1
|
Walrath T, Najarro KM, Giesy LE, Khair S, Orlicky DJ, McMahan RH, Kovacs EJ. Reducing the excessive inflammation after burn injury in aged mice by maintaining a healthier intestinal microbiome. FASEB J 2024; 38:e70065. [PMID: 39305117 PMCID: PMC11465428 DOI: 10.1096/fj.202401020r] [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: 05/06/2024] [Revised: 08/09/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024]
Abstract
One in six people are projected to be 65 years or older by 2050. As the population ages, better treatments for injuries that disproportionately impact the aged population will be needed. Clinical studies show that people aged 65 and older experience higher rates of morbidity and mortality after burn injury, including a greater incidence of pulmonary complications when compared to younger burn injured adults, which we and others believe is mediated, in part, by inflammation originating in the intestines. Herein, we use our clinically relevant model of scald burn injury in young and aged mice to determine whether cohousing aged mice with young mice or giving aged mice oral gavage of fecal material from young mice is sufficient to alter the microbiome of the aged mice and protect them from inflammation in the ileum and the lungs. Aged burn injured mice have less DNA expression of Bacteroidetes in the feces and an unhealthy Firmicutes/Bacteroidetes ratio. Both Bacteroidetes and the ratio of these two phyla are restored in aged burn injured by prior cohousing for a month with younger mice but not fecal transfer from young mice. This shift in the microbiome coincides with heightened expression of danger-associated molecular patterns (DAMP), and pro-inflammatory cytokine interleukin-6 (il6) in the ileum and lung of aged, burn injured mice, and heightened antimicrobial peptide camp in the lung. Cohousing reverses DAMP expression in the ileum and lung, and cathelicidin-related antimicrobial peptide protein (camp) in the lung, while fecal transfer heightened DAMPs while reducing camp in the lung, and also increased IL-6 protein in the lungs. These results highlight the importance of the intestinal microbiome in mediating inflammation within the gut-lung axis, giving insights into potential future treatments in the clinic.
Collapse
Affiliation(s)
- Travis Walrath
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kevin M. Najarro
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lauren E. Giesy
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Shanawaj Khair
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David J. Orlicky
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rachel H. McMahan
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Elizabeth J. Kovacs
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Veterans Health Administration, Eastern Colorado Health Care System, Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, Colorado, USA
| |
Collapse
|
2
|
Zhu S, Dai D, Li H, Huang J, Kang W, Yang Y, Zhong Y, Xiang Y, Liu C, He J, Liang Z. Bovine Neutrophil β-Defensin-5 Provides Protection against Multidrug-Resistant Klebsiella pneumoniae via Regulating Pulmonary Inflammatory Response and Metabolic Response. Int J Mol Sci 2024; 25:10506. [PMID: 39408834 PMCID: PMC11477005 DOI: 10.3390/ijms251910506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Klebsiella pneumoniae (K. pneumoniae), a kind of zoonotic bacteria, is among the most common antibiotic-resistant pathogens, and it causes nosocomial infections that pose a threat to public health. In this study, the roles of synthetic bovine neutrophil β-defensin-5 (B5) in regulating inflammatory response and metabolic response against multidrug-resistant K. pneumoniae infection in a mouse model were investigated. Mice were administrated intranasally with 20 μg of B5 twice and challenged with K. pneumoniae three days after B5 pretreatment. Results showed that B5 failed to directly kill K. pneumoniae in vitro, but it provided effective protection against multidrug-resistant K. pneumoniae via decreasing the bacterial load in the lungs and spleen, and by alleviating K. pneumoniae-induced histopathological damage in the lungs. Furthermore, B5 significantly enhanced the mRNA expression of TNF-α, IL-1β, Cxcl1, Cxcl5, Ccl17, and Ccl22 and obviously enhanced the rapid recruitment of macrophages and dendritic cells in the lungs in the early infection phase, but significantly down-regulated the levels of TNF-α, IL-1β, and IL-17 in the lungs in the later infection phase. Moreover, RNA-seq results showed that K. pneumoniae infection activated signaling pathways related to natural killer cell-mediated cytotoxicity, IL-17 signaling pathway, inflammatory response, apoptosis, and necroptosis in the lungs, while B5 inhibited these signaling pathways. Additionally, K. pneumoniae challenge led to the suppression of glycerophospholipid metabolism, the phosphotransferase system, the activation of microbial metabolism in diverse environments, and metabolic pathways in the lungs. However, B5 significantly reversed these metabolic responses. Collectively, B5 can effectively regulate the inflammatory response caused by K. pneumoniae and offer protection against K. pneumoniae. B5 may be applied as an adjuvant to the existing antimicrobial therapy to control multidrug-resistant K. pneumoniae infection. Our study highlights the potential of B5 in enhancing pulmonary bacterial clearance and alleviating K. pneumoniae-caused inflammatory damage.
Collapse
Affiliation(s)
- Shuxin Zhu
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Dejia Dai
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Han Li
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Jingsheng Huang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Weichao Kang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Yunmei Yang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Yawen Zhong
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Yifei Xiang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Chengzhi Liu
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Jiakang He
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| | - Zhengmin Liang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning 530004, China
| |
Collapse
|
3
|
Pastuszak K, Palusińska-Szysz M, Wiącek AE, Jurak M. Thermodynamic Study on Biomimetic Legionella gormanii Bacterial Membranes. Molecules 2024; 29:4367. [PMID: 39339363 PMCID: PMC11434087 DOI: 10.3390/molecules29184367] [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: 07/30/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
The presented studies were aimed at determining the interactions in model membranes (Langmuir monolayers) created of phospholipids (PL) isolated from Legionella gormanii bacteria cultured with (PL + choline) or without (PL - choline) choline and to describe the impact of an antimicrobial peptide, human cathelicidin LL-37, on PL's monolayer behavior. The addition of choline to the growth medium influenced the mutual proportions of phospholipids extracted from L. gormanii. Four classes of phospholipids-phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylglycerol (PG), cardiolipin (CL), and their mixtures-were used to register compression isotherms with or without the LL-37 peptide in the subphase. Based on them the excess area (Ae), excess (ΔGe), and total (ΔGm) Gibbs energy of mixing were determined. The thermodynamic analyses revealed that the PL - choline monolayer showed greater repulsive forces between molecules in comparison to the ideal system, while the PL + choline monolayer was characterized by greater attraction. The LL-37 peptide affected the strength of interactions between phospholipids' molecules and reduced the monolayers stability. Accordingly, the changes in interactions in the model membranes allowed us to determine the difference in their susceptibility to the LL-37 peptide depending on the choline supplementation of bacterial culture.
Collapse
Affiliation(s)
- Katarzyna Pastuszak
- Department of Interfacial Phenomena, Institute of Chemical Sciences, Faculty of Chemistry, Maria Curie-Skłodowska University, Maria Curie-Skłodowska Sq. 3, 20-031 Lublin, Poland; (K.P.); (A.E.W.)
| | - Marta Palusińska-Szysz
- Department of Genetics and Microbiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland;
| | - Agnieszka Ewa Wiącek
- Department of Interfacial Phenomena, Institute of Chemical Sciences, Faculty of Chemistry, Maria Curie-Skłodowska University, Maria Curie-Skłodowska Sq. 3, 20-031 Lublin, Poland; (K.P.); (A.E.W.)
| | - Małgorzata Jurak
- Department of Interfacial Phenomena, Institute of Chemical Sciences, Faculty of Chemistry, Maria Curie-Skłodowska University, Maria Curie-Skłodowska Sq. 3, 20-031 Lublin, Poland; (K.P.); (A.E.W.)
| |
Collapse
|
4
|
Zehr EP, Erzen CL, Oshima K, Langouet-Astrie CJ, LaRiviere WB, Shi D, Zhang F, McCollister BD, Windham SL, Rizzo AN, Bastarache JA, Horswill AR, Schmidt EP, Kwiecinski JM, Colbert JF. Bacterial pneumonia-induced shedding of epithelial heparan sulfate inhibits the bactericidal activity of cathelicidin in a murine model. Am J Physiol Lung Cell Mol Physiol 2024; 326:L206-L212. [PMID: 38113313 PMCID: PMC11280675 DOI: 10.1152/ajplung.00178.2023] [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: 06/05/2023] [Revised: 12/08/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023] Open
Abstract
Bacterial pneumonia is a common clinical syndrome leading to significant morbidity and mortality worldwide. In the current study, we investigate a novel, multidirectional relationship between the pulmonary epithelial glycocalyx and antimicrobial peptides in the setting of methicillin-resistant Staphylococcus aureus (MRSA) pneumonia. Using an in vivo pneumonia model, we demonstrate that highly sulfated heparan sulfate (HS) oligosaccharides are shed into the airspaces in response to MRSA pneumonia. In vitro, these HS oligosaccharides do not directly alter MRSA growth or gene transcription. However, in the presence of an antimicrobial peptide (cathelicidin), increasing concentrations of HS inhibit the bactericidal activity of cathelicidin against MRSA as well as other nosocomial pneumonia pathogens (Klebsiella pneumoniae and Pseudomonas aeruginosa) in a dose-dependent manner. Surface plasmon resonance shows avid binding between HS and cathelicidin with a dissociation constant of 0.13 μM. These findings highlight a complex relationship in which shedding of airspace HS may hamper host defenses against nosocomial infection via neutralization of antimicrobial peptides. These findings may inform future investigation into novel therapeutic targets designed to restore local innate immune function in patients suffering from primary bacterial pneumonia.NEW & NOTEWORTHY Primary Staphylococcus aureus pneumonia causes pulmonary epithelial heparan sulfate (HS) shedding into the airspace. These highly sulfated HS fragments do not alter bacterial growth or transcription, but directly bind with host antimicrobial peptides and inhibit the bactericidal activity of these cationic polypeptides. These findings highlight a complex local interaction between the pulmonary epithelial glycocalyx and antimicrobial peptides in the setting of bacterial pneumonia.
Collapse
Affiliation(s)
- Evan P Zehr
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Christopher L Erzen
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Kaori Oshima
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States
| | | | - Wells B LaRiviere
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Deling Shi
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States
| | - Fuming Zhang
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States
| | - Bruce D McCollister
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Samuel L Windham
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Alicia N Rizzo
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Julie A Bastarache
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States
| | - Alexander R Horswill
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, United States
| | - Eric P Schmidt
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States
| | | | - James F Colbert
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, United States
| |
Collapse
|
5
|
Abstract
The phenomenon of swarming has long been observed in nature as a strategic event that serves as a good offense toward prey and predators. Imaging studies have uncovered that neutrophils employ this swarm-like tactic within infected and inflamed tissues as part of the innate immune response. Much of our understanding of neutrophil swarming builds from observations during sterile inflammation and various bacterial, fungal, and parasitic infections of the skin. However, the architecture and function of the skin differ significantly from vital organs where highly specialized microenvironments carry out critical functions. Therefore, the detrimental extent this perturbation may have on organ function remains unclear. In this review, we examine organ-specific swarming within the skin, liver, and lungs, with a detailed focus on swarming within microvascular environments. In addition, we examine potential "swarmulants" that initiate both transient and persistent swarms that have been implicated in disease.
Collapse
Affiliation(s)
- Luke Brown
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Bryan G. Yipp
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
6
|
Duarte-Mata DI, Salinas-Carmona MC. Antimicrobial peptides´ immune modulation role in intracellular bacterial infection. Front Immunol 2023; 14:1119574. [PMID: 37056758 PMCID: PMC10086130 DOI: 10.3389/fimmu.2023.1119574] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Intracellular bacteria cause a wide range of diseases, and their intracellular lifestyle makes infections difficult to resolve. Furthermore, standard therapy antibiotics are often unable to eliminate the infection because they have poor cellular uptake and do not reach the concentrations needed to kill bacteria. In this context, antimicrobial peptides (AMPs) are a promising therapeutic approach. AMPs are short cationic peptides. They are essential components of the innate immune response and important candidates for therapy due to their bactericidal properties and ability to modulate host immune responses. AMPs control infections through their diverse immunomodulatory effects stimulating and/or boosting immune responses. This review focuses on AMPs described to treat intracellular bacterial infections and the known immune mechanisms they influence.
Collapse
|
7
|
Hu MY, Chen YW, Chai ZF, Wang YZ, Lin JQ, Fang SG. Antibacterial Properties and Potential Mechanism of Serum from Chinese Alligator. Microorganisms 2022; 10:2210. [PMID: 36363802 PMCID: PMC9698150 DOI: 10.3390/microorganisms10112210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 04/06/2025] Open
Abstract
The Chinese alligator (Alligator sinensis) is an ancient reptile with strong immunity that lives in wetland environments. This study tested the antibacterial ability of Chinese alligator serum (CAS) against Klebsiella pneumoniae, Escherichia coli, Staphylococcus aureus, and Pseudomonas aeruginosa and analyzed the potential underlying mechanisms. Results showed that the CAS had a marked antibacterial effect on K. pneumoniae, E. coli, and P. aeruginosa, while S. aureus was only mildly affected. However, these effects disappeared when Protease K was added to the serum. The serum proteome analysis revealed that the antibacterial ability of CAS was produced by interactions among various proteins and that the complement proteins played a major antibacterial role. Therefore, we made relevant predictions about the structure and function of complement component 3. In addition, sequence alignment and phylogenetic analysis of complement component 3d (C3d) in four mammalian species and two alligator species showed that the amino acids that make up the acid pocket on the concave surface of alligator C3d are not identical to those in mammals. This study provided evidence that CAS elicits significant antibacterial effects against some pathogens and provides the basis for further development of novel antibacterial drugs.
Collapse
Affiliation(s)
- Meng-Yuan Hu
- MOE Key Laboratory of Biosystems Homeostasis and Protection, State Conservation Center for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yi-Wen Chen
- MOE Key Laboratory of Biosystems Homeostasis and Protection, State Conservation Center for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhi-Fan Chai
- MOE Key Laboratory of Biosystems Homeostasis and Protection, State Conservation Center for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yin-Zhi Wang
- MOE Key Laboratory of Biosystems Homeostasis and Protection, State Conservation Center for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jian-Qing Lin
- Guangdong Provincial Key Laboratory of Marine Disaster Prediction and Prevention, Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Science, Shantou University, Shantou 515063, China
| | - Sheng-Guo Fang
- MOE Key Laboratory of Biosystems Homeostasis and Protection, State Conservation Center for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
8
|
Sung PS, Peng YC, Yang SP, Chiu CH, Hsieh SL. CLEC5A is critical in Pseudomonas aeruginosa-induced acute lung injury. JCI Insight 2022; 7:156613. [PMID: 36048544 PMCID: PMC9676025 DOI: 10.1172/jci.insight.156613] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
Pseudomonas aeruginosa is one of the most common nosocomial infections worldwide, and it frequently causes ventilator-associated acute pneumonia in immunocompromised patients. Abundant neutrophil extracellular traps (NETs) contribute to acute lung injury, thereby aggravating ventilator-induced lung damage. While pattern recognition receptors (PRRs) TLR4 and TLR5 are required for host defense against P. aeruginosa invasion, the PRR responsible for P. aeruginosa–induced NET formation, proinflammatory cytokine release, and acute lung injury remains unclear. We found that myeloid C-type lectin domain family 5 member A (CLEC5A) interacts with LPS of P. aeruginosa and is responsible for P. aeruginosa–induced NET formation and lung inflammation. P. aeruginosa activates CLEC5A to induce caspase-1–dependent NET formation, but it neither causes gasdermin D (GSDMD) cleavage nor contributes to P. aeruginosa–induced neutrophil death. Blockade of CLEC5A attenuates P. aeruginosa–induced NETosis and lung injury, and simultaneous administration of anti-CLEC5A mAb with ciprofloxacin increases survival rate and decreases collagen deposition in the lungs of mice challenged with a lethal dose of P. aeruginosa. Thus, CLEC5A is a promising therapeutic target to reduce ventilator-associated lung injury and fibrosis in P. aeruginosa–induced pneumonia.
Collapse
Affiliation(s)
- Pei-Shan Sung
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chun Peng
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shao-Ping Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Cheng-Hsun Chiu
- Department of Pediatrics, Chang Gung Children's Hospital, Taoyuan, Taiwan
| | | |
Collapse
|
9
|
Zhang X, He D, Jia J, Liang F, Mei J, Li W, Liu T, Wang Z, Liu Y, Zhang F, Zhang Z, Luo B. Erythropoietin mediates re-programming of endotoxin-tolerant macrophages through PI3K/AKT signaling and protects mice against secondary infection. Front Immunol 2022; 13:938944. [PMID: 36016936 PMCID: PMC9396350 DOI: 10.3389/fimmu.2022.938944] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 07/12/2022] [Indexed: 11/21/2022] Open
Abstract
Initial lipopolysaccharide (LPS) exposure leads to a hypo-responsive state by macrophages to a secondary stimulation of LPS, known as endotoxin tolerance. However, recent findings show that functions of endotoxin-tolerant macrophages are not completely suppressed, whereas they undergo a functional re-programming process with upregulation of a panel of molecules leading to enhanced protective functions including antimicrobial and tissue-remodeling activities. However, the underlying molecular mechanisms are still elusive. Erythropoietin (EPO), a glycoprotein regulated by hypoxia-inducible factor 1α (HIF-1α), exerts anti-inflammatory and tissue-protective activities. Nevertheless, the potential effects of EPO on functional re-programming of endotoxin-tolerant macrophages have not been investigated yet. Here, we found that initial LPS exposure led to upregulation of HIF-1α/EPO in macrophages and that EPO enhanced tolerance in tolerized macrophages and mice as demonstrated by suppressed proinflammatory genes such as Il1b, Il6, and Tnfa after secondary LPS stimulation. Moreover, we showed that EPO improved host protective genes in endotoxin-tolerant macrophages and mice, such as the anti-bacterial genes coding for cathelicidin-related antimicrobial peptide (Cnlp) and macrophage receptor with collagenous structure (Marco), and the tissue-repairing gene vascular endothelial growth factor C (Vegfc). Therefore, our findings indicate that EPO mediates the functional re-programming of endotoxin-tolerant macrophages. Mechanistically, we found that PI3K/AKT signaling contributed to EPO-mediated re-programming through upregulation of Irak3 and Wdr5 expression. Specifically, IL-1 receptor-associated kinase 3 (IRAK3) was responsible for inhibiting proinflammatory genes Il1b, Il6, and Tnfa in tolerized macrophages after LPS rechallenge, whereas WDR5 contributed to the upregulation of host beneficial genes including Cnlp, Marco, and Vegfc. In a septic model of mice, EPO pretreatment significantly promoted endotoxin-tolerant re-programming, alleviated lung injury, enhanced bacterial clearance, and decreased mortality in LPS-tolerized mice after secondary infection of Escherichia coli. Collectively, our results reveal a novel role for EPO in mediating functional re-programming of endotoxin-tolerant macrophages; thus, targeting EPO appears to be a new therapeutic option in sepsis and other inflammatory disorders.
Collapse
Affiliation(s)
- Xue Zhang
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dan He
- Medical College, Chongqing University, Chongqing, China
| | - Jialin Jia
- Medical College, Chongqing University, Chongqing, China
| | - Feihong Liang
- Department of Medical Science, Shunde Polytechnic, Foshan, China
| | - Jie Mei
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenhua Li
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tingting Liu
- Institute of Immunology, Army Medical University, Chongqing, China
| | - Zhiyu Wang
- Institute of Immunology, Army Medical University, Chongqing, China
| | - Yu Liu
- Institute of Immunology, Army Medical University, Chongqing, China
| | - Fengxue Zhang
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Bangwei Luo, ; Zhiren Zhang, ; Fengxue Zhang,
| | - Zhiren Zhang
- Institute of Immunology, Army Medical University, Chongqing, China
- *Correspondence: Bangwei Luo, ; Zhiren Zhang, ; Fengxue Zhang,
| | - Bangwei Luo
- Institute of Immunology, Army Medical University, Chongqing, China
- *Correspondence: Bangwei Luo, ; Zhiren Zhang, ; Fengxue Zhang,
| |
Collapse
|
10
|
Blair JMA, Zeth K, Bavro VN, Sancho-Vaello E. The role of bacterial transport systems in the removal of host antimicrobial peptides in Gram-negative bacteria. FEMS Microbiol Rev 2022; 46:6617596. [PMID: 35749576 PMCID: PMC9629497 DOI: 10.1093/femsre/fuac032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/23/2022] [Accepted: 06/22/2022] [Indexed: 01/09/2023] Open
Abstract
Antibiotic resistance is a global issue that threatens our progress in healthcare and life expectancy. In recent years, antimicrobial peptides (AMPs) have been considered as promising alternatives to the classic antibiotics. AMPs are potentially superior due to their lower rate of resistance development, since they primarily target the bacterial membrane ('Achilles' heel' of the bacteria). However, bacteria have developed mechanisms of AMP resistance, including the removal of AMPs to the extracellular space by efflux pumps such as the MtrCDE or AcrAB-TolC systems, and the internalization of AMPs to the cytoplasm by the Sap transporter, followed by proteolytic digestion. In this review, we focus on AMP transport as a resistance mechanism compiling all the experimental evidence for the involvement of efflux in AMP resistance in Gram-negative bacteria and combine this information with the analysis of the structures of the efflux systems involved. Finally, we expose some open questions with the aim of arousing the interest of the scientific community towards the AMPs-efflux pumps interactions. All the collected information broadens our understanding of AMP removal by efflux pumps and gives some clues to assist the rational design of AMP-derivatives as inhibitors of the efflux pumps.
Collapse
Affiliation(s)
- Jessica M A Blair
- College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Kornelius Zeth
- Department of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark
| | - Vassiliy N Bavro
- School of Life Sciences, University of Essex, Colchester, CO4 3SQ, United Kingdom
| | - Enea Sancho-Vaello
- Corresponding author. College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom. E-mail:
| |
Collapse
|
11
|
Bhusal A, Nam Y, Seo D, Rahman MH, Hwang EM, Kim S, Lee W, Suk K. Cathelicidin‐related antimicrobial peptide promotes neuroinflammation through astrocyte–microglia communication in experimental autoimmune encephalomyelitis. Glia 2022; 70:1902-1926. [DOI: 10.1002/glia.24227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/10/2022] [Accepted: 05/25/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Anup Bhusal
- Department of Pharmacology, School of Medicine Kyungpook National University Daegu Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine Kyungpook National University Daegu Republic of Korea
| | - Youngpyo Nam
- Department of Pharmacology, School of Medicine Kyungpook National University Daegu Republic of Korea
| | - Donggun Seo
- Department of Pharmacology, School of Medicine Kyungpook National University Daegu Republic of Korea
| | - Md Habibur Rahman
- Department of Pharmacology, School of Medicine Kyungpook National University Daegu Republic of Korea
- Division of Endocrinology, Department of Medicine Rutgers Robert Wood Johnson Medical School New Brunswick New Jersey USA
| | - Eun Mi Hwang
- Brain Science Institute, Korea Institute of Science and Technology Seoul Republic of Korea
| | - Seung‐Chan Kim
- Brain Science Institute, Korea Institute of Science and Technology Seoul Republic of Korea
| | - Won‐Ha Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group Kyungpook National University Daegu Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine Kyungpook National University Daegu Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine Kyungpook National University Daegu Republic of Korea
- Brain Science and Engineering Institute Kyungpook National University Daegu Republic of Korea
| |
Collapse
|
12
|
Escobar‐Salom M, Torrens G, Jordana‐Lluch E, Oliver A, Juan C. Mammals' humoral immune proteins and peptides targeting the bacterial envelope: from natural protection to therapeutic applications against multidrug‐resistant
Gram
‐negatives. Biol Rev Camb Philos Soc 2022; 97:1005-1037. [PMID: 35043558 PMCID: PMC9304279 DOI: 10.1111/brv.12830] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022]
Abstract
Mammalian innate immunity employs several humoral ‘weapons’ that target the bacterial envelope. The threats posed by the multidrug‐resistant ‘ESKAPE’ Gram‐negative pathogens (Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.) are forcing researchers to explore new therapeutic options, including the use of these immune elements. Here we review bacterial envelope‐targeting (peptidoglycan and/or membrane‐targeting) proteins/peptides of the mammalian immune system that are most likely to have therapeutic applications. Firstly we discuss their general features and protective activity against ESKAPE Gram‐negatives in the host. We then gather, integrate, and discuss recent research on experimental therapeutics harnessing their bactericidal power, based on their exogenous administration and also on the discovery of bacterial and/or host targets that improve the performance of this endogenous immunity, as a novel therapeutic concept. We identify weak points and knowledge gaps in current research in this field and suggest areas for future work to obtain successful envelope‐targeting therapeutic options to tackle the challenge of antimicrobial resistance.
Collapse
Affiliation(s)
- María Escobar‐Salom
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Gabriel Torrens
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Elena Jordana‐Lluch
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Antonio Oliver
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| | - Carlos Juan
- Department of Microbiology University Hospital Son Espases‐Health Research Institute of the Balearic Islands (IdISBa) Carretera de Valldemossa 79 Palma Balearic Islands 07010 Spain
| |
Collapse
|
13
|
Endogenous cathelicidin is required for protection against ZIKV-caused testis damage via inactivating virons. Antiviral Res 2022; 198:105248. [PMID: 35038500 DOI: 10.1016/j.antiviral.2022.105248] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/04/2022] [Accepted: 01/10/2022] [Indexed: 12/18/2022]
Abstract
Cathelicidins have been shown to effectively inhibit flavivirus replication in vitro. However, the effects of mouse and human endogenous cathelicidins on flavivirus infection in vivo are rarely known. We herein found that mouse endogenous cathelicidin CRAMP was significantly up-regulated upon Zika virus (ZIKV) infection. CRAMP deficiency markedly exacerbated ZIKV replication in testis, and aggravated ZIKV-induced testicular damage and ZIKV-induced spermatic damage in mice, indicating that endogenous cathelicidin is required for protection against ZIKV-caused male infertility in mice. In vitro antiviral assay showed that both mouse cathelidin CRAMP and human cathelicidin LL-37 obviously reduced ZIKV-caused cytopathic effect and inhibited ZIKV replication in Vero cells. Antiviral mechanism revealed that they both directly inactivated ZIKV virons by binding to ZIKV virons and inducing the leakage of ZIKV genomic RNA, consequently inactivated ZIKV virons. In vivo antiviral assay indicated that both of them effectively inhibited ZIKV replication in C57BL/6J and IFNα/β receptor-deficient (Ifnar1-/-) mice when CRAMP or LL-37 was intravenously injected in parallel with or at 1 h after intravenous injection of ZIKV, implying that mouse cathelidin CRAMP and human cathelicidin LL-37 effectively inactivated ZIKV particles and exhibited therapeutic potential against ZIKV infection in vivo. Our findings reveal that endogenous cahtelicidin CRAMP and LL-37 act as inactivators of ZIKV, and effectively protect against ZIKV replication and ZIKV-induced male infertility, highlighting their potential for therapy of ZIKV infection.
Collapse
|
14
|
Cabak A, Hovold G, Petersson AC, Ramstedt M, Påhlman LI. Activity of airway antimicrobial peptides against cystic fibrosis pathogens. Pathog Dis 2021; 78:5898671. [PMID: 32857857 DOI: 10.1093/femspd/ftaa048] [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: 06/08/2020] [Accepted: 08/26/2020] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial peptides are important players of the innate host defence against invading microorganisms. The aim of this study was to evaluate the activity of airway antimicrobial peptides against the common cystic fibrosis (CF) pathogen Pseudomonas aeruginosa, and to compare it to the emerging multi-drug resistant CF pathogens Achromobacter xylosoxidans and Stenotrophomonas maltophilia. Clinical bacterial isolates from CF patients were used, and the antimicrobial activity of human beta-defensin 2 and 3, LL37 and lysozyme was evaluated using radial diffusion assay and viable counts. The cell surface zeta potential was analysed to estimate the net charge at the bacterial surface. Of the bacterial species included in the study, A. xylosoxidans was the most resistant to antimicrobial peptides, whereas P. aeruginosa was the most susceptible. The net charge of the bacterial surface was significantly more negative for P. aeruginosa compared to A. xylosoxidans, which may in part explain the differences in susceptibility.
Collapse
Affiliation(s)
- Andrea Cabak
- Department of Clinical Sciences Lund, Division of Infection Medicine, Lund University, BMC B14, Sölvegatan 19, S-221 84 Lund, Sweden
| | - Gisela Hovold
- Department of Clinical Sciences Lund, Division of Infection Medicine, Lund University, BMC B14, Sölvegatan 19, S-221 84 Lund, Sweden
| | - Ann-Cathrine Petersson
- Department of Clinical Microbiology, Laboratory medicine, Region Skåne, Sölvegatan 23B, S-221 85 Lund, Sweden
| | - Madeleine Ramstedt
- Department of Chemistry, Umeå Centre of Microbial Research, Umeå University, S-901 87, Umeå, Sweden
| | - Lisa I Påhlman
- Department of Clinical Sciences Lund, Division of Infection Medicine, Lund University, BMC B14, Sölvegatan 19, S-221 84 Lund, Sweden.,Division of Infectious Diseases, Skåne University Hospital Lund, Hälsogatan 3, S-221 85 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Klinikgatan 32, S-221 84, Lund, Sweden
| |
Collapse
|
15
|
Rangasamy T, Ghimire L, Jin L, Le J, Periasamy S, Paudel S, Cai S, Jeyaseelan S. Host Defense against Klebsiella pneumoniae Pneumonia Is Augmented by Lung-Derived Mesenchymal Stem Cells. THE JOURNAL OF IMMUNOLOGY 2021; 207:1112-1127. [PMID: 34341173 DOI: 10.4049/jimmunol.2000688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 06/16/2021] [Indexed: 11/19/2022]
Abstract
Klebsiella pneumoniae is a common cause of Gram-negative pneumonia. The spread of antibiotic-resistant and hypervirulent strains has made treatment more challenging. This study sought to determine the immunomodulatory, antibacterial, and therapeutic potential of purified murine stem cell Ag-1+ (Sca-1+) lung mesenchymal stem cells (LMSCs) using in vitro cell culture and an in vivo mouse model of pneumonia caused by K pneumoniae. Sca-1+ LMSCs are plastic adherent, possess colony-forming capacity, express mesenchymal stem cell markers, differentiate into osteogenic and adipogenic lineages in vitro, and exhibit a high proliferative capacity. Further, these Sca-1+ LMSCs are morphologically similar to fibroblasts but differ ultrastructurally. Moreover, Sca-1+ LMSCs have the capacity to inhibit LPS-induced secretion of inflammatory cytokines by bone marrow-derived macrophages and neutrophils in vitro. Sca-1+ LMSCs inhibit the growth of K pneumoniae more potently than do neutrophils. Sca-1+ LMSCs also possess the intrinsic ability to phagocytize and kill K. pneumoniae intracellularly. Whereas the induction of autophagy promotes bacterial replication, inhibition of autophagy enhances the intracellular clearance of K. pneumoniae in Sca-1+ LMSCs during the early time of infection. Adoptive transfer of Sca-1+ LMSCs in K. pneumoniae-infected mice improved survival, reduced inflammatory cells in bronchoalveolar lavage fluid, reduced inflammatory cytokine levels and pathological lesions in the lung, and enhanced bacterial clearance in the lung and in extrapulmonary organs. To our knowledge, these results together illustrate for the first time the protective role of LMSCs in bacterial pneumonia.
Collapse
Affiliation(s)
- Tirumalai Rangasamy
- Center for Lung Biology and Disease, Louisiana State University, Baton Rouge, LA; .,Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Laxman Ghimire
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Liliang Jin
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - John Le
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Sivakumar Periasamy
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Sagar Paudel
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Shanshan Cai
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Samithamby Jeyaseelan
- Center for Lung Biology and Disease, Louisiana State University, Baton Rouge, LA; .,Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and.,Division of Pulmonary and Critical Care, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA
| |
Collapse
|
16
|
Schromm AB, Paulowski L, Kaconis Y, Kopp F, Koistinen M, Donoghue A, Keese S, Nehls C, Wernecke J, Garidel P, Sevcsik E, Lohner K, Sanchez-Gomez S, Martinez-de-Tejada G, Brandenburg K, Brameshuber M, Schütz GJ, Andrä J, Gutsmann T. Cathelicidin and PMB neutralize endotoxins by multifactorial mechanisms including LPS interaction and targeting of host cell membranes. Proc Natl Acad Sci U S A 2021; 118:e2101721118. [PMID: 34183393 PMCID: PMC8271772 DOI: 10.1073/pnas.2101721118] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Antimicrobial peptides (AMPs) contribute to an effective protection against infections. The antibacterial function of AMPs depends on their interactions with microbial membranes and lipids, such as lipopolysaccharide (LPS; endotoxin). Hyperinflammation induced by endotoxin is a key factor in bacterial sepsis and many other human diseases. Here, we provide a comprehensive profile of peptide-mediated LPS neutralization by systematic analysis of the effects of a set of AMPs and the peptide antibiotic polymyxin B (PMB) on the physicochemistry of endotoxin, macrophage activation, and lethality in mice. Mechanistic studies revealed that the host defense peptide LL-32 and PMB each reduce LPS-mediated activation also via a direct interaction of the peptides with the host cell. As a biophysical basis, we demonstrate modifications of the structure of cholesterol-rich membrane domains and the association of glycosylphosphatidylinositol (GPI)-anchored proteins. Our discovery of a host cell-directed mechanism of immune control contributes an important aspect in the development and therapeutic use of AMPs.
Collapse
Affiliation(s)
- Andra B Schromm
- Division of Immunobiophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany;
| | - Laura Paulowski
- Division of Immunobiophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Yani Kaconis
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Franziska Kopp
- Division of Immunobiophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Max Koistinen
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Annemarie Donoghue
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Susanne Keese
- Division of Immunobiophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Christian Nehls
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | - Julia Wernecke
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
- Deutsches Elektronen-Synchrotron, D-22607 Hamburg, Germany
| | - Patrick Garidel
- Biophysikalische Chemie, Martin-Luther-Universität Halle-Wittenberg, D-06108 Halle, Germany
| | - Eva Sevcsik
- Institute of Applied Physics at TU Wien, Vienna 1040, Austria
| | - Karl Lohner
- Institute of Molecular Biosciences, Biophysics Division, University of Graz, A-8010 Graz, Austria
- BioTechMed-Graz, A-8010 Graz, Austria
| | - Susana Sanchez-Gomez
- Department of Microbiology and Parasitology, University of Navarra, E-31008 Pamplona, Spain
| | - Guillermo Martinez-de-Tejada
- Department of Microbiology and Parasitology, University of Navarra, E-31008 Pamplona, Spain
- Navarra Institute for Health Research, E-31008 Pamplona, Spain
| | - Klaus Brandenburg
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| | | | | | - Jörg Andrä
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
- Department of Biotechnology, Faculty of Life Sciences, Hamburg University of Applied Sciences, D-21033 Hamburg, Germany
| | - Thomas Gutsmann
- Division of Biophysics, Research Center Borstel, Leibniz Lung Center, D-23845 Borstel, Germany
| |
Collapse
|
17
|
Holani R, Babbar A, Blyth GAD, Lopes F, Jijon H, McKay DM, Hollenberg MD, Cobo ER. Cathelicidin-mediated lipopolysaccharide signaling via intracellular TLR4 in colonic epithelial cells evokes CXCL8 production. Gut Microbes 2020; 12:1785802. [PMID: 32658599 PMCID: PMC7524372 DOI: 10.1080/19490976.2020.1785802] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We hypothesized that the antimicrobial peptide cathelicidin has a physiological role in regulating gut inflammatory homeostasis. We determined that cathelicidin synergizes with LPS to facilitate its internalization and signaling via endosomic TLR4 in colonic epithelium, evoking synthesis of the human neutrophil chemoattractant, CXCL8 (or murine homolog, CXCL1). Interaction of cathelicidin with LPS in the control of CXCL8/CXCL1 synthesis was assessed in human colon epithelial cells, murine colonoids and cathelicidin-null mice (Camp-/- ). Mechanistically, human cathelicidin (LL-37), as an extracellular complex with LPS, interacted with lipid raft-associated GM1 gangliosides to internalize and activate intracellular TLR4. Two signaling pathways converged on CXCL8/CXCL1 production: (1) a p38MAPK-dependent pathway regulated by Src-EGFR kinases; and, (2) a p38MAPK-independent, NF-κB-dependent pathway, regulated by MEK1/2-MAPK. Increased cathelicidin-dependent CXCL8 secretion in the colonic mucosa activated human blood-derived neutrophils. These cathelicidin effects occurred in vitro at concentrations well below those needed for microbicidal function. The important immunomodulatory role of cathelicidins was evident in cathelicidin-null/Camp-/- mice, which had diminished colonic CXCL1 secretion, decreased neutrophil recruitment-activation and reduced bacterial clearance when challenged with the colitis-inducing murine pathogen, Citrobacter rodentium. We conclude that in addition to its known microbicidal action, cathelicidin has a unique pathogen-sensing role, facilitating LPS-mediated intestinal responses, including the production of CXCL8/CXCL1 that would contribute to an integrated tissue response to recruit neutrophils during colitis.
Collapse
Affiliation(s)
- Ravi Holani
- Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Anshu Babbar
- Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Graham A. D. Blyth
- Microbiology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Fernando Lopes
- Institute of Parasitology, McGill University, Montreal, Canada
| | - Humberto Jijon
- Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Derek M. McKay
- Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Morley D. Hollenberg
- Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Eduardo R. Cobo
- Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada,CONTACT Eduardo R. Cobo Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, ABT2N 4N1, Canada
| |
Collapse
|
18
|
Singanayagam A, Glanville N, Cuthbertson L, Bartlett NW, Finney LJ, Turek E, Bakhsoliani E, Calderazzo MA, Trujillo-Torralbo MB, Footitt J, James PL, Fenwick P, Kemp SV, Clarke TB, Wedzicha JA, Edwards MR, Moffatt M, Cookson WO, Mallia P, Johnston SL. Inhaled corticosteroid suppression of cathelicidin drives dysbiosis and bacterial infection in chronic obstructive pulmonary disease. Sci Transl Med 2020; 11:11/507/eaav3879. [PMID: 31462509 DOI: 10.1126/scitranslmed.aav3879] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 02/12/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022]
Abstract
Bacterial infection commonly complicates inflammatory airway diseases such as chronic obstructive pulmonary disease (COPD). The mechanisms of increased infection susceptibility and how use of the commonly prescribed therapy inhaled corticosteroids (ICS) accentuates pneumonia risk in COPD are poorly understood. Here, using analysis of samples from patients with COPD, we show that ICS use is associated with lung microbiota disruption leading to proliferation of streptococcal genera, an effect that could be recapitulated in ICS-treated mice. To study mechanisms underlying this effect, we used cellular and mouse models of streptococcal expansion with Streptococcus pneumoniae, an important pathogen in COPD, to demonstrate that ICS impairs pulmonary clearance of bacteria through suppression of the antimicrobial peptide cathelicidin. ICS impairment of pulmonary immunity was dependent on suppression of cathelicidin because ICS had no effect on bacterial loads in mice lacking cathelicidin (Camp -/-) and exogenous cathelicidin prevented ICS-mediated expansion of streptococci within the microbiota and improved bacterial clearance. Suppression of pulmonary immunity by ICS was mediated by augmentation of the protease cathepsin D. Collectively, these data suggest a central role for cathepsin D/cathelicidin in the suppression of antibacterial host defense by ICS in COPD. Therapeutic restoration of cathelicidin to boost antibacterial immunity and beneficially modulate the lung microbiota might be an effective strategy in COPD.
Collapse
Affiliation(s)
- Aran Singanayagam
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK.
| | - Nicholas Glanville
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK
| | - Leah Cuthbertson
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Nathan W Bartlett
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK.,Faculty of Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, NSW 2305, Australia
| | - Lydia J Finney
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Elena Turek
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Eteri Bakhsoliani
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK
| | | | | | - Joseph Footitt
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK
| | - Phillip L James
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Peter Fenwick
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Samuel V Kemp
- Royal Brompton Hospital, Fulham Road, London SW2 6NP, UK
| | - Thomas B Clarke
- MRC Centre for Molecular Bacteriology and Infection, Department of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Jadwiga A Wedzicha
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Michael R Edwards
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK
| | - Miriam Moffatt
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - William O Cookson
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Patrick Mallia
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK
| | - Sebastian L Johnston
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK.
| |
Collapse
|
19
|
Ye J, Xu B, Fan B, Zhang J, Yuan F, Chen Y, Sun Z, Yan X, Song Y, Song S, Yang M, Yu JK. Discovery of Selenocysteine as a Potential Nanomedicine Promotes Cartilage Regeneration With Enhanced Immune Response by Text Mining and Biomedical Databases. Front Pharmacol 2020; 11:1138. [PMID: 32792959 PMCID: PMC7394085 DOI: 10.3389/fphar.2020.01138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/13/2020] [Indexed: 12/21/2022] Open
Abstract
Background Unlike bone tissue, little progress has been made regarding cartilage regeneration, and many challenges remain. Furthermore, the key roles of cartilage lesion caused by traumas, focal lesion, or articular overstress remain unclear. Traumatic injuries to the meniscus as well as its degeneration are important risk factors for long-term joint dysfunction, degenerative joint lesions, and knee osteoarthritis (OA) a chronic joint disease characterized by degeneration of articular cartilage and hyperosteogeny. Nearly 50% of the individuals with meniscus injuries develop OA over time. Due to the limited inherent self-repair capacity of cartilage lesion, the Biomaterial drug-nanomedicine is considered to be a promising alternative. Therefore, it is important to elucidate the gene potential regeneration mechanisms and discover novel precise medication, which are identified through this study to investigate their function and role in pathogenesis. Methods We downloaded the mRNA microarray statistics GSE117999, involving paired cartilage lesion tissue samples from 12 OA patients and 12 patients from a control group. First, we analyzed these statistics to recognize the differentially expressed genes (DEGs). We then exposed the gene ontology (GO) annotation and the Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway enrichment analyses for these DEGs. Protein-protein interaction (PPI) networks were then constructed, from which we attained eight significant genes after a functional interaction analysis. Finally, we identified a potential nanomedicine attained from this assay set, using a wide range of inhibitor information archived in the Search Tool for the Retrieval of Interacting Genes (STRING) database. Results Sixty-six DEGs were identified with our standards for meaning (adjusted P-value < 0.01, |log2 - FC| ≥1.2). Furthermore, we identified eight hub genes and one potential nanomedicine - Selenocysteine based on these integrative data. Conclusion We identified eight hub genes that could work as prospective biomarkers for the diagnostic and biomaterial drug treatment of cartilage lesion, involving the novel genes CAMP, DEFA3, TOLLIP, HLA-DQA2, SLC38A6, SLC3A1, FAM20A, and ANO8. Meanwhile, these genes were mainly associated with immune response, immune mediator induction, and cell chemotaxis. Significant support is provided for obtaining a series of novel gene targets, and we identify potential mechanisms for cartilage regeneration and final nanomedicine immunotherapy in regenerative medicine.
Collapse
Affiliation(s)
- Jing Ye
- Knee Surgery Department of the Institution of Sports Medicine, Peking University Third Hospital, Beijing Key Laboratory of Sports Injuries, Beijing, China
| | - Bingbing Xu
- Knee Surgery Department of the Institution of Sports Medicine, Peking University Third Hospital, Beijing Key Laboratory of Sports Injuries, Beijing, China
| | - Baoshi Fan
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Jiying Zhang
- Knee Surgery Department of the Institution of Sports Medicine, Peking University Third Hospital, Beijing Key Laboratory of Sports Injuries, Beijing, China
| | - Fuzhen Yuan
- Knee Surgery Department of the Institution of Sports Medicine, Peking University Third Hospital, Beijing Key Laboratory of Sports Injuries, Beijing, China
| | - Yourong Chen
- Knee Surgery Department of the Institution of Sports Medicine, Peking University Third Hospital, Beijing Key Laboratory of Sports Injuries, Beijing, China
| | - Zewen Sun
- Knee Surgery Department of the Institution of Sports Medicine, Peking University Third Hospital, Beijing Key Laboratory of Sports Injuries, Beijing, China
| | - Xin Yan
- Knee Surgery Department of the Institution of Sports Medicine, Peking University Third Hospital, Beijing Key Laboratory of Sports Injuries, Beijing, China
| | - Yifan Song
- Knee Surgery Department of the Institution of Sports Medicine, Peking University Third Hospital, Beijing Key Laboratory of Sports Injuries, Beijing, China
| | - Shitang Song
- Knee Surgery Department of the Institution of Sports Medicine, Peking University Third Hospital, Beijing Key Laboratory of Sports Injuries, Beijing, China
| | - Meng Yang
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Jia-Kuo Yu
- Knee Surgery Department of the Institution of Sports Medicine, Peking University Third Hospital, Beijing Key Laboratory of Sports Injuries, Beijing, China
| |
Collapse
|
20
|
Baer B, Veldhuizen EJA, Molchanova N, Jekhmane S, Weingarth M, Jenssen H, Lin JS, Barron AE, Yamashita C, Veldhuizen R. Optimizing Exogenous Surfactant as a Pulmonary Delivery Vehicle for Chicken Cathelicidin-2. Sci Rep 2020; 10:9392. [PMID: 32523049 PMCID: PMC7287084 DOI: 10.1038/s41598-020-66448-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/12/2020] [Indexed: 11/09/2022] Open
Abstract
The rising incidence of antibiotic-resistant lung infections has instigated a much-needed search for new therapeutic strategies. One proposed strategy is the use of exogenous surfactants to deliver antimicrobial peptides (AMPs), like CATH-2, to infected regions of the lung. CATH-2 can kill bacteria through a diverse range of antibacterial pathways and exogenous surfactant can improve pulmonary drug distribution. Unfortunately, mixing AMPs with commercially available exogenous surfactants has been shown to negatively impact their antimicrobial function. It was hypothesized that the phosphatidylglycerol component of surfactant was inhibiting AMP function and that an exogenous surfactant, with a reduced phosphatidylglycerol composition would increase peptide mediated killing at a distal site. To better understand how surfactant lipids interacted with CATH-2 and affected its function, isothermal titration calorimetry and solid-state nuclear magnetic resonance spectroscopy as well as bacterial killing curves against Pseudomonas aeruginosa were utilized. Additionally, the wet bridge transfer system was used to evaluate surfactant spreading and peptide transport. Phosphatidylglycerol was the only surfactant lipid to significantly inhibit CATH-2 function, showing a stronger electrostatic interaction with the peptide than other lipids. Although diluting the phosphatidylglycerol content in an existing surfactant, through the addition of other lipids, significantly improved peptide function and distal killing, it also reduced surfactant spreading. A synthetic phosphatidylglycerol-free surfactant however, was shown to further improve CATH-2 delivery and function at a remote site. Based on these in vitro experiments synthetic phosphatidylglycerol-free surfactants seem optimal for delivering AMPs to the lung.
Collapse
Affiliation(s)
- Brandon Baer
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada.
| | - Edwin J A Veldhuizen
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Natalia Molchanova
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
- Division of Biological Nanostructures, The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Shehrazade Jekhmane
- Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Utrecht, The Netherlands
| | - Markus Weingarth
- Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Utrecht, The Netherlands
| | - Håvard Jenssen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Jennifer S Lin
- Department of Bioengineering, School of Medicine & School of Engineering, Stanford University, Stanford, California, USA
| | - Annelise E Barron
- Department of Bioengineering, School of Medicine & School of Engineering, Stanford University, Stanford, California, USA
| | - Cory Yamashita
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
- Department of Medicine, Western University, London, Ontario, Canada
| | - Ruud Veldhuizen
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
- Department of Medicine, Western University, London, Ontario, Canada
| |
Collapse
|
21
|
Scheenstra MR, van Harten RM, Veldhuizen EJA, Haagsman HP, Coorens M. Cathelicidins Modulate TLR-Activation and Inflammation. Front Immunol 2020; 11:1137. [PMID: 32582207 PMCID: PMC7296178 DOI: 10.3389/fimmu.2020.01137] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/11/2020] [Indexed: 12/30/2022] Open
Abstract
Cathelicidins are short cationic peptides that are part of the innate immune system. At first, these peptides were studied mostly for their direct antimicrobial killing capacity, but nowadays they are more and more appreciated for their immunomodulatory functions. In this review, we will provide a comprehensive overview of the various effects cathelicidins have on the detection of damage- and microbe-associated molecular patterns, with a special focus on their effects on Toll-like receptor (TLR) activation. We review the available literature based on TLR ligand types, which can roughly be divided into lipidic ligands, such as LPS and lipoproteins, and nucleic-acid ligands, such as RNA and DNA. For both ligand types, we describe how direct cathelicidin-ligand interactions influence TLR activation, by for instance altering ligand stability, cellular uptake and receptor interaction. In addition, we will review the more indirect mechanisms by which cathelicidins affect downstream TLR-signaling. To place all this information in a broader context, we discuss how these cathelicidin-mediated effects can have an impact on how the host responds to infectious organisms as well as how these effects play a role in the exacerbation of inflammation in auto-immune diseases. Finally, we discuss how these immunomodulatory activities can be exploited in vaccine development and cancer therapies.
Collapse
Affiliation(s)
- Maaike R Scheenstra
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Roel M van Harten
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Edwin J A Veldhuizen
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Henk P Haagsman
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Maarten Coorens
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Laboratory, Stockholm, Sweden
| |
Collapse
|
22
|
Kang MJ, Jang AR, Park JY, Ahn JH, Lee TS, Kim DY, Jung DH, Song EJ, Hong JJ, Park JH. Cathelicidin-related Antimicrobial Peptide Contributes to Host Immune Responses Against Pulmonary Infection with Acinetobacter baumannii in Mice. Immune Netw 2020; 20:e25. [PMID: 32655973 PMCID: PMC7327154 DOI: 10.4110/in.2020.20.e25] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022] Open
Abstract
Acinetobacter baumannii is known for its multidrug antibiotic resistance. New approaches to treating drug-resistant bacterial infections are urgently required. Cathelicidin-related antimicrobial peptide (CRAMP) is a murine antimicrobial peptide that exerts diverse immune functions, including both direct bacterial cell killing and immunomodulatory effects. In this study, we sought to identify the role of CRAMP in the host immune response to multidrug-resistant Acinetobacter baumannii. Wild-type (WT) and CRAMP knockout mice were infected intranasally with the bacteria. CRAMP−/− mice exhibited increased bacterial colony-forming units (CFUs) in bronchoalveolar lavage (BAL) fluid after A. baumannii infection compared to WT mice. The loss of CRAMP expression resulted in a significant decrease in the recruitment of immune cells, primarily neutrophils. The levels of IL-6 and CXCL1 were lower, whereas the levels of IL-10 were significantly higher in the BAL fluid of CRAMP−/− mice compared to WT mice 1 day after infection. In an in vitro assay using thioglycollate-induced peritoneal neutrophils, the ability of bacterial phagocytosis and killing was impaired in CRAMP−/− neutrophils compared to the WT cells. CRAMP was also essential for the production of cytokines and chemokines in response to A. baumannii in neutrophils. In addition, the A. baumannii-induced inhibitor of κB-α degradation and phosphorylation of p38 MAPK were impaired in CRAMP−/− neutrophils, whereas ERK and JNK phosphorylation was upregulated. Our results indicate that CRAMP plays an important role in the host defense against pulmonary infection with A. baumannii by promoting the antibacterial activity of neutrophils and regulating the innate immune responses.
Collapse
Affiliation(s)
- Min-Jung Kang
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Ah-Ra Jang
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Ji-Yeon Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Jae-Hun Ahn
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Tae-Sung Lee
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Dong-Yeon Kim
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Do-Hyeon Jung
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Eun-Jung Song
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Jung Joo Hong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk 28116, Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| |
Collapse
|
23
|
Mookherjee N, Anderson MA, Haagsman HP, Davidson DJ. Antimicrobial host defence peptides: functions and clinical potential. Nat Rev Drug Discov 2020; 19:311-332. [DOI: 10.1038/s41573-019-0058-8] [Citation(s) in RCA: 425] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2019] [Indexed: 12/18/2022]
|
24
|
Zhou Y, Shi Y, Yang L, Sun Y, Han Y, Zhao Z, Wang Y, Liu Y, Ma Y, Zhang T, Ren T, Dale TP, Forsyth NR, Jin F, Qu J, Zuo W, Xu J. Genetically engineered distal airway stem cell transplantation protects mice from pulmonary infection. EMBO Mol Med 2020; 12:e10233. [PMID: 31782624 PMCID: PMC6949487 DOI: 10.15252/emmm.201810233] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 10/26/2019] [Accepted: 11/04/2019] [Indexed: 12/31/2022] Open
Abstract
Severe pulmonary infection is a major threat to human health accompanied by substantial medical costs, prolonged inpatient requirements, and high mortality rates. New antimicrobial therapeutic strategies are urgently required to address the emergence of antibiotic resistance and persistent bacterial infections. In this study, we show that the constitutive expression of a native antimicrobial peptide LL-37 in transgenic mice aids in clearing Pseudomonas aeruginosa (PAO1), a major pathogen of clinical pulmonary infection. Orthotopic transplantation of adult mouse distal airway stem cells (DASCs), genetically engineered to express LL-37, into injured mouse lung foci enabled large-scale incorporation of cells and long-term release of the host defense peptide, protecting the mice from bacterial pneumonia and hypoxemia. Further, correlates of DASCs in adult humans were isolated, expanded, and genetically engineered to demonstrate successful construction of an anti-infective artificial lung. Together, our stem cell-based gene delivery therapeutic platform proposes a new strategy for addressing recurrent pulmonary infections with future translational opportunities.
Collapse
Affiliation(s)
- Yue‐qing Zhou
- Department of Respiratory and Critical Care MedicineClinical Translation Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Yun Shi
- Shanghai East HospitalTongji University School of MedicineShanghaiChina
- Department of Respiratory and Critical Care MedicineTangdu HospitalFourth Military Medical University of PLAXi'anChina
| | - Ling Yang
- Department of Respiratory and Critical Care MedicineClinical Translation Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Yu‐fen Sun
- Department of Respiratory and Critical Care MedicineClinical Translation Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Yu‐fei Han
- Department of Respiratory and Critical Care MedicineClinical Translation Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Zi‐xian Zhao
- Department of Respiratory and Critical Care MedicineClinical Translation Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Yu‐jia Wang
- Department of Respiratory and Critical Care MedicineClinical Translation Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Ying Liu
- Shanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Yu Ma
- Shanghai East HospitalTongji University School of MedicineShanghaiChina
- Regend Therapeutics Co. LtdZhejiangChina
| | - Ting Zhang
- Regend Therapeutics Co. LtdZhejiangChina
| | - Tao Ren
- Shanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Tina P Dale
- Guy Hilton Research CenterSchool of Pharmacy and BioengineeringKeele UniversityStaffordshireUK
| | - Nicholas R Forsyth
- Guy Hilton Research CenterSchool of Pharmacy and BioengineeringKeele UniversityStaffordshireUK
| | - Fa‐guang Jin
- Department of Respiratory and Critical Care MedicineTangdu HospitalFourth Military Medical University of PLAXi'anChina
| | - Jie‐ming Qu
- Ruijin HospitalShanghai Jiaotong University School of MedicineShanghaiChina
- Institute of Respiratory DiseasesShanghai Jiaotong University School of MedicineShanghaiChina
| | - Wei Zuo
- Department of Respiratory and Critical Care MedicineClinical Translation Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
- Shanghai East HospitalTongji University School of MedicineShanghaiChina
- Regend Therapeutics Co. LtdZhejiangChina
- Guangzhou Institute of Respiratory DiseaseThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Ningxia Medical UniversityYinchuanChina
| | - Jin‐fu Xu
- Department of Respiratory and Critical Care MedicineClinical Translation Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| |
Collapse
|
25
|
Burkes RM, Ceppe AS, Couper DJ, Comellas AP, Wells JM, Peters SP, Criner GJ, Kanner RE, Paine R, Christenson SA, Cooper CB, Barjaktarevic IZ, Krishnan JA, Labaki WW, Han MK, Curtis JL, Hansel NN, Wise RA, Drummond MB. Plasma Cathelicidin is Independently Associated with Reduced Lung Function in COPD: Analysis of the Subpopulations and Intermediate Outcome Measures in COPD Study Cohort. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2020; 7:370-381. [PMID: 33108110 DOI: 10.15326/jcopdf.7.4.2020.0142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ratrionale The antimicrobial peptide cathelicidin, also known in humans as LL-37, is a defensin secreted by immune and airway epithelial cells. Deficiencies in this peptide may contribute to adverse pulmonary outcomes in chronic obstructive pulmonary disease (COPD). Objectives Using clinical and biological samples from the SubPopulations and InteRmediate Outcome Measures In COPD Study (SPIROMICS), we assessed the associations of plasma cathelicidin levels with cross-sectional and longitudinal COPD outcomes. Methods A total of 1609 SPIROMICS participants with COPD and available plasma samples were analyzed. Cathelicidin was modeled dichotomously (lowest quartile [< 50 ng/ml] versus highest 75% [≥ 50 ng/ml]) and continuously per 10 ng/ml. Fixed-effect multilevel regression analyses were used to assess associations between cathelicidin and cross-sectional as well as longitudinal lung function. The associations between cathelicidin and participant-reported retrospective and prospective COPD exacerbations were assessed via logistic regression. Measurements and Main Results Cathelicidin < 50 ng/ml (N=383) was associated with female sex, black race, and lower body mass index (BMI).At baseline,cathelicidin < 50 ng/ml was independently associated with 3.55% lower % predicted forced expiratory volume in 1 second (FEV1)(95% confidence interval [CI] -6.22% to -0.88% predicted; p=0.01), while every 10 ng/ml lower cathelicidin was independently associated with 0.65% lower % predicted FEV1 (95% CI -1.01% to -0.28% predicted; p< 0.001). No independent associations with longitudinal lung function decline or participant-reported COPD exacerbations were observed. Conclusions Reduced cathelicidin is associated with lower lung function at baseline. Plasma cathelicidin may potentially identify COPD patients at increased risk for more severe lung disease.
Collapse
Affiliation(s)
- Robert M Burkes
- Division of Pulmonary Diseases and Critical Care Medicine, University of North Carolina, Chapel Hill
| | - Agathe S Ceppe
- Marsico Lung Institute, University of North Carolina, Chapel Hill
| | - David J Couper
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill
| | - Alejandro P Comellas
- Division of Pulmonary, Critical Care, and Occupational Medicine, Carver College of Medicine, University of Iowa, Iowa City
| | - J Michael Wells
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama, Birmingham
| | - Stephen P Peters
- Section of Pulmonary, Critical Care, Allergy, and Immunologic Disease, Wake Forest University, Winston-Salem, North Carolina
| | - Gerard J Criner
- Division of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania
| | - Richard E Kanner
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City
| | - Robert Paine
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City
| | - Stephanie A Christenson
- Division of Pulmonary Critical Care, Allergy, and Sleep Medicine, University of California, San Francisco
| | - Christopher B Cooper
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Igor Z Barjaktarevic
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Jerry A Krishnan
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, University of Illinois, Chicago
| | - Wassim W Labaki
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor
| | - MeiLan K Han
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor
| | - Jeffrey L Curtis
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor.,Medicine Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Nadia N Hansel
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University Baltimore, Maryland
| | - Robert A Wise
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University Baltimore, Maryland
| | - M Bradley Drummond
- Division of Pulmonary Diseases and Critical Care Medicine, University of North Carolina, Chapel Hill.,Marsico Lung Institute, University of North Carolina, Chapel Hill
| | | |
Collapse
|
26
|
He L, Li F, Yin X, Bohman P, Kim S, McClain CJ, Feng W, Zhang X. Profiling of Polar Metabolites in Mouse Feces Using Four Analytical Platforms to Study the Effects Of Cathelicidin-Related Antimicrobial Peptide in Alcoholic Liver Disease. J Proteome Res 2019; 18:2875-2884. [PMID: 31188604 PMCID: PMC6748645 DOI: 10.1021/acs.jproteome.9b00181] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alterations in gut bacterial homeostasis result in changes in intestinal metabolites. To investigate the effects of alcohol on fecal metabolites and the role of cathelicidin-related antimicrobial peptide (CRAMP) in alcoholic liver disease (ALD), CRAMP knockout (KO) and their control wild type (WT) mice were fed a Lieber-DeCarli liquid diet with or without alcohol. Polar metabolites in mouse feces were analyzed by GC × GC-MS and 2DLC-MS, and the concentrations of short chain fatty acids (SCFAs) were measured by GC-MS. A total of 95 and 190 metabolites were detected by GC × GC-MS and 2DLC-MS, respectively. Among the significantly changed metabolites, taurine and nicotinic acid were decreased in WT mice fed alcohol, which were also down-regulated in KO mice fed without alcohol. Interestingly, these two metabolites were increased in KO mice fed alcohol compared to them in WT controls. Additionally, SCFAs were significantly decreased in WT mice fed alcohol and in KO mice fed without alcohol, whereas two branched-chain SCFAs were increased by alcohol treatment in KO mice. In summary, the analytical platforms employed in this study successfully dissected the alterations of polar metabolites and SCFAs in fecal samples, which helped understand the effects of alcohol consumption and CRAMP in intestinal metabolism and alcohol-induced liver injury.
Collapse
Affiliation(s)
- Liqing He
- Department of Chemistry, University of Louisville, Louisville, Kentucky 40208, United States
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40208, United States
- University of Louisville Hepatobiology & Toxicology Center, University of Louisville, Louisville, Kentucky 40208, United States
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, Kentucky 40208, United States
| | - Fengyuan Li
- University of Louisville Hepatobiology & Toxicology Center, University of Louisville, Louisville, Kentucky 40208, United States
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, Kentucky 40208, United States
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, Kentucky 40208, United States
| | - Xinmin Yin
- Department of Chemistry, University of Louisville, Louisville, Kentucky 40208, United States
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40208, United States
- University of Louisville Hepatobiology & Toxicology Center, University of Louisville, Louisville, Kentucky 40208, United States
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, Kentucky 40208, United States
| | - Patrick Bohman
- Thermo Fisher Scientific International Inc., 3000 Lakeside Dr., Bannockburn, Illinois 60015, United States
| | - Seongho Kim
- Biostatistics Core, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan 48201, United States
| | - Craig J. McClain
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40208, United States
- University of Louisville Hepatobiology & Toxicology Center, University of Louisville, Louisville, Kentucky 40208, United States
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, Kentucky 40208, United States
- Department of Medicine, University of Louisville, Louisville, Kentucky 40208, United States
- Robley Rex Louisville VAMC, Louisville, Kentucky 40292, United States
| | - Wenke Feng
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40208, United States
- University of Louisville Hepatobiology & Toxicology Center, University of Louisville, Louisville, Kentucky 40208, United States
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, Kentucky 40208, United States
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, Kentucky 40208, United States
- Department of Medicine, University of Louisville, Louisville, Kentucky 40208, United States
| | - Xiang Zhang
- Department of Chemistry, University of Louisville, Louisville, Kentucky 40208, United States
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40208, United States
- University of Louisville Hepatobiology & Toxicology Center, University of Louisville, Louisville, Kentucky 40208, United States
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, Kentucky 40208, United States
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, Kentucky 40208, United States
| |
Collapse
|
27
|
Myszor IT, Parveen Z, Ottosson H, Bergman P, Agerberth B, Strömberg R, Gudmundsson GH. Novel aroylated phenylenediamine compounds enhance antimicrobial defense and maintain airway epithelial barrier integrity. Sci Rep 2019; 9:7114. [PMID: 31068616 PMCID: PMC6506505 DOI: 10.1038/s41598-019-43350-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/18/2019] [Indexed: 01/19/2023] Open
Abstract
Aroylated phenylenediamines (APDs) are novel inducers of innate immunity enhancing cathelicidin gene expression in human bronchial epithelial cell lines. Here we present two newly developed APDs and aimed at defining the response and signaling pathways for these compounds with reference to innate immunity and antimicrobial peptide (AMP) expression. Induction was initially defined with respect to dose and time and compared with the APD Entinostat (MS-275). The induction applies to several innate immunity effectors, indicating that APDs trigger a broad spectrum of antimicrobial responses. The bactericidal effect was shown in an infection model against Pseudomonas aeruginosa by estimating bacteria entering cells. Treatment with a selected APD counteracted Pseudomonas mediated disruption of epithelial integrity. This double action by inducing AMPs and enhancing epithelial integrity for one APD compound is unique and taken as a positive indication for host directed therapy (HDT). The APD effects are mediated through Signal transducer and activator of transcription 3 (STAT3) activation. Utilization of induced innate immunity to fight infections can reduce antibiotic usage, might be effective against multidrug resistant bacteria and is in line with improved stewardship in healthcare.
Collapse
Affiliation(s)
- Iwona T Myszor
- Biomedical Center, University of Iceland, Reykjavik, 101, Iceland
| | - Zahida Parveen
- Biomedical Center, University of Iceland, Reykjavik, 101, Iceland
| | - Håkan Ottosson
- Department of Biosciences and Nutrition, Karolinska Institutet, S-14183, Huddinge, Sweden
| | - Peter Bergman
- Department of Laboratory Medicine, Clinical Microbiology, Karolinska Institutet, S-14186, Huddinge, Sweden
| | - Birgitta Agerberth
- Department of Laboratory Medicine, Clinical Microbiology, Karolinska Institutet, S-14186, Huddinge, Sweden
| | - Roger Strömberg
- Department of Biosciences and Nutrition, Karolinska Institutet, S-14183, Huddinge, Sweden
| | | |
Collapse
|
28
|
McHugh BJ, Wang R, Li HN, Beaumont PE, Kells R, Stevens H, Young L, Rossi AG, Gray RD, Dorin JR, Gwyer Findlay EL, Brough D, Davidson DJ. Cathelicidin is a "fire alarm", generating protective NLRP3-dependent airway epithelial cell inflammatory responses during infection with Pseudomonas aeruginosa. PLoS Pathog 2019; 15:e1007694. [PMID: 30978238 PMCID: PMC6481867 DOI: 10.1371/journal.ppat.1007694] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 04/24/2019] [Accepted: 03/11/2019] [Indexed: 01/23/2023] Open
Abstract
Pulmonary infections are a major global cause of morbidity, exacerbated by an increasing threat from antibiotic-resistant pathogens. In this context, therapeutic interventions aimed at protectively modulating host responses, to enhance defence against infection, take on ever greater significance. Pseudomonas aeruginosa is an important multidrug-resistant, opportunistic respiratory pathogen, the clearance of which can be enhanced in vivo by the innate immune modulatory properties of antimicrobial host defence peptides from the cathelicidin family, including human LL-37. Initially described primarily as bactericidal agents, cathelicidins are now recognised as multifunctional antimicrobial immunomodulators, modifying host responses to pathogens, but the key mechanisms involved in these protective functions are not yet defined. We demonstrate that P. aeruginosa infection of airway epithelial cells promotes extensive infected cell internalisation of LL-37, in a manner that is dependent upon epithelial cell interaction with live bacteria, but does not require bacterial Type 3 Secretion System (T3SS). Internalised LL-37 acts as a second signal to induce inflammasome activation in airway epithelial cells, which, in contrast to myeloid cells, are relatively unresponsive to P. aeruginosa. We demonstrate that this is mechanistically dependent upon cathepsin B release, and NLRP3-dependent activation of caspase 1. These result in LL-37-mediated release of IL-1β and IL-18 in a manner that is synergistic with P. aeruginosa infection, and can induce caspase 1-dependent death of infected epithelial cells, and promote neutrophil chemotaxis. We propose that cathelicidin can therefore act as a second signal, required by P. aeruginosa infected epithelial cells to promote an inflammasome-mediated altruistic cell death of infection-compromised epithelial cells and act as a "fire alarm" to enhance rapid escalation of protective inflammatory responses to an uncontrolled infection. Understanding this novel modulatory role for cathelicidins, has the potential to inform development of novel therapeutic strategies to antibiotic-resistant pathogens, harnessing innate immunity as a complementation or alternative to current interventions.
Collapse
Affiliation(s)
- Brian J. McHugh
- Centre for Inflammation Research at the University of Edinburgh, Queens’ Medical Research Institute, Edinburgh BioQuarter, Little France Crescent, Edinburgh, United Kingdom
| | - Rongling Wang
- Centre for Inflammation Research at the University of Edinburgh, Queens’ Medical Research Institute, Edinburgh BioQuarter, Little France Crescent, Edinburgh, United Kingdom
| | - Hsin-Ni Li
- Centre for Inflammation Research at the University of Edinburgh, Queens’ Medical Research Institute, Edinburgh BioQuarter, Little France Crescent, Edinburgh, United Kingdom
| | - Paula E. Beaumont
- Centre for Inflammation Research at the University of Edinburgh, Queens’ Medical Research Institute, Edinburgh BioQuarter, Little France Crescent, Edinburgh, United Kingdom
| | - Rebekah Kells
- Centre for Inflammation Research at the University of Edinburgh, Queens’ Medical Research Institute, Edinburgh BioQuarter, Little France Crescent, Edinburgh, United Kingdom
| | - Holly Stevens
- Centre for Inflammation Research at the University of Edinburgh, Queens’ Medical Research Institute, Edinburgh BioQuarter, Little France Crescent, Edinburgh, United Kingdom
| | - Lisa Young
- Centre for Inflammation Research at the University of Edinburgh, Queens’ Medical Research Institute, Edinburgh BioQuarter, Little France Crescent, Edinburgh, United Kingdom
| | - Adriano G. Rossi
- Centre for Inflammation Research at the University of Edinburgh, Queens’ Medical Research Institute, Edinburgh BioQuarter, Little France Crescent, Edinburgh, United Kingdom
| | - Robert D. Gray
- Centre for Inflammation Research at the University of Edinburgh, Queens’ Medical Research Institute, Edinburgh BioQuarter, Little France Crescent, Edinburgh, United Kingdom
| | - Julia R. Dorin
- Centre for Inflammation Research at the University of Edinburgh, Queens’ Medical Research Institute, Edinburgh BioQuarter, Little France Crescent, Edinburgh, United Kingdom
| | - Emily L. Gwyer Findlay
- Centre for Inflammation Research at the University of Edinburgh, Queens’ Medical Research Institute, Edinburgh BioQuarter, Little France Crescent, Edinburgh, United Kingdom
| | - David Brough
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Donald J. Davidson
- Centre for Inflammation Research at the University of Edinburgh, Queens’ Medical Research Institute, Edinburgh BioQuarter, Little France Crescent, Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
29
|
Liu C, Yang L, Han Y, Ouyang W, Yin W, Xu F. Mast cells participate in regulation of lung-gut axis during Staphylococcus aureus pneumonia. Cell Prolif 2019; 52:e12565. [PMID: 30729611 PMCID: PMC6496676 DOI: 10.1111/cpr.12565] [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: 09/20/2018] [Revised: 11/15/2018] [Accepted: 11/27/2018] [Indexed: 12/18/2022] Open
Abstract
Objectives The lung‐gut axis is known to be involved in the pathogenesis of Staphylococcus aureus pneumonia. However, the underlying mechanisms remain unclear. We examined the role of pulmonary mast cells (MCs) in the regulation of the lung‐gut axis during S. aureus pneumonia. Materials and Methods We created a mouse model of S. aureus pneumonia using MC‐deficient mice (KitW‐sh/W‐sh) and examined the level of inflammation, bacterial burden, expression of cathelicidin‐related antimicrobial peptide (CRAMP) and composition of the gut microbiota. We further evaluated anti‐bacterial immunity by administering bone marrow MCs (BMMCs) or CRAMP into the lungs of KitW‐sh/W‐sh mice. Results After S. aureus challenge, the MC‐deficient mice, compared with wild‐type (WT) mice, displayed attenuated lung inflammation, decreased expression of CRAMP, higher bacterial lung load and disturbance of the intestinal microbiota. Adoptive transfer of BMMCs into the lung effectively reconstituted the host defence against S. aureus in KitW‐sh/W‐sh mice, thus resulting in recovery of S. aureus pneumonia‐induced intestinal dysfunction. Similarly, exogenous administration of CRAMP significantly enhanced anti‐bacterial immunity in the lungs of MC‐deficient mice. Conclusions This study provides evidence for the involvement of MCs in the regulation of the lung‐gut axis during S. aureus pneumonia.
Collapse
Affiliation(s)
- Chao Liu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liping Yang
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Han
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Ouyang
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Yin
- Core Facilities, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Xu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
30
|
Haney EF, Straus SK, Hancock REW. Reassessing the Host Defense Peptide Landscape. Front Chem 2019; 7:43. [PMID: 30778385 PMCID: PMC6369191 DOI: 10.3389/fchem.2019.00043] [Citation(s) in RCA: 237] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/15/2019] [Indexed: 12/18/2022] Open
Abstract
Current research has demonstrated that small cationic amphipathic peptides have strong potential not only as antimicrobials, but also as antibiofilm agents, immune modulators, and anti-inflammatories. Although traditionally termed antimicrobial peptides (AMPs) these additional roles have prompted a shift in terminology to use the broader term host defense peptides (HDPs) to capture the multi-functional nature of these molecules. In this review, we critically examined the role of AMPs and HDPs in infectious diseases and inflammation. It is generally accepted that HDPs are multi-faceted mediators of a wide range of biological processes, with individual activities dependent on their polypeptide sequence. In this context, we explore the concept of chemical space as it applies to HDPs and hypothesize that the various functions and activities of this class of molecule exist on independent but overlapping activity landscapes. Finally, we outline several emerging functions and roles of HDPs and highlight how an improved understanding of these processes can potentially be leveraged to more fully realize the therapeutic promise of HDPs.
Collapse
Affiliation(s)
- Evan F Haney
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Suzana K Straus
- Department of Chemistry, 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
|
31
|
Huynh E, Akhtar N, Li J. Efficient Production of Recombinant Protegrin-1 From Pichia pastoris, and Its Antimicrobial and in vitro Cell Migration Activity. Front Microbiol 2018; 9:2300. [PMID: 30319593 PMCID: PMC6170612 DOI: 10.3389/fmicb.2018.02300] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 09/10/2018] [Indexed: 01/08/2023] Open
Abstract
Protegrin (PG) belongs to the antimicrobial peptide cathelicidin family. To date, five protegrin sequences have been identified in pigs, PG-1 to PG-5. Of these, PG-1 exhibits potent antimicrobial activity against a broad range of antibiotic-resistant microorganisms as well as viruses. However, the other potential role(s) of PG beyond antimicrobial has largely been unexplored. The aim of this study was to use nonpathogenic yeast Pichia pastoris to express antimicrobially active recombinant protegrin (rPG-1). Additionally, the effect of PG-1 on cell migration and proliferation was also examined in vitro using pig intestinal epithelial cells as a model. Highest level of rPG-1 (104 ± 11 μg/mL) was detected at 24 h in fermentation culture medium. Similar to rPG-1, 0.8 ± 0.10 g/L of proform PG-1 (rProPG-1) and 0.2 ± 0.02 g/L of the PG-1 cathelin domain (rCath) was detected in fermentation culture medium. Resulting recombinant PG-1 and cleaved rProPG-1 exerted antimicrobial activity against Escherichia coli DH5α at the same level as chemically synthesized PG-1. Enhanced cell migration was observed (p < 0.05) in groups treated with rProPG-1, rCath, and rPG-1 compared to the control. Furthermore, rPG-1 was stable at temperatures ranging from 25°C to 80°C. In summary, biologically active recombinant protegrin in its pro-, cathelin-, and mature- forms were successfully expressed in P. pastoris suggesting potential feasibility for future therapeutic applications.
Collapse
Affiliation(s)
- Evanna Huynh
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Nadeem Akhtar
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Julang Li
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
- College of Life Science and Engineering, Foshan University, Foshan, China
| |
Collapse
|
32
|
Skerrett SJ, Braff MH, Liggitt HD, Rubens CE. Toll-like receptor 2 has a prominent but nonessential role in innate immunity to Staphylococcus aureus pneumonia. Physiol Rep 2018; 5:5/21/e13491. [PMID: 29142002 PMCID: PMC5688782 DOI: 10.14814/phy2.13491] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/08/2017] [Accepted: 10/11/2017] [Indexed: 11/30/2022] Open
Abstract
Staphylococcus aureus is an important cause of acute bacterial pneumonia. Toll‐like receptor 2 (TLR2) recognizes multiple components of the bacterial cell wall and activates innate immune responses to gram‐positive bacteria. We hypothesized that TLR2 would have an important role in pulmonary host defense against S. aureus. TLR null (TLR2−/−) mice and wild type (WT) C57BL/6 controls were challenged with aerosolized S. aureus at a range of inocula for kinetic studies of cytokine and antimicrobial peptide expression, lung inflammation, bacterial killing by alveolar macrophages, and bacterial clearance. Survival was measured after intranasal infection. Pulmonary induction of most pro‐inflammatory cytokines was significantly blunted in TLR2−/− mice 4 and 24 h after infection in comparison with WT controls. Bronchoalveolar concentrations of cathelicidin‐related antimicrobial peptide also were reduced in TLR2−/− mice. Lung inflammation, measured by enumeration of bronchoalveolar neutrophils and scoring of histological sections, was significantly blunted in TLR2−/− mice. Phagocytosis of S. aureus by alveolar macrophages in vivo after low‐dose infection was unimpaired, but viability of ingested bacteria was significantly greater in TLR2−/− mice. Bacterial clearance from the lungs was slightly impaired in TLR2−/− mice after low‐dose infection only; bacterial elimination from the lungs was slightly accelerated in the TLR2−/− mice after high‐dose infection. Survival after high‐dose intranasal challenge was 50–60% in both groups. TLR2 has a significant role in early innate immune responses to S. aureus in the lungs but is not required for bacterial clearance and survival from S. aureus pneumonia.
Collapse
Affiliation(s)
- Shawn J Skerrett
- Department of Medicine, University of Washington, Seattle, Washington
| | - Marissa H Braff
- Seattle Children's Hospital Research Institute, Seattle, Washington
| | - H Denny Liggitt
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Craig E Rubens
- Seattle Children's Hospital Research Institute, Seattle, Washington
| |
Collapse
|
33
|
Cuperus T, Kraaij MD, Zomer AL, van Dijk A, Haagsman HP. Immunomodulation and effects on microbiota after in ovo administration of chicken cathelicidin-2. PLoS One 2018; 13:e0198188. [PMID: 29870564 PMCID: PMC5988267 DOI: 10.1371/journal.pone.0198188] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 05/15/2018] [Indexed: 01/06/2023] Open
Abstract
Host Defense Peptides (HDPs) such as cathelicidins are multifunctional effectors of the innate immune system with both antimicrobial and pleiotropic immunomodulatory functions. Chicken cathelicidin-2 (CATH-2) has multiple immunomodulatory effects in vitro and the D-amino acid analog of this peptide has been shown to partially protect young chicks from a bacterial infection. However, the mechanisms responsible for CATH-2 mediated in vivo protection have not been investigated so far. In this study, D-CATH-2 was administered in ovo and the immune status and microbiota of the chicks were investigated at 7 days posthatch to elucidate the in vivo mechanisms of the peptide. In three consecutive studies, no effects on numbers and functions of immune cells were found and only small changes were seen in gene expression of Peripheral Blood Mononuclear Cells (PBMCs). In two studies, intestinal microbiota composition was determined which was highly variable, suggesting that it was strongly influenced by environmental factors. In both studies, in ovo D-CATH-2 treatment caused significant reduction of Ruminococcaceae and Butyricicoccus in the cecum and Escherichia/Shigella in both ileum and cecum. In conclusion, this study shows that, in the absence of an infectious stimulus, in ovo administration of a CATH-2 analog alters the microbiota composition but does not affect the chicks' immune system posthatch.
Collapse
Affiliation(s)
- Tryntsje Cuperus
- Division of Molecular Host Defence, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Marina D. Kraaij
- Division of Molecular Host Defence, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Aldert L. Zomer
- Division Clinical Infectiology, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Albert van Dijk
- Division of Molecular Host Defence, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Henk P. Haagsman
- Division of Molecular Host Defence, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
34
|
Zhao L, Tan S, Zhang H, Liu P, Tan YZ, Li JC, Jia D, Shen XF. Astragalus polysaccharides exerts anti-infective activity by inducing human cathelicidin antimicrobial peptide LL-37 in respiratory epithelial cells. Phytother Res 2018; 32:1521-1529. [PMID: 29672953 DOI: 10.1002/ptr.6080] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/05/2018] [Accepted: 03/06/2018] [Indexed: 12/30/2022]
Abstract
Astragalus polysaccharides (APS), one of the major active components in Astragalus membranaceus, is an effective immunomodulator used in the treatment of immunological diseases in China. However, the anti-infective action and mechanism of APS is not fully known. In the present study, we found that APS induced the expression of human cathelicidin antimicrobial peptide LL-37, a key host anti-infective molecule, in both mRNA and protein levels in respiratory epithelial cells HBE16 and A549. Furthermore, the lysate and supernatant from APS-treated HBE16 cells both exhibited an obvious antibacterial action, which was partially neutralizated by LL-37 monoclonal antibody. In addition, APS also significantly elevated the phosphorylation of p38 MAPK and JNK and caused the degradation of IκBα. Specific inhibitors of p38 MAPK, JNK, or NF-κB obviously abolished APS-induced LL-37 synthesis and antibacterial activity, respectively. Taken together, our results confirmed the enhancement of APS on LL-37 induction and antibacterial action in respiratory epithelial cells, which may be attributed to activation of p38 MAPK/JNK and NF-κB pathways. Furthermore, these results also supported the clinical application of APS in the treatment of infectious diseases.
Collapse
Affiliation(s)
- Lin Zhao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shuai Tan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hai Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peng Liu
- Shaanxi TASLY Plant Pharmaceutical Co Ltd., Shangluo, Shaanxi, China
| | - Yu-Zhu Tan
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia-Chuan Li
- School of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiao-Fei Shen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
35
|
Sham HP, Walker KH, Abdulnour REE, Krishnamoorthy N, Douda DN, Norris PC, Barkas I, Benito-Figueroa S, Colby JK, Serhan CN, Levy BD. 15-epi-Lipoxin A 4, Resolvin D2, and Resolvin D3 Induce NF-κB Regulators in Bacterial Pneumonia. THE JOURNAL OF IMMUNOLOGY 2018. [PMID: 29523657 DOI: 10.4049/jimmunol.1602090] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Specialized proresolving mediators (SPMs) decrease NF-κB activity to prevent excessive tissue damage and promote the resolution of acute inflammation. Mechanisms for NF-κB regulation by SPMs remain to be determined. In this study, after LPS challenge, the SPMs 15-epi-lipoxin A4 (15-epi-LXA4), resolvin D1, resolvin D2, resolvin D3, and 17-epi-resolvin D1 were produced in vivo in murine lungs. In LPS-activated human bronchial epithelial cells, select SPMs increased expression of the NF-κB regulators A20 and single Ig IL-1R-related molecule (SIGIRR). Of interest, 15-epi-LXA4 induced A20 and SIGIRR in an lipoxin A4 receptor/formyl peptide receptor 2 (ALX/FPR2) receptor-dependent manner in epithelial cells and in murine pneumonia. This SPM regulated NF-κB-induced cytokines to decrease pathogen-mediated inflammation. In addition to dampening lung inflammation, surprisingly, 15-epi-LXA4 also enhanced pathogen clearance with increased antimicrobial peptide expression. Taken together, to our knowledge these results are the first to identify endogenous agonists for A20 and SIGIRR expression to regulate NF-κB activity and to establish mechanisms for NF-κB regulation by SPMs for pneumonia resolution.
Collapse
Affiliation(s)
- Ho Pan Sham
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| | - Katherine H Walker
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| | - Raja-Elie E Abdulnour
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| | - Nandini Krishnamoorthy
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| | - David N Douda
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| | - Paul C Norris
- Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Ioanna Barkas
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| | - Sarah Benito-Figueroa
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| | - Jennifer K Colby
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| |
Collapse
|
36
|
Mansbach JM, Hasegawa K, Ajami NJ, Petrosino JF, Piedra PA, Tierney CN, Espinola JA, Camargo CA. Serum LL-37 Levels Associated With Severity of Bronchiolitis and Viral Etiology. Clin Infect Dis 2018; 65:967-975. [PMID: 28541502 DOI: 10.1093/cid/cix483] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/18/2017] [Indexed: 12/18/2022] Open
Abstract
Background LL-37 is a host defense peptide with antimicrobial and immunomodulatory properties. We examined the relation of serum LL-37 levels to the severity of bronchiolitis and viral etiology. Methods We performed a 17-center prospective cohort study in infants hospitalized with bronchiolitis over 3 winters (2011-2014). Site teams collected clinical data, nasopharyngeal aspirates and serum. We used real-time polymerase chain reaction to test nasopharyngeal aspirates for 16 viruses. We tested serum for LL-37. Severity of bronchiolitis was defined by intensive care use and hospital length of stay. Viral etiology was defined as respiratory syncytial virus (RSV) or rhinovirus (RV), including coinfections with other viruses. Results The median age of the 1005 enrolled infants was 3 months (interquartile range, 2-6 months). After adjustment for 12 variables, LL-37 levels in the lowest quartile, compared with the highest, were associated both with intensive care use (adjusted odds ratio [aOR], 1.97; P = .01) and longer hospital stay (1.34; P < .001). In separate multivariable models, infants with LL-37 levels in the lowest 3 quartiles, compared with the highest, were more likely to have RSV (eg, aOR, 2.6 [lowest quartile]; P < .001 [all quartiles]). By contrast, infants with the lowest 3 LL-37 quartiles were less likely to have RV (eg, aOR, 0.5 [lowest quartile]; Pall quartiles ≤ .03 [all quartiles]). Conclusions In a large multicenter study of infants hospitalized with bronchiolitis, lower levels of serum LL-37 were associated with increased severity of illness. There was also an inverse relationship between LL-37 levels and the most common virus causing bronchiolitis, RSV. These findings highlight the role of LL-37 in the pathogenesis of bronchiolitis.
Collapse
Affiliation(s)
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Nadim J Ajami
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology
| | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology and Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Courtney N Tierney
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Janice A Espinola
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
37
|
Horibe K, Hosoya A, Hiraga T, Nakamura H. Expression and localization of CRAMP in rat tooth germ and during reparative dentin formation. Clin Oral Investig 2018; 22:2559-2566. [DOI: 10.1007/s00784-018-2353-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 01/22/2018] [Indexed: 12/19/2022]
|
38
|
Chen K, Bao Z, Gong W, Tang P, Yoshimura T, Wang JM. Regulation of inflammation by members of the formyl-peptide receptor family. J Autoimmun 2017; 85:64-77. [PMID: 28689639 PMCID: PMC5705339 DOI: 10.1016/j.jaut.2017.06.012] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 06/29/2017] [Indexed: 12/14/2022]
Abstract
Inflammation is associated with a variety of diseases. The hallmark of inflammation is leukocyte infiltration at disease sites in response to pathogen- or damage-associated chemotactic molecular patterns (PAMPs and MAMPs), which are recognized by a superfamily of seven transmembrane, Gi-protein-coupled receptors (GPCRs) on cell surface. Chemotactic GPCRs are composed of two major subfamilies: the classical GPCRs and chemokine GPCRs. Formyl-peptide receptors (FPRs) belong to the classical chemotactic GPCR subfamily with unique properties that are increasingly appreciated for their expression on diverse host cell types and the capacity to interact with a plethora of chemotactic PAMPs and MAMPs. Three FPRs have been identified in human: FPR1-FPR3, with putative corresponding mouse counterparts. FPR expression was initially described in myeloid cells but subsequently in many non-hematopoietic cells including cancer cells. Accumulating evidence demonstrates that FPRs possess multiple functions in addition to controlling inflammation, and participate in the processes of many pathophysiologic conditions. They are not only critical mediators of myeloid cell trafficking, but are also implicated in tissue repair, angiogenesis and protection against inflammation-associated tumorigenesis. A series recent discoveries have greatly expanded the scope of FPRs in host defense which uncovered the essential participation of FPRs in step-wise trafficking of myeloid cells including neutrophils and dendritic cells (DCs) in host responses to bacterial infection, tissue injury and wound healing. Also of great interest is the FPRs are exploited by malignant cancer cells for their growth, invasion and metastasis. In this article, we review the current understanding of FPRs concerning their expression in a vast array of cell types, their involvement in guiding leukocyte trafficking in pathophysiological conditions, and their capacity to promote the differentiation of immune cells, their participation in tumor-associated inflammation and cancer progression. The close association of FPRs with human diseases and cancer indicates their potential as targets for the development of therapeutics.
Collapse
Affiliation(s)
- Keqiang Chen
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Zhiyao Bao
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA; Department of Pulmonary & Critical Care Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Peng Tang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA; Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA.
| |
Collapse
|
39
|
Mohammed I, Said DG, Dua HS. Human antimicrobial peptides in ocular surface defense. Prog Retin Eye Res 2017; 61:1-22. [DOI: 10.1016/j.preteyeres.2017.03.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 01/17/2023]
|
40
|
Bosso M, Ständker L, Kirchhoff F, Münch J. Exploiting the human peptidome for novel antimicrobial and anticancer agents. Bioorg Med Chem 2017; 26:2719-2726. [PMID: 29122440 DOI: 10.1016/j.bmc.2017.10.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 10/28/2017] [Indexed: 12/22/2022]
Abstract
Infectious diseases and cancers are leading causes of death and pose major challenges to public health. The human peptidome encompasses millions of compounds that display an enormous structural and functional diversity and represents an excellent source for the discovery of endogenous agents with antimicrobial and/or anticancer activity. Here, we discuss how to exploit the human peptidome for novel antimicrobial and anticancer agents through the generation of peptide libraries from human body fluids and tissues and stepwise purification of bioactive compounds.
Collapse
Affiliation(s)
- Matteo Bosso
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstrasse 1, 89081 Ulm, Germany
| | - Ludger Ständker
- Ulm Peptide Pharmaceuticals, Ulm University, Albert-Einstein-Allee 47, 89081 Ulm, Germany; Core Facility Functional Peptidomics, Ulm University Medical Center, Albert-Einstein-Allee 47, 89081 Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstrasse 1, 89081 Ulm, Germany; Ulm Peptide Pharmaceuticals, Ulm University, Albert-Einstein-Allee 47, 89081 Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstrasse 1, 89081 Ulm, Germany; Ulm Peptide Pharmaceuticals, Ulm University, Albert-Einstein-Allee 47, 89081 Ulm, Germany; Core Facility Functional Peptidomics, Ulm University Medical Center, Albert-Einstein-Allee 47, 89081 Ulm, Germany.
| |
Collapse
|
41
|
Munguia J, LaRock DL, Tsunemoto H, Olson J, Cornax I, Pogliano J, Nizet V. The Mla pathway is critical for Pseudomonas aeruginosa resistance to outer membrane permeabilization and host innate immune clearance. J Mol Med (Berl) 2017; 95:1127-1136. [PMID: 28844103 PMCID: PMC5671890 DOI: 10.1007/s00109-017-1579-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 08/14/2017] [Accepted: 08/14/2017] [Indexed: 12/11/2022]
Abstract
Pseudomonas aeruginosa is an important opportunistic pathogen that has become a serious problem due to increased rates of antibiotic resistance. Due to this along with a dearth in novel antibiotic development, especially against Gram-negative pathogens, new therapeutic strategies are needed to prevent a post-antibiotic era. Here, we describe the importance of the vacJ/Mla pathway in resisting bactericidal actions of the host innate immune response. P. aeruginosa tn5 transposon mutants in genes from the VacJ/Mla pathway showed increased susceptibility to killing by the host cathelicidin antimicrobial peptide, LL-37, when compared to the wild-type parent strain. The P. aeruginosa vacJ - mutant demonstrated increased membrane permeability upon damage as well as sensitivity to killing in the presence of the detergent sodium dodecyl sulfate and the divalent cation chelator EDTA. When exposed to human whole blood and serum complement, the vacJ - mutant was killed more rapidly when compared to the wild-type parent strain and complemented mutant. Finally, in an in vivo mouse lung infection model, infection with the vacJ - mutant resulted in reduced mortality, lower bacterial burden, and reduced lung damage when compared to the wild-type strain. This study highlights the potential in therapeutically targeting the VacJ/Mla pathway in sensitizing P. aeruginosa to killing by the host innate immune response. KEY MESSAGES • The Mla pathway regulates outer membrane dynamics in human pathogen Pseudomonas aeruginosa (PA). • Disruption of Mla pathway gene vacJ sensitizes PA to host cathelicidin antimicrobial peptide LL-37. • Loss of vacJ expression renders PA more sensitive to human whole blood and serum killing. • Loss of vacJ expression reduces PA survival and virulence in a murine lung infection model. • The Mla pathway merits exploration as a pharmacologic target to sensitize PA to host innate immunity.
Collapse
Affiliation(s)
- Jason Munguia
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Doris L LaRock
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Hannah Tsunemoto
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Joshua Olson
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Ingrid Cornax
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Joseph Pogliano
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Victor Nizet
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.
- Rady Children's Hospital, San Diego, CA, USA.
| |
Collapse
|
42
|
IL-36γ is a crucial proximal component of protective type-1-mediated lung mucosal immunity in Gram-positive and -negative bacterial pneumonia. Mucosal Immunol 2017; 10:1320-1334. [PMID: 28176791 PMCID: PMC5548659 DOI: 10.1038/mi.2016.130] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 12/01/2016] [Indexed: 02/04/2023]
Abstract
Interleukin-36γ (IL-36γ) is a member of novel IL-1-like proinflammatory cytokine family that are highly expressed in epithelial tissues and several myeloid-derived cell types. Little is known about the role of the IL-36 family in mucosal immunity, including lung anti-bacterial responses. We used murine models of IL-36γ deficiency to assess the contribution of IL-36γ in the lung during experimental pneumonia. Induction of IL-36γ was observed in the lung in response to Streptococcus pneumoniae (Sp) infection, and mature IL-36γ protein was secreted primarily in microparticles. IL-36γ-deficient mice challenged with Sp demonstrated increased mortality, decreased lung bacterial clearance and increased bacterial dissemination, in association with reduced local expression of type-1 cytokines, and impaired lung macrophage M1 polarization. IL-36γ directly stimulated type-1 cytokine induction from dendritic cells in vitro in a MyD88-dependent manner. Similar protective effects of IL-36γ were observed in a Gram-negative pneumonia model (Klebsiella pneumoniae). Intrapulmonary delivery of IL-36γ-containing microparticles reconstituted immunity in IL-36γ-/- mice. Enhanced expression of IL-36γ was also observed in plasma and bronchoalveolar lavage fluid of patients with acute respiratory distress syndrome because of pneumonia. These studies indicate that IL-36γ assumes a vital proximal role in the lung innate mucosal immunity during bacterial pneumonia by driving protective type-1 responses and classical macrophage activation.
Collapse
|
43
|
Xia X, Cheng L, Zhang S, Wang L, Hu J. The role of natural antimicrobial peptides during infection and chronic inflammation. Antonie van Leeuwenhoek 2017; 111:5-26. [PMID: 28856473 DOI: 10.1007/s10482-017-0929-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 08/15/2017] [Indexed: 01/12/2023]
Abstract
Natural antimicrobial peptides (AMPs), a family of small polypeptides that are produced by constitutive or inducible expression in organisms, are integral components of the host innate immune system. In addition to their broad-spectrum antibacterial activity, natural AMPs also have many biological activities against fungi, viruses and parasites. Natural AMPs exert multiple immunomodulatory roles that may predominate under physiological conditions where they lose their microbicidal properties in serum and tissue environments. Increased drug resistance among microorganisms is occurring far more quickly than the discovery of new antibiotics. Natural AMPs have shown promise as 'next generation antibiotics' due to their broad-spectrum curative effects, low toxicity, the fact that they are not residual in animals, and the low rates of resistance exhibited by many pathogens. Many types of synthetic AMPs are currently being tested in clinical trials for the prevention and treatment of various diseases such as chemotherapy-associated infections, diabetic foot ulcers, catheter-related infections, and other conditions. Here, we provide an overview of the types and functions of natural AMPs and their role in combating microorganisms and different infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Xiaojing Xia
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, No. 90, Hualan Street, Xinxiang, 453003, People's Republic of China
| | - Likun Cheng
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, 256600, People's Republic of China
| | - Shouping Zhang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, No. 90, Hualan Street, Xinxiang, 453003, People's Republic of China
| | - Lei Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, No. 90, Hualan Street, Xinxiang, 453003, People's Republic of China
| | - Jianhe Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, No. 90, Hualan Street, Xinxiang, 453003, People's Republic of China.
| |
Collapse
|
44
|
Receptor for advanced glycation endproducts (RAGE) maintains pulmonary structure and regulates the response to cigarette smoke. PLoS One 2017; 12:e0180092. [PMID: 28678851 PMCID: PMC5497997 DOI: 10.1371/journal.pone.0180092] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 06/09/2017] [Indexed: 12/15/2022] Open
Abstract
The receptor for advanced glycation endproducts (RAGE) is highly expressed in the lung but its physiological functions in this organ is still not completely understood. To determine the contribution of RAGE to physiological functions of the lung, we analyzed pulmonary mechanics and structure of wildtype and RAGE deficient (RAGE-/-) mice. RAGE deficiency spontaneously resulted in a loss of lung structure shown by an increased mean chord length, increased respiratory system compliance, decreased respiratory system elastance and increased concentrations of serum protein albumin in bronchoalveolar lavage fluids. Pulmonary expression of RAGE was mainly localized on alveolar epithelial cells and alveolar macrophages. Primary murine alveolar epithelial cells isolated from RAGE-/- mice revealed an altered differentiation and defective barrier formation under in vitro conditions. Stimulation of interferone-y (IFNy)-activated alveolar macrophages deficient for RAGE with Toll-like receptor (TLR) ligands resulted in significantly decreased release of proinflammatory cytokines and chemokines. Exposure to chronic cigarette smoke did not affect emphysema-like changes in lung parenchyma in RAGE-/- mice. Acute cigarette smoke exposure revealed a modified inflammatory response in RAGE-/- mice that was characterized by an influx of macrophages and a decreased keratinocyte-derived chemokine (KC) release. Our data suggest that RAGE regulates the differentiation of alveolar epithelial cells and impacts on the development and maintenance of pulmonary structure. In cigarette smoke-induced lung pathology, RAGE mediates inflammation that contributes to lung damage.
Collapse
|
45
|
Abstract
Supplemental Digital Content is available in the text. Objectives: Vitamin D deficiency has been implicated as a pathogenic factor in sepsis and ICU mortality but causality of these associations has not been demonstrated. To determine whether sepsis and severe sepsis are associated with vitamin D deficiency and to determine whether vitamin D deficiency influences the severity of sepsis. Design, Setting, and Patients: Sixty-one patients with sepsis and severe sepsis from two large U.K. hospitals and 20 healthy controls were recruited. Murine models of cecal ligation and puncture and intratracheal lipopolysaccharide were undertaken in normal and vitamin D deficient mice to address the issue of causality. Measurements and Main Results: Patients with severe sepsis had significantly lower concentrations of 25-hydroxyvitamin D3 than patients with either mild sepsis or age-matched healthy controls (15.7 vs 49.5 vs 66.5 nmol/L; p = 0.0001). 25-hydroxyvitamin D3 concentrations were significantly lower in patients who had positive microbiologic culture than those who were culture negative (p = 0.0023) as well as those who died within 30 days of hospital admission (p = 0.025). Vitamin D deficiency in murine sepsis was associated with increased peritoneal (p = 0.037), systemic (p = 0.019), and bronchoalveolar lavage (p = 0.011) quantitative bacterial culture. This was associated with reduced local expression of the cathelicidin-related antimicrobial peptide as well as evidence of defective macrophage phagocytosis (p = 0.029). In the intratracheal lipopolysaccharide model, 1,500 IU of intraperitoneal cholecalciferol treatment 6 hours postinjury reduced alveolar inflammation, cellular damage, and hypoxia. Conclusions: Vitamin D deficiency is common in severe sepsis. This appears to contribute to the development of the condition in clinically relevant murine models and approaches to correct vitamin D deficiency in patients with sepsis should be developed.
Collapse
|
46
|
Pinheiro da Silva F, Machado MCC. The dual role of cathelicidins in systemic inflammation. Immunol Lett 2017; 182:57-60. [PMID: 28082134 DOI: 10.1016/j.imlet.2017.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/11/2016] [Accepted: 01/04/2017] [Indexed: 01/10/2023]
Abstract
Antimicrobial peptides are key components of the innate immune system. They act as broad-spectrum antimicrobial agents against Gram-positive and -negative bacteria, viruses, and fungi. More recently, antimicrobial peptides have been ascribed immunomodulatory functions, including roles in wound healing, induction of cytokines, and altering host gene expression. Cathelicidins are a class of antimicrobial peptide found in humans, mice, and rats, among others. Known as LL-37 in humans and cathelin-related antimicrobial peptide (CRAMP) in rodents, cathelicidins are produced by many different cells, including macrophages, neutrophils, and epithelial cells. The role of cathelicidins is somewhat confounding, as they exhibit both pro-and anti-inflammatory activity. A major obstacle in the study of cathelicidins is the inability of exogenous LL-37 or CRAMP to mimic the activity of their endogenous counterparts. Nevertheless, studies have shown that LL-37 is recognized by multiple receptors, and may stabilize or modulate Toll-like receptor signaling. In addition, cathelicidins play a role in apoptosis, inflammasome activation, and phagocytosis. However, many studies are revealing the dual effects of cathelicidins. For example, CRAMP appears to be protective in models of group A Streptococcus skin infection, pneumonia, and meningitis, but detrimental in cases of severe bacterial infection, such as septic shock. It is becoming increasingly clear that the activity of cathelicidins is modulated by complex interactions with the microenvironment, as well as the disease background. This article reviews what is currently known about the activity of cathelicidins in an attempt to understand their complex roles in systemic diseases.
Collapse
|
47
|
Kolar SS, Baidouri H, Mangoni ML, McDermott AM. Methods for In Vivo/Ex Vivo Analysis of Antimicrobial Peptides in Bacterial Keratitis: siRNA Knockdown, Colony Counts, Myeloperoxidase, Immunostaining, and RT-PCR Assays. Methods Mol Biol 2017; 1548:411-425. [PMID: 28013522 PMCID: PMC11991856 DOI: 10.1007/978-1-4939-6737-7_30] [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] [Indexed: 04/13/2025]
Abstract
Antimicrobial peptides (AMPs) are essential components of the innate immune response. They have direct killing ability as well as immunomodulatory functions. Here, we describe techniques to identify specific AMPs involved in the protection against microbial keratitis, a vision threatening infection of the cornea of the eye which is the most serious complication of contact lens wear. Specifically we detail the use of siRNA technology to temporarily knockdown AMP expression at the murine ocular surface in vivo and then describe ex vivo assays to determine the level of bacteria, relative number of neutrophils, and levels of cytokines, chemokines, and AMPs in infected corneas.
Collapse
Affiliation(s)
- Satya Sree Kolar
- The Ocular Surface Institute, College of Optometry, University of Houston, 4901 Calhoun Road, Houston, TX, 77204, USA
| | - Hasna Baidouri
- The Ocular Surface Institute, College of Optometry, University of Houston, 4901 Calhoun Road, Houston, TX, 77204, USA
| | - Maria Luisa Mangoni
- Department of Biochemical Sciences "A. Rossi Fanelli", Istituto Pasteur-Fondaziona Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Alison M McDermott
- The Ocular Surface Institute, College of Optometry, University of Houston, 4901 Calhoun Road, Houston, TX, 77204, USA.
| |
Collapse
|
48
|
Bergman P, Seyedoleslami Esfahani S, Engström Y. Drosophila as a Model for Human Diseases—Focus on Innate Immunity in Barrier Epithelia. Curr Top Dev Biol 2017; 121:29-81. [DOI: 10.1016/bs.ctdb.2016.07.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
49
|
Abstract
Antimicrobial peptides (AMPs), also known as host defense peptides, are small naturally occurring microbicidal molecules produced by the host innate immune response that function as a first line of defense to kill pathogenic microorganisms by inducing deleterious cell membrane damage. AMPs also possess signaling and chemoattractant activities and can modulate the innate immune response to enhance protective immunity or suppress inflammation. Human pathogens have evolved defense molecules and strategies to counter and survive the AMPs released by host immune cells such as neutrophils and macrophages. Here, we review the various mechanisms used by human bacterial pathogens to resist AMP-mediated killing, including surface charge modification, active efflux, alteration of membrane fluidity, inactivation by proteolytic digestion, and entrapment by surface proteins and polysaccharides. Enhanced understanding of AMP resistance at the molecular level may offer insight into the mechanisms of bacterial pathogenesis and augment the discovery of novel therapeutic targets and drug design for the treatment of recalcitrant multidrug-resistant bacterial infections.
Collapse
|
50
|
Anti-fungal activity of Ctn[15-34], the C-terminal peptide fragment of crotalicidin, a rattlesnake venom gland cathelicidin. J Antibiot (Tokyo) 2016; 70:231-237. [PMID: 27876749 DOI: 10.1038/ja.2016.135] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/25/2016] [Accepted: 10/17/2016] [Indexed: 12/11/2022]
Abstract
Crotalicidin (Ctn), a 34-residue cathelicidin from a South American rattlesnake, and its fragment (Ctn[15-34]) have shown anti-infective and cytotoxic activities against Gram-negative bacteria and certain tumor lines, respectively. The extent of such effects has been related to physicochemical characteristics such as helicity and hydrophobicity. We now report the anti-fungal activity of Ctn and its fragments (Ctn[1-14]) and (Ctn[15-34]). MIC determination and luminescent cell viability assays were used to evaluate the anti-infective activity of Ctn and its fragments (Ctn[1-14]) and (Ctn[15-34]) as anti-fungal agents against opportunistic yeast and dermatophytes. Cytotoxicity towards healthy eukaryotic cells was assessed in vitro with healthy human kidney-2 (HK-2) cells and erythrocytes. The checkerboard technique was performed to estimate the effects of combining either one of the peptides with amphotericin B. Ctn was the most active peptide against dermatophytes and also the most toxic to healthy eukaryotic cells. Fragments Ctn[1-14] and Ctn[15-35] lost activity against dermatophytes, but became more active against pathogenic yeasts, including several Candida species, both clinical isolates and standard strains, with MICs as low as 5 μm. Interestingly, the two peptide fragments were less cytotoxic to healthy HK-2 cells and less hemolytic to human erythrocytes than the standard-of-care amphotericin B. Also noteworthy was the synergy between Ctn peptides and amphotericin B, with consequent reduction in MICs of both drug and peptides. Altogether, Ctn and its fragments, particularly Ctn[15-34], are promising leads, either alone or in combined regimen with amphotericin B, for the treatment of fungal diseases.
Collapse
|