
Collapse
|
18
|
Krishnan M, Choi J, Jang A, Kim Y. A Novel Peptide Antibiotic, Pro10-1D, Designed from Insect Defensin Shows Antibacterial and Anti-Inflammatory Activities in Sepsis Models. Int J Mol Sci 2020; 21:ijms21176216. [PMID: 32867384 PMCID: PMC7504360 DOI: 10.3390/ijms21176216] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/19/2020] [Accepted: 08/25/2020] [Indexed: 01/28/2023] Open
Abstract
Owing to the challenges faced by conventional therapeutics, novel peptide antibiotics against multidrug-resistant (MDR) gram-negative bacteria need to be urgently developed. We had previously designed Pro9-3 and Pro9-3D from the defensin of beetle Protaetia brevitarsis; they showed high antimicrobial activity with cytotoxicity. Here, we aimed to develop peptide antibiotics with bacterial cell selectivity and potent antibacterial activity against gram-negative bacteria. We designed 10-meric peptides with increased cationicity by adding Arg to the N-terminus of Pro9-3 (Pro10-1) and its D-enantiomeric alteration (Pro10-1D). Among all tested peptides, the newly designed Pro10-1D showed the strongest antibacterial activity against Escherichia coli, Acinetobacter baumannii, and MDR strains with resistance against protease digestion. Pro10-1D can act as a novel potent peptide antibiotic owing to its outstanding inhibitory activities against bacterial film formation with high bacterial cell selectivity. Dye leakage and scanning electron microscopy revealed that Pro10-1D targets the bacterial membrane. Pro10-1D inhibited inflammation via Toll Like Receptor 4 (TLR4)/Nuclear factor-κB (NF-κB) signaling pathways in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. Furthermore, Pro10-1D ameliorated multiple-organ damage and attenuated systemic infection-associated inflammation in an E. coli K1-induced sepsis mouse model. Overall, our results suggest that Pro10-1D can potentially serve as a novel peptide antibiotic for the treatment of gram-negative sepsis.
Collapse
Affiliation(s)
| | | | | | - Yangmee Kim
- Correspondence: ; Tel.: +82-2-450-3421; Fax: +82-2-447-5987
| |
Collapse
|
19
|
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: 0.3] [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
|
20
|
Cigana C, Ranucci S, Rossi A, De Fino I, Melessike M, Bragonzi A. Antibiotic efficacy varies based on the infection model and treatment regimen for Pseudomonas aeruginosa. Eur Respir J 2020; 55:13993003.02456-2018. [PMID: 31624114 PMCID: PMC7057181 DOI: 10.1183/13993003.02456-2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 10/10/2019] [Indexed: 12/14/2022]
Abstract
Antibiotic discovery and preclinical testing are needed to combat the Pseudomonas aeruginosa health threat. Most frequently, antibiotic efficacy is tested in models of acute respiratory infection, with chronic pneumonia remaining largely unexplored. This approach generates serious concerns about the evaluation of treatment for chronically infected patients, and highlights the need for animal models that mimic the course of human disease. In this study, the efficacy of the marketed antibacterial drugs tobramycin (TOB) and colistin (COL) was tested in murine models of acute and chronic P. aeruginosa pulmonary infection. Different administration routes (intranasal, aerosol or subcutaneous) and treatment schedules (soon or 7 days post-infection) were tested. In the acute infection model, aerosol and subcutaneous administration of TOB reduced the bacterial burden and inflammatory response, while intranasal treatment showed modest efficacy. COL reduced the bacterial burden less effectively but dampened inflammation. Mice treated soon after chronic infection for 7 days with daily aerosol or subcutaneous administration of TOB showed higher and more rapid body weight recovery and reduced bacterial burden and inflammation than vehicle-treated mice. COL-treated mice showed no improvement in body weight or change in inflammation. Modest bacterial burden reduction was recorded only with aerosol COL administration. When treatment for chronic infection was commenced 7 days after infection, both TOB and COL failed to reduce P. aeruginosa burden and inflammation, or aid in recovery of body weight. Our findings suggest that the animal model and treatment regimen should be carefully chosen based on the type of infection to assess antibiotic efficacy. Disease-specific animal models and treatment regimens are essential in order to optimise anti-Pseudomonas drug testinghttp://bit.ly/2ISfBiB
Collapse
Affiliation(s)
- Cristina Cigana
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Both authors contributed equally and share senior authorship
| | - Serena Ranucci
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Alice Rossi
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Ida De Fino
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Medede Melessike
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Alessandra Bragonzi
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy .,Both authors contributed equally and share senior authorship
| |
Collapse
|
21
|
Schoergenhofer C, Matzneller P, Mühlbacher J, Hell L, Zeitlinger M, Jilma B. PCSK9 decreases during experimental endotoxemia. J Intern Med 2020; 287:333-335. [PMID: 31660669 DOI: 10.1111/joim.13003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/16/2019] [Accepted: 10/24/2019] [Indexed: 11/30/2022]
Affiliation(s)
- C Schoergenhofer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - P Matzneller
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - J Mühlbacher
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - L Hell
- Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - M Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - B Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
22
|
Wang J, Shao W, Niu H, Yang T, Wang Y, Cai Y. Immunomodulatory Effects of Colistin on Macrophages in Rats by Activating the p38/MAPK Pathway. Front Pharmacol 2019; 10:729. [PMID: 31297059 PMCID: PMC6606943 DOI: 10.3389/fphar.2019.00729] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/05/2019] [Indexed: 01/25/2023] Open
Abstract
Objectives: Colistin has been identified in a Caenorhabditis elegans chemical screening as an immunostimulatory agent that activates the conserved p38/PMK-1 pathway and provides protection against pathogens. Here we aimed to extend those findings to a mammalian model and evaluate the immunomodulatory effects of colistin on rat macrophages. Methods: Macrophages were isolated from Sprague-Dawley (SD) rat. The effects of colistin on the cytokine secretion, phagocytic activity and protein expression were determined by enzyme-linked immunosorbent assay (ELISA), flow cytometry, and Western blotting analysis, respectively. The relative microRNA expression was determined by microarray, and Kyoto Encylopedia of Genes and Genomes (KEGG) was used to identify potential signaling pathways. Results: Our data showed that 5, 10, and 20 µg/ml colistin significantly increased the secretion of TNF-α, while 20 and 5 µg/ml colistin significantly increased the levels of IL-1β and IL-6, respectively. Flow cytometry results showed that the relative mean fluorescence intensity and percentage of phagocytosis in colistin treatment groups were significantly higher compared with the control group, while the increased phagocytosis phenomenon can be blocked by p38 inhibitor. The phagocytic ability of macrophages against Staphylococcus aureus was significantly increased after colistin treatment. Microarray and KEGG pathway analyses revealed that mitogen-activated protein kinase (MAPK), mammalian target of rapamycin (mTOR), chemokine, and B cell receptor were the main pathways involved in the colistin stimulation process. Western blotting analysis demonstrated that the phosphorylated p38 protein level of colistin treatment groups was increased in a dose dependent manner. Conclusions: Present study is the first to demonstrate that colistin had immunomodulatory effects on macrophages in mammals, and the p38/MAPK pathway was involved in such colistin-induced immunomodulatory effect.
Collapse
Affiliation(s)
- Jin Wang
- Center of Medicine Clinical Research, Department of Pharmacy, PLA General Hospital, Beijing, China
| | - Weili Shao
- Center of Medicine Clinical Research, Department of Pharmacy, PLA General Hospital, Beijing, China
| | - Hui Niu
- Center of Medicine Clinical Research, Department of Pharmacy, PLA General Hospital, Beijing, China
| | - Tianli Yang
- Center of Medicine Clinical Research, Department of Pharmacy, PLA General Hospital, Beijing, China
| | - Yuning Wang
- Clinical Surgery Division, PLA General Hospital, Beijing, China
| | - Yun Cai
- Center of Medicine Clinical Research, Department of Pharmacy, PLA General Hospital, Beijing, China
| |
Collapse
|
23
|
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: 7.8] [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
|
24
|
Avendaño-Ortiz J, Llanos-González E, Toledano V, Del Campo R, Cubillos-Zapata C, Lozano-Rodríguez R, Ismail A, Prados C, Gómez-Campelo P, Aguirre LA, García-Río F, López-Collazo E. Pseudomonas aeruginosa colonization causes PD-L1 overexpression on monocytes, impairing the adaptive immune response in patients with cystic fibrosis. J Cyst Fibros 2018; 18:630-635. [PMID: 30442491 DOI: 10.1016/j.jcf.2018.11.002] [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: 07/18/2018] [Revised: 10/29/2018] [Accepted: 11/04/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND Cystic fibrosis (CF) is an endotoxin tolerance (ET)-related disease. Given that increased PD-L1 has been reported in ET, its expression and physiological effects on cystic fibrosis monocytes should be studied. METHODS We analyzed the phenotype and ex vivo response of immune system cells in 32 patients with CF, 19 of them colonized by Pseudomonas aeruginosa. An in vitro model was developed of Pseudomonas aeruginosa colonization using purified lipopolysaccharides (LPS) from one of the most prevalent strains in patients with CF (a CF-adapted Pseudomonas aeruginosa ST395 clone). Changes in the immune response, including cytokine production and T-lymphocyte proliferation, as well as expression of PD-L1, were evaluated. RESULTS PD-L1 was overexpressed in the monocytes of patients with CF compared with healthy volunteers, and levels of this immune checkpoint were associated with Pseudomonas aeruginosa colonization. In addition, patients with Pseudomonas aeruginosa colonization showed a patent ET status, including poor inflammatory response, reduced HLA-DR expression and T-lymphocyte proliferation impairment. PD-L1/PD-1 blocking assays reverted the impaired adaptive response. Ultimately, monocytes from healthy volunteers cultured in the presence of the clinically relevant strain of Pseudomonas aeruginosa or serum collected from patients with CF colonized by Pseudomonas aeruginosa reproduced the previous observed features. CONCLUSIONS Pseudomonas aeruginosa colonization in patients with CF was associated with PD-L1 overexpression and impaired T cell response, and LPS from this pathogen induced the observed phenotype. Our findings open new avenues for the use of anti-PD-1/PD-L1 immunotherapy in patients with CF who are colonized by Pseudomonas aeruginosa.
Collapse
Affiliation(s)
- José Avendaño-Ortiz
- Innate Immunity Group, IdiPAZ, La Paz University Hospital, Madrid, Spain; Tumor Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain; Center for Biomedical Research Network, CIBEres, Spain
| | - Emilio Llanos-González
- Innate Immunity Group, IdiPAZ, La Paz University Hospital, Madrid, Spain; Tumor Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Víctor Toledano
- Innate Immunity Group, IdiPAZ, La Paz University Hospital, Madrid, Spain; Tumor Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain; Center for Biomedical Research Network, CIBEres, Spain
| | - Rosa Del Campo
- Department of Microbiology, University Hospital Ramón y Cajal and IRYCIS, Madrid, Spain
| | - Carolina Cubillos-Zapata
- Center for Biomedical Research Network, CIBEres, Spain; Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Roberto Lozano-Rodríguez
- Innate Immunity Group, IdiPAZ, La Paz University Hospital, Madrid, Spain; Tumor Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Ahmad Ismail
- Innate Immunity Group, IdiPAZ, La Paz University Hospital, Madrid, Spain; Tumor Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Concepción Prados
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Paloma Gómez-Campelo
- Innate Immunity Group, IdiPAZ, La Paz University Hospital, Madrid, Spain; Tumor Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Luis A Aguirre
- Innate Immunity Group, IdiPAZ, La Paz University Hospital, Madrid, Spain; Tumor Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Francisco García-Río
- Center for Biomedical Research Network, CIBEres, Spain; Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Eduardo López-Collazo
- Innate Immunity Group, IdiPAZ, La Paz University Hospital, Madrid, Spain; Tumor Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain; Center for Biomedical Research Network, CIBEres, Spain.
| |
Collapse
|
25
|
Ostorhazi E, Hoffmann R, Herth N, Wade JD, Kraus CN, Otvos L. Advantage of a Narrow Spectrum Host Defense (Antimicrobial) Peptide Over a Broad Spectrum Analog in Preclinical Drug Development. Front Chem 2018; 6:359. [PMID: 30186829 PMCID: PMC6111444 DOI: 10.3389/fchem.2018.00359] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 07/30/2018] [Indexed: 12/27/2022] Open
Abstract
The APO-type proline-arginine-rich host defense peptides exhibit potent in vitro killing parameters against Enterobacteriaceae but not to other bacteria. Because of the excellent in vivo properties against systemic and local infections, attempts are regularly made to further improve the activity spectrum. A C-terminal hydrazide analog of the Chex1-Arg20 amide (ARV-1502) shows somewhat improved minimal inhibitory concentration against Moraxellaceae. Here we compared the activity of the two peptides as well as an inactive dimeric reverse amide analog in a systemic Acinetobacter baumannii infection. Only the narrow spectrum amide derivative reduced the 6-h blood bacterial burden by >2 log10 units reaching statistical significance (p = 0.03 at 5 mg/kg and 0.031 at 2 mg/kg administered intramuscularly). The hydrazide derivative, probably due to stronger activity on cell membranes, lysed erythrocytes at lower concentrations, and caused toxic effects at lower doses (10 mg/kg vs. 25 mg/kg). In a limited study, the amide induced a >5-fold production of the anti-inflammatory cytokine IL-10 over untreated naïve mice and minor increases in the anti-inflammatory IL-4 and pro-inflammatory cytokines TNF-α and IL-6, in blood. The blood of hydrazide-treated mice exhibited significantly lowered levels of IL-10 and slightly decreased IL-4 and TNF-α. These results suggest that the improved efficacy of the narrow-spectrum amide analog is likely associated with increased anti-inflammatory cytokine production and better stimulation of the immune system. Although blood IL-6 and TNF-α levels are frequently used as markers of potential toxicity in drug development, we did not observe any notable increase in mice receiving the toxic polyamide antibiotic colistin.
Collapse
Affiliation(s)
- Eszter Ostorhazi
- Institute of Medical Microbiology, Semmelweis University, Budapest, Hungary
| | - Ralf Hoffmann
- Institute of Bioanalytical Chemistry, Leipzig University, Leipzig, Germany
| | - Nicole Herth
- Institute of Bioanalytical Chemistry, Leipzig University, Leipzig, Germany
| | - John D Wade
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia.,School of Chemistry, University of Melbourne, Melbourne, VIC, Australia
| | | | - Laszlo Otvos
- Institute of Medical Microbiology, Semmelweis University, Budapest, Hungary.,Arrevus, Inc, Raleigh, NC, United States.,OLPE, LLC, Audubon, PA, United States
| |
Collapse
|
26
|
Wang L, Zhao H, Wang D. Inflammatory cytokine expression in patients with sepsis at an intensive care unit. Exp Ther Med 2018; 16:2126-2131. [PMID: 30186449 DOI: 10.3892/etm.2018.6376] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 01/18/2018] [Indexed: 12/24/2022] Open
Abstract
Sepsis is a systemic inflammatory response syndrome caused by infection of bacteria, fungi and/or viruses in clinical patients. It is known that inflammatory cytokine levels have an essential role in the progression of sepsis. The present study investigated the role of inflammatory markers in human peripheral blood mononuclear cells (hPBMCs) of patients with sepsis at an intensive care unit. In addition, the plasma levels of inflammatory cytokines were compared between sepsis patients and healthy individuals. The results demonstrated that the serum levels of interleukin-1, -17 and -6, as well as tumor necrosis factor-α, were upregulated in sepsis patients. The serum levels of high mobility group box 1 and C-reactive protein were increased in sepsis patients compared with those in healthy individuals. The expression levels of nuclear factor-κB-p65 and its inhibitor IκBα, as well as the ratio of CD25+ cells, and the levels of neutrophil gelatinase-associated lipocalin and peptidoglycan recognition protein were higher in hPBMCs in sepsis patients compared with those in healthy individuals. It was also indicated that balance of T helper type 1/2 cytokines was also disturbed in patients with sepsis compared with that in healthy individuals. In conclusion, these results indicated that inflammation is involved in the progression of sepsis by interfering with the expression of various molecules, suggesting a potential therapeutic strategy for the treatment of sepsis patients.
Collapse
Affiliation(s)
- Lili Wang
- Intensive Care Unit, Daqing Oil Field General Hospital, Daqing, Heilongjiang 163001, P.R. China
| | - Hongyan Zhao
- Intensive Care Unit, Daqing Oil Field General Hospital, Daqing, Heilongjiang 163001, P.R. China
| | - Dongxu Wang
- Intensive Care Unit, Daqing Oil Field General Hospital, Daqing, Heilongjiang 163001, P.R. China
| |
Collapse
|
27
|
Lee S, Ghosh P, Kwon H, Park SS, Kim GL, Choi SY, Kim EH, Tran TDH, Seon SH, Le NT, Iqbal H, Lee S, Pyo S, Rhee DK. Induction of the pneumococcal vncRS operon by lactoferrin is essential for pneumonia. Virulence 2018; 9:1562-1575. [PMID: 30246592 PMCID: PMC6177237 DOI: 10.1080/21505594.2018.1526529] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/12/2018] [Indexed: 12/29/2022] Open
Abstract
Streptococcus pneumoniae (pneumococcus), the major pathogen for pneumonia, commonly colonizes the lung, but the mechanism underlying the coordination of virulence factors during invasion via the host protein remains poorly understood. Bacterial lysis releases the components of the cell wall, and triggers innate immunity and the subsequent secretion of pro-inflammatory cytokines. Previously, the virulence of the pep27 mutant was shown to be attenuated as a feasible candidate for vaccine development. However, the role of pep27 gene, belonging to the vancomycin-resistance locus (vncRS operon), in virulence, is largely unknown. This study demonstrates that transferrin in the host serum reduces the survival of the host during S. pneumoniae infections in mice. The exposure of the pneumococcal D39 strain to lactoferrin induced the vncRS operon, lysis, and subsequent in vivo cytokine production, resulting in lung inflammation. However, these responses were significantly attenuated in pneumococci harboring a mutation in pep27. Mechanistically, the VncS ligand, identified as lactoferrin, induced the vncRS operon and increased the in vivo mortality rates. Thus, serum-induced activation of vncRS plays an essential role in inducing pneumonia.
Collapse
Affiliation(s)
- Seungyeop Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | | | - Hyogyoung Kwon
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan, Korea
| | - Sang-Sang Park
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Gyu-Lee Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Sang-Yoon Choi
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Eun-Hye Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | | | - Seung Han Seon
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Nhat Tu Le
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Hamid Iqbal
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Sangho Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Suhkneung Pyo
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Dong-Kwon Rhee
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
28
|
Schoergenhofer C, Matzneller P, Mußbacher M, Schmid JA, Jilma-Stohlawetz P, Zeitlinger M, Jilma B. Colistin dampens fibrinolysis and endothelial activation during endotoxaemia. A randomised, double blind trial. Thromb Haemost 2017; 117:1714-1721. [PMID: 28796276 PMCID: PMC6292133 DOI: 10.1160/th17-03-0196] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/20/2017] [Indexed: 12/20/2022]
Abstract
Colistin electrostatically interacts with lipopolysaccharides (LPS). Preclinical studies demonstrated beneficial effects of colistin on LPS-induced coagulation and fibrinolysis. The objective of this trial was to investigate the effects of colistin during experimental endotoxaemia. In this randomised, double-blind, placebo-controlled, crossover trial 16 healthy volunteers received a 2 ng/kg LPS bolus after infusion of 2.5 million IU colistin or placebo. Plasma levels of F1+2 prothrombin fragments, thrombin-antithrombin complexes (TAT), von Willebrand factor antigen levels (vWF), E-selectin, plasmin-antiplasmin complexes (PAP), tissue-type plasminogen activator (t-PA) antigen and activity, plasminogen activator inhibitor-1 (PAI-1) were measured. Infusion of colistin significantly reduced peak concentrations of PAP complexes by 70 %, t-PA antigen levels by 63 % and t-PA activity by 48 %, while PAI-1 levels decreased numerically by 63 %. Two hours after the LPS bolus F1+2 levels and TAT complexes were slightly reduced in the colistin period, but peak concentrations were similar in both periods. Colistin blunted the LPS induced four-fold increase in soluble E-Selectin levels by ∼50 % and the two-fold increase in vWF antigen levels by ∼70 %. The LPS-scavenging actions of colistin significantly reduce endothelial activation and fibrinolytic response in the human endotoxaemia model, while the activation of the coagulation system remains largely unaffected. Note: This work was conducted at the Medical University of Vienna. EudraCT-Nr.: 2014–00285720
Supplementary Material to this article is available online at
http://www.thrombosis-online.com
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bernd Jilma
- Bernd Jilma, MD, Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria, Tel.: +43 1 40400 29810, Fax: +43 1 40400 29980, E-mail:
| |
Collapse
|