151
|
Platelets Enhance Dendritic Cell Responses against Staphylococcus aureus through CD40-CD40L. Infect Immun 2018; 86:IAI.00186-18. [PMID: 29914928 DOI: 10.1128/iai.00186-18] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/12/2018] [Indexed: 12/24/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen that can cause mild to severe life-threatening infections in many tissues and organs. Platelets are known to participate in protection against S. aureus by direct killing and by enhancing the activities of neutrophils and macrophages in clearing S. aureus infection. Platelets have also been shown to induce monocyte differentiation into dendritic cells and to enhance activation of dendritic cells. Therefore, in the present study, we explored the role of platelets in enhancing bone marrow-derived dendritic cell (BMDC) function against S. aureus We observed a significant increase in dendritic cell phagocytosis and intracellular killing of a methicillin-resistant Staphylococcus aureus (MRSA) strain (USA300) by thrombin-activated platelets or their releasates. Enhancement of bacterial uptake and killing by DCs is mediated by platelet-derived CD40L. Coculture of USA300 and BMDCs in the presence of thrombin-activated platelet releasates invokes upregulation of the maturation marker CD80 on DCs and enhanced production of the proinflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin 12 (IL-12), and IL-6. Overall, these observations support our hypothesis that platelets play a critical role in the host defense against S. aureus infection. Platelets stimulate DCs, leading to direct killing of S. aureus and enhanced DC maturation, potentially leading to adaptive immune responses against S. aureus.
Collapse
|
152
|
Nazario-Toole AE, Robalino J, Okrah K, Corrada-Bravo H, Mount SM, Wu LP. The Splicing Factor RNA-Binding Fox Protein 1 Mediates the Cellular Immune Response in Drosophila melanogaster. THE JOURNAL OF IMMUNOLOGY 2018; 201:1154-1164. [PMID: 29997126 DOI: 10.4049/jimmunol.1800496] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022]
Abstract
The uptake and destruction of bacteria by phagocytic cells is an essential defense mechanism in metazoans. To identify novel genes involved in the phagocytosis of Staphylococcus aureus, a major human pathogen, we assessed the phagocytic capacity of adult blood cells (hemocytes) of the fruit fly, Drosophila melanogaster, by testing several lines of the Drosophila Genetic Reference Panel. Natural genetic variation in the gene RNA-binding Fox protein 1 (Rbfox1) correlated with low phagocytic capacity in hemocytes, pointing to Rbfox1 as a candidate regulator of phagocytosis. Loss of Rbfox1 resulted in increased expression of the Ig superfamily member Down syndrome adhesion molecule 4 (Dscam4). Silencing of Dscam4 in Rbfox1-depleted blood cells rescued the fly's cellular immune response to S. aureus, indicating that downregulation of Dscam4 by Rbfox1 is critical for S. aureus phagocytosis in Drosophila To our knowledge, this study is the first to demonstrate a link between Rbfox1, Dscam4, and host defense against S. aureus.
Collapse
Affiliation(s)
- Ashley E Nazario-Toole
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742.,Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742; and
| | - Javier Robalino
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Kwame Okrah
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742
| | - Hector Corrada-Bravo
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742
| | - Stephen M Mount
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742.,Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742
| | - Louisa P Wu
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742; .,Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742; and
| |
Collapse
|
153
|
Brandt SL, Klopfenstein N, Wang S, Winfree S, McCarthy BP, Territo PR, Miller L, Serezani CH. Macrophage-derived LTB4 promotes abscess formation and clearance of Staphylococcus aureus skin infection in mice. PLoS Pathog 2018; 14:e1007244. [PMID: 30102746 PMCID: PMC6107286 DOI: 10.1371/journal.ppat.1007244] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 08/23/2018] [Accepted: 07/26/2018] [Indexed: 01/26/2023] Open
Abstract
The early events that shape the innate immune response to restrain pathogens during skin infections remain elusive. Methicillin-resistant Staphylococcus aureus (MRSA) infection engages phagocyte chemotaxis, abscess formation, and microbial clearance. Upon infection, neutrophils and monocytes find a gradient of chemoattractants that influence both phagocyte direction and microbial clearance. The bioactive lipid leukotriene B4 (LTB4) is quickly (seconds to minutes) produced by 5-lipoxygenase (5-LO) and signals through the G protein-coupled receptors LTB4R1 (BLT1) or BLT2 in phagocytes and structural cells. Although it is known that LTB4 enhances antimicrobial effector functions in vitro, whether prompt LTB4 production is required for bacterial clearance and development of an inflammatory milieu necessary for abscess formation to restrain pathogen dissemination is unknown. We found that LTB4 is produced in areas near the abscess and BLT1 deficient mice are unable to form an abscess, elicit neutrophil chemotaxis, generation of neutrophil and monocyte chemokines, as well as reactive oxygen species-dependent bacterial clearance. We also found that an ointment containing LTB4 synergizes with antibiotics to eliminate MRSA potently. Here, we uncovered a heretofore unknown role of macrophage-derived LTB4 in orchestrating the chemoattractant gradient required for abscess formation, while amplifying antimicrobial effector functions.
Collapse
Affiliation(s)
- Stephanie L. Brandt
- Indiana University School of Medicine, Department of Microbiology & Immunology, Indianapolis, Indiana, United States of America
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Disease, Nashville, Tennessee, United States of America
| | - Nathan Klopfenstein
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Disease, Nashville, Tennessee, United States of America
- Vanderbilt University Medical Center, Department of Pathology, Microbiology and Immunology, Nashville, Tennessee, United States of America
| | - Soujuan Wang
- Indiana University School of Medicine, Department of Microbiology & Immunology, Indianapolis, Indiana, United States of America
| | - Seth Winfree
- Indiana Center for Biological Microscopy, Indianapolis, Indiana, United States of America
| | - Brian P. McCarthy
- Indiana Institute for Biomedical Imaging Sciences, Department of Radiology, Indianapolis, Indiana, United States of America
| | - Paul R. Territo
- Indiana Institute for Biomedical Imaging Sciences, Department of Radiology, Indianapolis, Indiana, United States of America
| | - Lloyd Miller
- Johns Hopkins University School of Medicine, Department of Dermatology, Baltimore, Maryland, United States of America
| | - C. Henrique Serezani
- Indiana University School of Medicine, Department of Microbiology & Immunology, Indianapolis, Indiana, United States of America
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Disease, Nashville, Tennessee, United States of America
- Vanderbilt University Medical Center, Department of Pathology, Microbiology and Immunology, Nashville, Tennessee, United States of America
- Vanderbilt Institute of Infection, Immunology and Inflammation, Nashville, Tennessee, United States of America
| |
Collapse
|
154
|
Yu J, Francisco AMC, Patel BG, Cline JM, Zou E, Berga SL, Taylor RN. IL-1β Stimulates Brain-Derived Neurotrophic Factor Production in Eutopic Endometriosis Stromal Cell Cultures: A Model for Cytokine Regulation of Neuroangiogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2281-2292. [PMID: 30031725 DOI: 10.1016/j.ajpath.2018.06.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 05/15/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022]
Abstract
Endometriosis implants are comprised of glandular and stromal elements, macrophages, nerves, and blood vessels and are commonly accompanied by pelvic pain. We propose that activated macrophages are recruited to and infiltrate nascent lesions, where they secrete proinflammatory cytokines, promoting the production of chemokines, neurotrophins, and angiogenic growth factors that sustain an inflammatory microenvironment. Immunohistochemical evaluation of endometriosis lesions reveals in situ colocalization of concentrated macrophages, brain-derived neurotrophic factor (BDNF), and nerve fibers. These observations were coupled with biochemical analyses of primary eutopic endometriosis stromal cell (EESC) cultures, which allowed defining potential pathways leading to the neuroangiogenic phenotype of these lesions. Our findings indicate that IL-1β potently (EC50 = 7 ± 2 ng/mL) stimulates production of EESC BDNF at the mRNA and protein levels in an IL-1 receptor-dependent fashion. Selective kinase inhibitors demonstrate that this IL-1β effect is mediated by c-Jun N-terminal kinase (JNK), NF-κB, and mechanistic target of rapamycin signal transduction pathways. IL-1β regulation of regulated on activation normal T cell expressed and secreted (RANTES), a prominent EESC chemokine, also relies on JNK and NF-κB. An important clinical implication of the study is that interference with BDNF and RANTES production, by selectively targeting the JNK and NF-κB cascades, may offer a tractable therapeutic strategy to mitigate the pain and inflammation associated with endometriosis.
Collapse
Affiliation(s)
- Jie Yu
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Antônio M C Francisco
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina; Health Sciences School, University of Vale do Sapucaí, Pouso Alegre, Brazil
| | - Bansari G Patel
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - J Mark Cline
- Department of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina; Molecular Medicine and Translational Sciences Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Eric Zou
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Sarah L Berga
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Robert N Taylor
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina; Molecular Medicine and Translational Sciences Program, Wake Forest School of Medicine, Winston-Salem, North Carolina; Clinical and Translational Science Institute, Wake Forest School of Medicine, Winston-Salem, North Carolina.
| |
Collapse
|
155
|
Shibue Y, Kimura S, Kajiwara C, Iwakura Y, Yamaguchi K, Tateda K. Role of interleukin-17 in a murine community-associated methicillin-resistant Staphylococcus aureus pneumonia model. Microbes Infect 2018; 21:33-39. [PMID: 30026066 DOI: 10.1016/j.micinf.2018.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/20/2018] [Accepted: 06/28/2018] [Indexed: 11/30/2022]
Abstract
Interleukin (IL)-17 is a key member of the Th17 cytokines and has been reported to be involved in the pathomechanisms underlying various diseases, including infectious diseases. Infections with community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) have garnered worldwide attention, and the representative USA300 strain is known to cause pneumonia in healthy people, which can be lethal. However, little is known about the role of IL-17 in CA-MRSA pneumonia. In this study, we investigated the role of IL-17 in a CA-MRSA pneumonia animal model. Mortality was higher and occurred at an earlier stage of infection in the IL-17A-knockout mice than in the wild-type (P < 0.01) and IL-17A/F-knockout mice (P < 0.05); however, no significant difference in the intrapulmonary bacterial counts was observed among the three groups of mice. Moreover, the IL-17A-knockout group showed significantly higher levels of IL-17F and granulocyte-colony stimulating factor (G-CSF) and a significantly higher neutrophil count in the bronchoalveolar lavage fluid than the other groups. These results confirmed that G-CSF expression significantly increased, and significant neutrophilic inflammation occurred under conditions of IL-17A deficiency in the murine CA-MRSA pneumonia model.
Collapse
Affiliation(s)
- Yasushi Shibue
- Department of Microbiology and Infectious Diseases, Toho University School of Medicine, 5-21-16, Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan; First Department of Medicine, Hokkaido University School of Medicine, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Soichiro Kimura
- Department of Microbiology and Infectious Diseases, Toho University School of Medicine, 5-21-16, Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan.
| | - Chiaki Kajiwara
- Department of Microbiology and Infectious Diseases, Toho University School of Medicine, 5-21-16, Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamasaki, Noda, Chiba, 278-0022, Japan
| | - Keizo Yamaguchi
- Department of Microbiology and Infectious Diseases, Toho University School of Medicine, 5-21-16, Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Kazuhiro Tateda
- Department of Microbiology and Infectious Diseases, Toho University School of Medicine, 5-21-16, Omori-nishi, Ota-ku, Tokyo, 143-8540, Japan
| |
Collapse
|
156
|
Poolman JT, Anderson AS. Escherichia coliandStaphylococcus aureus: leading bacterial pathogens of healthcare associated infections and bacteremia in older-age populations. Expert Rev Vaccines 2018; 17:607-618. [DOI: 10.1080/14760584.2018.1488590] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Jan T. Poolman
- Bacterial Vaccines Discovery & Early Development, Janssen Vaccines and Prevention, Leiden, The Netherlands
| | | |
Collapse
|
157
|
Cela EM, Gonzalez CD, Friedrich A, Ledo C, Paz ML, Leoni J, Gómez MI, González Maglio DH. Daily very low UV dose exposure enhances adaptive immunity, compared with a single high-dose exposure. Consequences for the control of a skin infection. Immunology 2018; 154:510-521. [PMID: 29377107 PMCID: PMC6002207 DOI: 10.1111/imm.12901] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/24/2017] [Accepted: 01/12/2018] [Indexed: 12/22/2022] Open
Abstract
Ultraviolet radiation (UVr) promotes several well-known molecular changes, which may ultimately impact on health. Some of these effects are detrimental, like inflammation, carcinogenesis and immunosuppression. On the other hand, UVr also promotes vitamin D synthesis and other beneficial effects. We recently demonstrated that exposure to very low doses of UVr on four consecutive days [repetitive low UVd (rlUVd)] does not promote an inflammatory state, nor the recruitment of neutrophils or lymphocytes, as the exposure to a single high UV dose (shUVd) does. Moreover, rlUVd reinforce the epithelium by increasing antimicrobial peptides transcription and epidermal thickness. The aim of this study was to evaluate the adaptive immune response after shUVd and rlUVd, determining T-cell and B-cell responses. Finally, we challenged animals exposed to both irradiation procedures with Staphylococcus aureus to study the overall effects of both innate and adaptive immunity during a cutaneous infection. We observed, as expected, a marked suppression of T-cell and B-cell responses after exposure to an shUVd but a novel and significant increase in both specific responses after exposure to rlUVd. However, the control of the cutaneous S. aureus infection was defective in this last group, suggesting that responses against pathogens cannot be ruled out from isolated stimuli.
Collapse
Affiliation(s)
- Eliana M. Cela
- Universidad de Buenos AiresFacultad de Farmacia y BioquímicaCátedra de InmunologíaBuenos AiresArgentina
- CONICET – Universidad de Buenos AiresInstituto de Estudios de la Inmunidad Humoral (IDEHU)Buenos AiresArgentina
| | - Cintia Daniela Gonzalez
- Universidad de Buenos AiresFacultad de MedicinaDepartamento de Microbiología, Parasitología e InmunologíaBuenos AiresArgentina
- CONICET – Universidad de Buenos AiresInstituto de Microbiología y Parasitología Médica (IMPaM)Buenos AiresArgentina
| | - Adrian Friedrich
- Universidad de Buenos AiresFacultad de Farmacia y BioquímicaCátedra de InmunologíaBuenos AiresArgentina
| | - Camila Ledo
- Universidad de Buenos AiresFacultad de MedicinaDepartamento de Microbiología, Parasitología e InmunologíaBuenos AiresArgentina
- CONICET – Universidad de Buenos AiresInstituto de Microbiología y Parasitología Médica (IMPaM)Buenos AiresArgentina
| | - Mariela Laura Paz
- Universidad de Buenos AiresFacultad de Farmacia y BioquímicaCátedra de InmunologíaBuenos AiresArgentina
- CONICET – Universidad de Buenos AiresInstituto de Estudios de la Inmunidad Humoral (IDEHU)Buenos AiresArgentina
| | - Juliana Leoni
- CONICET – Universidad de Buenos AiresInstituto de Estudios de la Inmunidad Humoral (IDEHU)Buenos AiresArgentina
| | - Marisa Inés Gómez
- Universidad de Buenos AiresFacultad de MedicinaDepartamento de Microbiología, Parasitología e InmunologíaBuenos AiresArgentina
- CONICET – Universidad de Buenos AiresInstituto de Microbiología y Parasitología Médica (IMPaM)Buenos AiresArgentina
| | - Daniel H. González Maglio
- Universidad de Buenos AiresFacultad de Farmacia y BioquímicaCátedra de InmunologíaBuenos AiresArgentina
- CONICET – Universidad de Buenos AiresInstituto de Estudios de la Inmunidad Humoral (IDEHU)Buenos AiresArgentina
| |
Collapse
|
158
|
Brandt SL, Putnam NE, Cassat JE, Serezani CH. Innate Immunity to Staphylococcus aureus: Evolving Paradigms in Soft Tissue and Invasive Infections. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:3871-3880. [PMID: 29866769 PMCID: PMC6028009 DOI: 10.4049/jimmunol.1701574] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/12/2018] [Indexed: 01/18/2023]
Abstract
Staphylococcus aureus causes a wide range of diseases that together embody a significant public health burden. Aided by metabolic flexibility and a large virulence repertoire, S. aureus has the remarkable ability to hematogenously disseminate and infect various tissues, including skin, lung, heart, and bone, among others. The hallmark lesions of invasive staphylococcal infections, abscesses, simultaneously denote the powerful innate immune responses to tissue invasion as well as the ability of staphylococci to persist within these lesions. In this article, we review the innate immune responses to S. aureus during infection of skin and bone, which serve as paradigms for soft tissue and bone disease, respectively.
Collapse
Affiliation(s)
- Stephanie L Brandt
- Division of Infectious Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Nicole E Putnam
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - James E Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232;
- Division of Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232; and
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232
| | - C Henrique Serezani
- Division of Infectious Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232;
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
159
|
Salat M, Petkova P, Hoyo J, Perelshtein I, Gedanken A, Tzanov T. Durable antimicrobial cotton textiles coated sonochemically with ZnO nanoparticles embedded in an in-situ enzymatically generated bioadhesive. Carbohydr Polym 2018; 189:198-203. [DOI: 10.1016/j.carbpol.2018.02.033] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 02/06/2018] [Accepted: 02/10/2018] [Indexed: 02/07/2023]
|
160
|
Montanari E, Oates A, Di Meo C, Meade J, Cerrone R, Francioso A, Devine D, Coviello T, Mancini P, Mosca L, Matricardi P. Hyaluronan-Based Nanohydrogels for Targeting Intracellular S. Aureus in Human Keratinocytes. Adv Healthc Mater 2018; 7:e1701483. [PMID: 29696813 DOI: 10.1002/adhm.201701483] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/13/2018] [Indexed: 11/05/2022]
Abstract
Staphylococcus aureus is one of the most significant human pathogens that is frequently isolated in a wide range of superficial and systemic infections. The ability of S. aureus to invade and survive within host cells such as keratinocytes and host immune cells has been increasingly recognized as a potential factor in persistent infections and treatment failures. The incorporation of antibiotics into hyaluronan-cholesterol nanohydrogels represents a novel paradigm in the delivery of therapeutic agents against intracellular bacteria. The work presented herein shows that NHs quickly enter human keratinocytes and accumulate into lysosomes. When used for targeting intracellular S. aureus the antimicrobial activity of loaded levofloxacin is enhanced, possibly changing the antibiotic intracellular fate from cytosol to lysosome. Indeed, gentamicin, an antibiotic that predominantly accumulates in lysosomes, shows significant and equal antibacterial activity when entrapped into NHs. These results strongly suggest that lysosomal formulations may display preferential activity toward intracellular S. aureus, opening new avenues for the use of HA-based NHs for treatment of such skin infections.
Collapse
Affiliation(s)
- Elita Montanari
- Department of Drug Chemistry and Technologies; Sapienza University of Rome; P.le Aldo Moro 5 Rome 00185 Italy
| | - Angela Oates
- School of Healthcare; Faculty of Medicine and Health; University of Leeds; Leeds LS2 9JT UK
| | - Chiara Di Meo
- Department of Drug Chemistry and Technologies; Sapienza University of Rome; P.le Aldo Moro 5 Rome 00185 Italy
| | - Josephine Meade
- Division of Oral Biology; Faculty of Medicine and Health; School of Dentistry; University of Leeds; Leeds LS7 9TF UK
| | - Rugiada Cerrone
- Department of Drug Chemistry and Technologies; Sapienza University of Rome; P.le Aldo Moro 5 Rome 00185 Italy
| | - Antonio Francioso
- Department of Biochemical Sciences “A. Rossi Fanelli”; Sapienza University of Rome; P.le Aldo Moro 5 Rome 00185 Italy
| | - Deirdre Devine
- Division of Oral Biology; School of Dentistry; Faculty of Medicine and Health; University of Leeds; Leeds LS2 9LU UK
| | - Tommasina Coviello
- Department of Drug Chemistry and Technologies; Sapienza University of Rome; P.le Aldo Moro 5 Rome 00185 Italy
| | - Patrizia Mancini
- Department of Experimental Medicine; Sapienza University of Rome; V.le Regina Elena 291 Rome 00161 Italy
| | - Luciana Mosca
- Department of Biochemical Sciences “A. Rossi Fanelli”; Sapienza University of Rome; P.le Aldo Moro 5 Rome 00185 Italy
| | - Pietro Matricardi
- Department of Drug Chemistry and Technologies; Sapienza University of Rome; P.le Aldo Moro 5 Rome 00185 Italy
| |
Collapse
|
161
|
A Phage Lysin Fused to a Cell-Penetrating Peptide Kills Intracellular Methicillin-Resistant Staphylococcus aureus in Keratinocytes and Has Potential as a Treatment for Skin Infections in Mice. Appl Environ Microbiol 2018; 84:AEM.00380-18. [PMID: 29625989 DOI: 10.1128/aem.00380-18] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/03/2018] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus is the main pathogen that causes skin and skin structure infections and is able to survive and persist in keratinocytes of the epidermis. Since the evolution of multidrug-resistant bacteria, the use of phages and their lysins has presented a promising alternative approach to treatment. In this study, a cell wall hydrolase (also called lysin) derived from Staphylococcus phage JD007 (JDlys) was identified. JDlys showed strong lytic activity against methicillin-resistant Staphylococcus aureus (MRSA) strains from different sources and of different multilocus sequence typing (MLST) types. Furthermore, a fusion protein consisting of a cell-penetrating peptide derived from the trans-activating transcription (Tat) factor fused to JDlys (CPPTat-JDlys) was used to kill MRSA bacteria causing intracellular infections. CPPTat-JDlys, in which the fusion of CPPTat to JDlys had almost no effect on the bacteriolytic activity of JDlys, was able to effectively eliminate intracellular MRSA bacteria and alleviate the inflammatory response and cell damage caused by MRSA. Specifically, CPPTat-JDlys was able to combat MRSA-induced murine skin infections and, consequently, expedite the healing of cutaneous abscesses. These data suggest that the novel antimicrobial CPP-JDlys may be a worthwhile candidate as a treatment for skin and skin structure infections caused by MRSA.IMPORTANCES. aureus is the main cause of skin and skin structure infections due to its ability to invade and survive in the epithelial barrier. Due to the overuse of antibiotics in humans and animals, S. aureus has shown a high capacity for acquiring and accumulating mechanisms of resistance to antibiotics. Moreover, most antibiotics are usually limited in their ability to overcome the intracellular persistence of bacteria causing skin and skin structure infections. So, it is critical to seek a novel antimicrobial agent to eradicate intracellular S. aureus In this study, a cell-penetrating peptide fused to lysin (CPP-JDlys) was engineered. Our results show that CPP-JDlys can enter keratinocytes and effectively eliminate intracellular MRSA. Meanwhile, experiments with mice revealed that CPP-JDlys efficiently inhibits the proliferation of MRSA in murine skin and thus shortens the course of wound healing. Our results indicate that the CPP-fused lysin has potential for use for the treatment of skin infections caused by MRSA.
Collapse
|
162
|
Dillen CA, Pinsker BL, Marusina AI, Merleev AA, Farber ON, Liu H, Archer NK, Lee DB, Wang Y, Ortines RV, Lee SK, Marchitto MC, Cai SS, Ashbaugh AG, May LS, Holland SM, Freeman AF, Miller LG, Yeaman MR, Simon SI, Milner JD, Maverakis E, Miller LS. Clonally expanded γδ T cells protect against Staphylococcus aureus skin reinfection. J Clin Invest 2018; 128:1026-1042. [PMID: 29400698 PMCID: PMC5824877 DOI: 10.1172/jci96481] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 12/19/2017] [Indexed: 12/19/2022] Open
Abstract
The mechanisms that mediate durable protection against Staphylococcus aureus skin reinfections are unclear, as recurrences are common despite high antibody titers and memory T cells. Here, we developed a mouse model of S. aureus skin reinfection to investigate protective memory responses. In contrast with WT mice, IL-1β-deficient mice exhibited poor neutrophil recruitment and bacterial clearance during primary infection that was rescued during secondary S. aureus challenge. The γδ T cells from skin-draining LNs utilized compensatory T cell-intrinsic TLR2/MyD88 signaling to mediate rescue by trafficking and producing TNF and IFN-γ, which restored neutrophil recruitment and promoted bacterial clearance. RNA-sequencing (RNA-seq) of the LNs revealed a clonotypic S. aureus-induced γδ T cell expansion with a complementarity-determining region 3 (CDR3) aa sequence identical to that of invariant Vγ5+ dendritic epidermal T cells. However, this T cell receptor γ (TRG) aa sequence of the dominant CDR3 sequence was generated from multiple gene rearrangements of TRGV5 and TRGV6, indicating clonotypic expansion. TNF- and IFN-γ-producing γδ T cells were also expanded in peripheral blood of IRAK4-deficient humans no longer predisposed to S. aureus skin infections. Thus, clonally expanded γδ T cells represent a mechanism for long-lasting immunity against recurrent S. aureus skin infections.
Collapse
Affiliation(s)
- Carly A. Dillen
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bret L. Pinsker
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alina I. Marusina
- Department of Dermatology, School of Medicine, UCD, Sacramento, California, USA
| | | | - Orly N. Farber
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nathan K. Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Da B. Lee
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Roger V. Ortines
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Steven K. Lee
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mark C. Marchitto
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shuting S. Cai
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alyssa G. Ashbaugh
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Larissa S. May
- Department of Emergency Medicine, School of Medicine, UCD, Sacramento, California, USA
| | - Steven M. Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | | | - Michael R. Yeaman
- Division of Infectious Diseases
- Division of Molecular Medicine, and
- St. John’s Cardiovascular Research Center, Los Angeles Biomedical Research Institute, Harbor–UCLA Medical Center, Torrance, California, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Scott I. Simon
- Department of Biomedical Engineering, UCD, Davis, California, USA
| | - Joshua D. Milner
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Emanual Maverakis
- Department of Dermatology, School of Medicine, UCD, Sacramento, California, USA
| | - Lloyd S. Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Division of Infectious Diseases, and
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
163
|
Neutralizing Alpha-Toxin Accelerates Healing of Staphylococcus aureus-Infected Wounds in Nondiabetic and Diabetic Mice. Antimicrob Agents Chemother 2018; 62:AAC.02288-17. [PMID: 29311091 DOI: 10.1128/aac.02288-17] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/03/2018] [Indexed: 12/26/2022] Open
Abstract
Staphylococcus aureus wound infections delay healing and result in invasive complications such as osteomyelitis, especially in the setting of diabetic foot ulcers. In preclinical animal models of S. aureus skin infection, antibody neutralization of alpha-toxin (AT), an S. aureus-secreted pore-forming cytolytic toxin, reduces disease severity by inhibiting skin necrosis and restoring effective host immune responses. However, whether therapeutic neutralization of alpha-toxin is effective against S. aureus-infected wounds is unclear. Herein, the efficacy of prophylactic treatment with a human neutralizing anti-AT monoclonal antibody (MAb) was evaluated in an S. aureus skin wound infection model in nondiabetic and diabetic mice. In both nondiabetic and diabetic mice, anti-AT MAb treatment decreased wound size and bacterial burden and enhanced reepithelialization and wound resolution compared to control MAb treatment. Anti-AT MAb had distinctive effects on the host immune response, including decreased neutrophil and increased monocyte and macrophage infiltrates in nondiabetic mice and decreased neutrophil extracellular traps (NETs) in diabetic mice. Similar therapeutic efficacy was achieved with an active vaccine targeting AT. Taken together, neutralization of AT had a therapeutic effect against S. aureus-infected wounds in both nondiabetic and diabetic mice that was associated with differential effects on the host immune response.
Collapse
|
164
|
Chamoun MN, Blumenthal A, Sullivan MJ, Schembri MA, Ulett GC. Bacterial pathogenesis and interleukin-17: interconnecting mechanisms of immune regulation, host genetics, and microbial virulence that influence severity of infection. Crit Rev Microbiol 2018; 44:465-486. [PMID: 29345518 DOI: 10.1080/1040841x.2018.1426556] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Interleukin-17 (IL-17) is a pro-inflammatory cytokine involved in the control of many different disorders, including autoimmune, oncogenic, and diverse infectious diseases. In the context of infectious diseases, IL-17 protects the host against various classes of microorganisms but, intriguingly, can also exacerbate the severity of some infections. The regulation of IL-17 expression stems, in part, from the activity of Interleukin-23 (IL-23), which drives the maturation of different classes of IL-17-producing cells that can alter the course of infection. In this review, we analyze IL-17/IL-23 signalling in bacterial infection, and examine the interconnecting mechanisms that link immune regulation, host genetics, and microbial virulence in the context of bacterial pathogenesis. We consider the roles of IL-17 in both acute and chronic bacterial infections, with a focus on mouse models of human bacterial disease that involve infection of mucosal surfaces in the lungs, urogenital, and gastrointestinal tracts. Polymorphisms in IL-17-encoding genes in humans, which have been associated with heightened host susceptibility to some bacterial pathogens, are discussed. Finally, we examine the implications of IL-17 biology in infectious diseases for the development of novel therapeutic strategies targeted at preventing bacterial infection.
Collapse
Affiliation(s)
- Michelle N Chamoun
- a School of Medical Science, and Menzies Health Institute Queensland , Griffith University , Southport , Australia
| | - Antje Blumenthal
- b The University of Queensland Diamantina Institute, Translational Research Institute , Brisbane , Australia
| | - Matthew J Sullivan
- a School of Medical Science, and Menzies Health Institute Queensland , Griffith University , Southport , Australia
| | - Mark A Schembri
- c School of Chemistry and Molecular Biosciences, and Australian Infectious Disease Research Centre , The University of Queensland , Brisbane , Australia
| | - Glen C Ulett
- a School of Medical Science, and Menzies Health Institute Queensland , Griffith University , Southport , Australia
| |
Collapse
|
165
|
Ruzek MC, Huang L, Zhang TT, Bryant S, Slivka PF, Cuff CA, Tripp C, Blaich G. Dual Blockade of Interleukin-1β and Interleukin-17A Reduces Murine Arthritis Pathogenesis but Also Leads to Spontaneous Skin Infections in Nonhuman Primates. J Pharmacol Exp Ther 2018; 364:474-484. [DOI: 10.1124/jpet.117.243493] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/12/2017] [Indexed: 01/10/2023] Open
|
166
|
Abstract
Safe and efficacious vaccines are arguably the most successful medical interventions of all time. Yet the ongoing discovery of new pathogens, along with emergence of antibiotic-resistant pathogens and a burgeoning population at risk of such infections, imposes unprecedented public health challenges. To meet these challenges, innovative strategies to discover and develop new or improved anti-infective vaccines are necessary. These approaches must intersect the most meaningful insights into protective immunity and advanced technologies with capabilities to deliver immunogens for optimal immune protection. This goal is considered through several recent advances in host-pathogen relationships, conceptual strides in vaccinology, and emerging technologies. Given a clear and growing risk of pandemic disease should the threat of infection go unmet, developing vaccines that optimize protective immunity against high-priority and antibiotic-resistant pathogens represents an urgent and unifying imperative.
Collapse
Affiliation(s)
- Michael R Yeaman
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90024.,Division of Molecular Medicine, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California 90509; .,Division of Infectious Diseases, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California 90509.,Los Angeles Biomedical Research Institute, Torrance, California 90502
| | | |
Collapse
|
167
|
Liu X, Pichulik T, Wolz OO, Dang TM, Stutz A, Dillen C, Delmiro Garcia M, Kraus H, Dickhöfer S, Daiber E, Münzenmayer L, Wahl S, Rieber N, Kümmerle-Deschner J, Yazdi A, Franz-Wachtel M, Macek B, Radsak M, Vogel S, Schulte B, Walz JS, Hartl D, Latz E, Stilgenbauer S, Grimbacher B, Miller L, Brunner C, Wolz C, Weber ANR. Human NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasome activity is regulated by and potentially targetable through Bruton tyrosine kinase. J Allergy Clin Immunol 2017; 140:1054-1067.e10. [PMID: 28216434 DOI: 10.1016/j.jaci.2017.01.017] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 12/23/2016] [Accepted: 01/11/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND The Nod-like receptor NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) and Bruton tyrosine kinase (BTK) are protagonists in innate and adaptive immunity, respectively. NLRP3 senses exogenous and endogenous insults, leading to inflammasome activation, which occurs spontaneously in patients with Muckle-Wells syndrome; BTK mutations cause the genetic immunodeficiency X-linked agammaglobulinemia (XLA). However, to date, few proteins that regulate NLRP3 inflammasome activity in human primary immune cells have been identified, and clinically promising pharmacologic targeting strategies remain elusive. OBJECTIVE We sought to identify novel regulators of the NLRP3 inflammasome in human cells with a view to exploring interference with inflammasome activity at the level of such regulators. METHODS After proteome-wide phosphoproteomics, the identified novel regulator BTK was studied in human and murine cells by using pharmacologic and genetic BTK ablation. RESULTS Here we show that BTK is a critical regulator of NLRP3 inflammasome activation: pharmacologic (using the US Food and Drug Administration-approved inhibitor ibrutinib) and genetic (in patients with XLA and Btk knockout mice) BTK ablation in primary immune cells led to reduced IL-1β processing and secretion in response to nigericin and the Staphylococcus aureus toxin leukocidin AB (LukAB). BTK affected apoptosis-associated speck-like protein containing a CARD (ASC) speck formation and caspase-1 cleavage and interacted with NLRP3 and ASC. S aureus infection control in vivo and IL-1β release from cells of patients with Muckle-Wells syndrome were impaired by ibrutinib. Notably, IL-1β processing and release from immune cells isolated from patients with cancer receiving ibrutinib therapy were reduced. CONCLUSION Our data suggest that XLA might result in part from genetic inflammasome deficiency and that NLRP3 inflammasome-linked inflammation could potentially be targeted pharmacologically through BTK.
Collapse
Affiliation(s)
- Xiao Liu
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Tica Pichulik
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Olaf-Oliver Wolz
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Truong-Minh Dang
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Andrea Stutz
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Carly Dillen
- Department of Dermatology, Johns Hopkins University, Baltimore, Md
| | - Magno Delmiro Garcia
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Helene Kraus
- Centre of Chronic Immunodeficiency, University Hospital Freiburg, Freiburg, Germany
| | - Sabine Dickhöfer
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Ellen Daiber
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Lisa Münzenmayer
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Silke Wahl
- Proteome Center Tübingen, University of Tübingen, Tübingen, Germany
| | - Nikolaus Rieber
- Department of Pediatrics I, University Hospital Tübingen, Tübingen, Germany
| | | | - Amir Yazdi
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | | | - Boris Macek
- Proteome Center Tübingen, University of Tübingen, Tübingen, Germany
| | - Markus Radsak
- Medical Hospital III, University Hospital Mainz, Mainz, Germany
| | - Sebastian Vogel
- Department of Cardiology and Cardiovascular Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Berit Schulte
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Juliane Sarah Walz
- Medical Hospital II (Department of Hematology and Oncology), University Hospital Tübingen, Tübingen, Germany
| | - Dominik Hartl
- Department of Pediatrics I, University Hospital Tübingen, Tübingen, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany; Division of Infectious Diseases & Immunology, University of Massachusetts, Worcester, Mass
| | | | - Bodo Grimbacher
- Centre of Chronic Immunodeficiency, University Hospital Freiburg, Freiburg, Germany
| | - Lloyd Miller
- Department of Dermatology, Johns Hopkins University, Baltimore, Md
| | - Cornelia Brunner
- Department of Otorhinolaryngology, Ulm University Medical Center, Ulm, Germany
| | - Christiane Wolz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Alexander N R Weber
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
168
|
Ng CY, Huang YH, Chu CF, Wu TC, Liu SH. Risks for Staphylococcus aureus colonization in patients with psoriasis: a systematic review and meta-analysis. Br J Dermatol 2017; 177:967-977. [PMID: 28160277 DOI: 10.1111/bjd.15366] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2017] [Indexed: 12/29/2022]
Abstract
Evidence on whether patients with psoriasis have a higher risk for staphylococcal colonization than healthy controls remains controversial. To synthesize the current literature, we performed a systematic review on the prevalence and relative risk (RR) of Staphylococcus aureus colonization in patients with psoriasis. We modified the QUADAS-2 instrument to assess the reporting quality of individual studies and applied random-effects models in meta-analysis. Overall we identified 21 eligible studies, of which 15 enrolled one or more comparison groups. The pooled prevalence of staphylococcal colonization in patients with psoriasis was 35·3% [95% confidence interval (CI) 25·0-45·6] on lesional skin and 39·2% (95% CI 33·7-44·8) in the nares. Patients with psoriasis were 4·5 times more likely to be colonized by S. aureus than healthy controls were on the skin (RR 5·54, 95% CI 3·21-9·57) and 60% more in the nares (RR 1·60, 95% CI 1·11-2·32). Cutaneous and nasal colonization by meticillin-resistant S. aureus also appeared higher in patients with psoriasis (pooled prevalence 8·6%) than in healthy controls (2·6%), yet the difference was not statistically significant (P = 0·74). In contrast, despite of a similar risk for nasal staphylococcal colonization (RR 0·67, 95% CI 0·38-1·18), patients with psoriasis were less likely to carry S. aureus on lesional skin than atopic patients (RR 0·64, 95% CI 0·40-1·02). In summarizing the current literature, we found that patients with psoriasis were at an increased risk for staphylococcal colonization compared with healthy individuals. Prospective studies on how bacterial loads correlate with disease activity can guide the clinical management of bacterial colonization while preventing the emergence of drug-resistant strains.
Collapse
Affiliation(s)
- C Y Ng
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan.,College of Medicine, Chang Gung University, Gueishan District, Taoyuan City, Taiwan.,Graduate Institute of Clinical Medical Sciences, Chang Gung University, Gueishan District, Taoyuan City, Taiwan
| | - Y H Huang
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan.,College of Medicine, Chang Gung University, Gueishan District, Taoyuan City, Taiwan
| | - C F Chu
- College of Medicine, Chang Gung University, Gueishan District, Taoyuan City, Taiwan
| | - T C Wu
- College of Medicine, Chang Gung University, Gueishan District, Taoyuan City, Taiwan
| | - S H Liu
- College of Medicine, Chang Gung University, Gueishan District, Taoyuan City, Taiwan.,Department of Family Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan
| |
Collapse
|
169
|
Abstract
Covering: up to 2017.Natural products are important secondary metabolites produced by bacterial and fungal species that play important roles in cellular growth and signaling, nutrient acquisition, intra- and interspecies communication, and virulence. A subset of natural products is produced by nonribosomal peptide synthetases (NRPSs), a family of large, modular enzymes that function in an assembly line fashion. Because of the pharmaceutical activity of many NRPS products, much effort has gone into the exploration of their biosynthetic pathways and the diverse products they make. Many interesting NRPS pathways have been identified and characterized from both terrestrial and marine bacterial sources. Recently, several NRPS pathways in human commensal bacterial species have been identified that produce molecules with antibiotic activity, suggesting another source of interesting NRPS pathways may be the commensal and pathogenic bacteria that live on the human body. The ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.) have been identified as a significant cause of human bacterial infections that are frequently multidrug resistant. The emerging resistance profile of these organisms has prompted calls from multiple international agencies to identify novel antibacterial targets and develop new approaches to treat infections from ESKAPE pathogens. Each of these species contains several NRPS biosynthetic gene clusters. While some have been well characterized and produce known natural products with important biological roles in microbial physiology, others have yet to be investigated. This review catalogs the NRPS pathways of ESKAPE pathogens. The exploration of novel NRPS products may lead to a better understanding of the chemical communication used by human pathogens and potentially to the discovery of novel therapeutic approaches.
Collapse
Affiliation(s)
- Andrew M Gulick
- Hauptman-Woodward Medical Research Institute, 700 Ellicott Street, Buffalo, NY 14203, USA.
| |
Collapse
|
170
|
Falahee PC, Anderson LS, Reynolds MB, Pirir M, McLaughlin BE, Dillen CA, Cheung AL, Miller LS, Simon SI. α-Toxin Regulates Local Granulocyte Expansion from Hematopoietic Stem and Progenitor Cells in Staphylococcus aureus-Infected Wounds. THE JOURNAL OF IMMUNOLOGY 2017; 199:1772-1782. [PMID: 28733486 DOI: 10.4049/jimmunol.1700649] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/21/2017] [Indexed: 12/18/2022]
Abstract
The immune response to Staphylococcus aureus infection in skin involves the recruitment of polymorphonuclear neutrophils (PMNs) from the bone marrow via the circulation and local granulopoiesis from hematopoietic stem and progenitor cells (HSPCs) that also traffic to infected skin wounds. We focus on regulation of PMN number and function and the role of pore-forming α-toxin (AT), a virulence factor that causes host cell lysis and elicits inflammasome-mediated IL-1β secretion in wounds. Infection with wild-type S. aureus enriched in AT reduced PMN recruitment and resulted in sustained bacterial burden and delayed wound healing. In contrast, PMN recruitment to wounds infected with an isogenic AT-deficient S. aureus strain was unimpeded, exhibiting efficient bacterial clearance and hastened wound resolution. HSPCs recruited to infected wounds were unaffected by AT production and were activated to expand PMN numbers in proportion to S. aureus abundance in a manner regulated by TLR2 and IL-1R signaling. Immunodeficient MyD88-knockout mice infected with S. aureus experienced lethal sepsis that was reversed by PMN expansion mediated by injection of wild-type HSPCs directly into wounds. We conclude that AT-induced IL-1β promotes local granulopoiesis and effective resolution of S. aureus-infected wounds, revealing a potential antibiotic-free strategy for tuning the innate immune response to treat methicillin-resistant S. aureus infection in immunodeficient patients.
Collapse
Affiliation(s)
- Patrick C Falahee
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616
| | - Leif S Anderson
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616
| | - Mack B Reynolds
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616
| | - Mauricio Pirir
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616
| | - Bridget E McLaughlin
- Comprehensive Cancer Center Flow Cytometry Shared Resource, University of California, Davis, Davis, CA 95616
| | - Carly A Dillen
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231; and
| | - Ambrose L Cheung
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231; and
| | - Scott I Simon
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616;
| |
Collapse
|
171
|
Scumpia PO, Botten GA, Norman JS, Kelly-Scumpia KM, Spreafico R, Ruccia AR, Purbey PK, Thomas BJ, Modlin RL, Smale ST. Opposing roles of Toll-like receptor and cytosolic DNA-STING signaling pathways for Staphylococcus aureus cutaneous host defense. PLoS Pathog 2017; 13:e1006496. [PMID: 28704551 PMCID: PMC5526579 DOI: 10.1371/journal.ppat.1006496] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/25/2017] [Accepted: 06/27/2017] [Indexed: 01/01/2023] Open
Abstract
Successful host defense against pathogens requires innate immune recognition of the correct pathogen associated molecular patterns (PAMPs) by pathogen recognition receptors (PRRs) to trigger the appropriate gene program tailored to the pathogen. While many PRR pathways contribute to the innate immune response to specific pathogens, the relative importance of each pathway for the complete transcriptional program elicited has not been examined in detail. Herein, we used RNA-sequencing with wildtype and mutant macrophages to delineate the innate immune pathways contributing to the early transcriptional response to Staphylococcus aureus, a ubiquitous microorganism that can activate a wide variety of PRRs. Unexpectedly, two PRR pathways—the Toll-like receptor (TLR) and Stimulator of Interferon Gene (STING) pathways—were identified as dominant regulators of approximately 95% of the genes that were potently induced within the first four hours of macrophage infection with live S. aureus. TLR signaling predominantly activated a pro-inflammatory program while STING signaling activated an antiviral/type I interferon response with live but not killed S. aureus. This STING response was largely dependent on the cytosolic DNA sensor cyclic guanosine-adenosine synthase (cGAS). Using a cutaneous infection model, we found that the TLR and STING pathways played opposite roles in host defense to S. aureus. TLR signaling was required for host defense, with its absence reducing interleukin (IL)-1β production and neutrophil recruitment, resulting in increased bacterial growth. In contrast, absence of STING signaling had the opposite effect, enhancing the ability to restrict the infection. These results provide novel insights into the complex interplay of innate immune signaling pathways triggered by S. aureus and uncover opposing roles of TLR and STING in cutaneous host defense to S. aureus. Individual pathogen associated molecular patterns (PAMPs) induce gene expression in immune cells through distinct signaling pathways to protect cells from infection. However, pathogens typically possess many PAMPs, and the precise contribution of each PAMP to the gene expression program elicited by a live pathogen has not been clearly defined. Herein, we used gene expression profiling to examine the full early response of macrophages to Staphylococcus aureus, a major human opportunistic pathogen. Surprisingly, we found that two pathogen-sensing pathways, Toll-like receptor (TLR) and Stimulator of Interferon Signaling Gene (STING) pathways, contribute to the activation of ~95% of the genes induced by S. aureus infection. The remaining genes may be induced by hypoxia pathways. When the bacterium is dead, 98% of the gene induction occurs through TLR signaling, and neither STING nor hypoxia contributes greatly to the response. STING activation requires sensing of S. aureus DNA by the cytosolic DNA sensor, cGAS. During S. aureus skin infection, the TLR and STING pathways compete with each other to induce or suppress host defense, respectively, by counter regulating interleukin 1β production and neutrophil recruitment. A similar approach may allow delineation of the relative contributions of immune pathways in the response to various live pathogens.
Collapse
Affiliation(s)
- Philip O Scumpia
- Department of Medicine, Division of Dermatology, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Giovanni A Botten
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Joshua S Norman
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Kindra M Kelly-Scumpia
- Department of Medicine, Division of Dermatology, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Roberto Spreafico
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America.,Institute for Quantitative and Computational Biosciences, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Amber R Ruccia
- Department of Medicine, Division of Dermatology, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Prabhat K Purbey
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Brandon J Thomas
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Robert L Modlin
- Department of Medicine, Division of Dermatology, University of California at Los Angeles, Los Angeles, California, United States of America.,Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Stephen T Smale
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
172
|
Propofol Sedation Exacerbates Kidney Pathology and Dissemination of Bacteria during Staphylococcus aureus Bloodstream Infections. Infect Immun 2017; 85:IAI.00097-17. [PMID: 28461390 DOI: 10.1128/iai.00097-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/13/2017] [Indexed: 02/06/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is responsible for large numbers of postsurgical nosocomial infections across the United States and worldwide. Propofol anesthesia is widely used in surgery and in intensive care units, and recent evidence indicates that even brief exposure to propofol can substantially increase host susceptibility to microbial infection. Here, we delineate the impact of propofol sedation on MRSA bloodstream infections in mice in the presence and absence of prophylactic antibiotic treatment. Consistent with previous reports, brief periods of anesthesia with propofol were sufficient to significantly increase bacterial burdens and kidney pathology in mice infected with MRSA. Propofol exposure increased neutrophilic infiltrates into the kidney and enhanced bacterial dissemination throughout kidney tissue. Propofol sedation reduced populations of effector phagocytes and mature dendritic cells within the kidney and led to the apparent expansion of myeloid-derived suppressor cell-like populations. When propofol was coadministered with vancomycin prophylaxis, it dramatically increased kidney abscess formation and bacterial dissemination throughout kidney tissue at early times post-S. aureus infection compared to antibiotic-treated but nonsedated animals. Taken together, our data indicate that short-term sedation with propofol significantly increases the severity of bloodstream MRSA infection, even when administered in conjunction with vancomycin prophylaxis.
Collapse
|
173
|
Rerknimitr P, Otsuka A, Nakashima C, Kabashima K. The etiopathogenesis of atopic dermatitis: barrier disruption, immunological derangement, and pruritus. Inflamm Regen 2017; 37:14. [PMID: 29259713 PMCID: PMC5725646 DOI: 10.1186/s41232-017-0044-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/12/2017] [Indexed: 02/07/2023] Open
Abstract
Atopic dermatitis (AD) is a common chronic skin inflammatory disorder characterized by recurrent eczema accompanied by an intractable itch that leads to an impaired quality of life. Extensive recent studies have shed light on the multifaceted pathogenesis of the disease. The complex interplay among skin barrier deficiency, immunological derangement, and pruritus contributes to the development, progression, and chronicity of the disease. Abnormalities in filaggrin, other stratum corneum constituents, and tight junctions induce and/or promote skin inflammation. This inflammation, in turn, can further deteriorate the barrier function by downregulating a myriad of essential barrier-maintaining molecules. Pruritus in AD, which may be due to hyperinnervation of the epidermis, increases pruritogens, and central sensitization compromises the skin integrity and promotes inflammation. There are unmet needs in the treatment of AD. Based on the detailed evidence available to date, certain disease mechanisms can be chosen as treatment targets. Numerous clinical trials of biological agents are currently being conducted and are expected to provide treatments for patients suffering from AD in the future. This review summarizes the etiopathogenesis of the disease and provides a rationale for choosing the novel targeted therapy that will be available in the future.
Collapse
Affiliation(s)
- Pawinee Rerknimitr
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawara, Sakyo, Kyoto, 606-8507 Japan.,Division of Dermatology, Department of Medicine, Faculty of Medicine, Skin and Allergy Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Atsushi Otsuka
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawara, Sakyo, Kyoto, 606-8507 Japan
| | - Chisa Nakashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawara, Sakyo, Kyoto, 606-8507 Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawara, Sakyo, Kyoto, 606-8507 Japan.,Singapore Immunology Network (SIgN) and Institute of Medical Biology, Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore
| |
Collapse
|
174
|
Ebner F, Sedlyarov V, Tasciyan S, Ivin M, Kratochvill F, Gratz N, Kenner L, Villunger A, Sixt M, Kovarik P. The RNA-binding protein tristetraprolin schedules apoptosis of pathogen-engaged neutrophils during bacterial infection. J Clin Invest 2017; 127:2051-2065. [PMID: 28504646 PMCID: PMC5451238 DOI: 10.1172/jci80631] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/16/2017] [Indexed: 02/06/2023] Open
Abstract
Protective responses against pathogens require a rapid mobilization of resting neutrophils and the timely removal of activated ones. Neutrophils are exceptionally short-lived leukocytes, yet it remains unclear whether the lifespan of pathogen-engaged neutrophils is regulated differently from that in the circulating steady-state pool. Here, we have found that under homeostatic conditions, the mRNA-destabilizing protein tristetraprolin (TTP) regulates apoptosis and the numbers of activated infiltrating murine neutrophils but not neutrophil cellularity. Activated TTP-deficient neutrophils exhibited decreased apoptosis and enhanced accumulation at the infection site. In the context of myeloid-specific deletion of Ttp, the potentiation of neutrophil deployment protected mice against lethal soft tissue infection with Streptococcus pyogenes and prevented bacterial dissemination. Neutrophil transcriptome analysis revealed that decreased apoptosis of TTP-deficient neutrophils was specifically associated with elevated expression of myeloid cell leukemia 1 (Mcl1) but not other antiapoptotic B cell leukemia/lymphoma 2 (Bcl2) family members. Higher Mcl1 expression resulted from stabilization of Mcl1 mRNA in the absence of TTP. The low apoptosis rate of infiltrating TTP-deficient neutrophils was comparable to that of transgenic Mcl1-overexpressing neutrophils. Our study demonstrates that posttranscriptional gene regulation by TTP schedules the termination of the antimicrobial engagement of neutrophils. The balancing role of TTP comes at the cost of an increased risk of bacterial infections.
Collapse
Affiliation(s)
- Florian Ebner
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Vitaly Sedlyarov
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Saren Tasciyan
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Masa Ivin
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | | | - Nina Gratz
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Lukas Kenner
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Unit of Pathology of Laboratory Animals (UPLA), University of Veterinary Medicine Vienna, Vienna, Austria
| | - Andreas Villunger
- Medical University of Innsbruck, Division of Developmental Immunology, Innsbruck, Austria
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Michael Sixt
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Pavel Kovarik
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| |
Collapse
|
175
|
Antigen delivery to dendritic cells shapes human CD4+ and CD8+ T cell memory responses to Staphylococcus aureus. PLoS Pathog 2017; 13:e1006387. [PMID: 28542586 PMCID: PMC5444865 DOI: 10.1371/journal.ppat.1006387] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/27/2017] [Indexed: 12/15/2022] Open
Abstract
Intracellular persistence of Staphylococcus aureus favors bacterial spread and chronic infections. Here, we provide evidence for the existence of human CD4+ and CD8+ T cell memory against staphylococcal antigens. Notably, the latter could provide a missing link in our understanding of immune control of intracellular S. aureus. The analyses showed that pulsing of monocyte-derived dendritic cells (MoDC) with native staphylococcal protein antigens induced release of Th2-associated cytokines and mediators linked to T regulatory cell development (G-CSF, IL-2 and IL-10) from both CD4+ and CD8+ T cells, thus revealing a state of tolerance predominantly arising from preformed memory T cells. Furthermore, G-CSF was identified as a suppressor of CD8+ T cell-derived IFNγ secretion, thus confirming a tolerogenic role of this cytokine in the regulation of T cell responses to S. aureus. Nevertheless, delivery of in vitro transcribed mRNA-encoded staphylococcal antigens triggered Th1-biased responses, e.g. IFNγ and TNF release from both naïve and memory T cells. Collectively, our data highlight the potential of mRNA-adjuvanted antigen presentation to enable inflammatory responses, thus overriding the existing Th2/Treg-biased memory T cell response to native S. aureus antigens. Staphylococcus aureus is deemed one of the most important nosocomial pathogens but, to date, there are no safe and protective vaccines. In this study we investigate the nature of the preformed T cell response to S. aureus antigens in healthy donors. Our data reveal that CD4+ and—so far not described—CD8+ T cell memory responses against native staphylococcal antigens exist but are skewed towards minimizing inflammation and promoting tolerance. The T cell response to staphylococcal antigens is characterized by the secretion of typical Th2 cytokines such as IL-5 and IL-13 and mediators associated with formation of T regulatory cells. Most importantly, G-CSF suppresses IFNγ release from pre-existent memory T cells. However, our data reveal that the use of mRNA-encoded antigens to trigger S. aureus-specific T cell responses bears the potential to override the tolerogenic bias. It favors TNF- and IFNγ-releasing T cells and may, thus, represent an innovative tool in prophylactic and therapeutic vaccine development.
Collapse
|
176
|
Bishayi B, Nandi A, Dey R, Adhikary R. Expression of CXCR1 (IL-8 receptor A) in splenic, peritoneal macrophages and resident bone marrow cells after acute live or heat killed Staphylococcus aureus stimulation in mice. Microb Pathog 2017; 109:131-150. [PMID: 28552636 DOI: 10.1016/j.micpath.2017.05.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/15/2017] [Accepted: 05/19/2017] [Indexed: 10/19/2022]
Abstract
Literature reveals that interaction with live Staphylococcus aureus (S. aureus) or heat killed S. aureus (HKSA) promotes secretion of CXCL-8 or interleukin-8 (IL-8) from leukocytes, however, the expressions of CXCR1 in murine splenic (SPM), peritoneal macrophages (PM) and resident fresh bone marrow cells (FBMC) have not been identified. Currently, very few studies are available on the functional characterization of CXCR1 in mouse macrophage subtypes and its modulation in relation to acute S. aureus infection. SPM, PM and FBMCs were infected with viable S. aureus or stimulated with HKSA in presence and absence of anti-CXCR1 antibody in this study. We reported here that CXCR1 was not constitutively expressed by macrophage subtypes and the receptor was induced only after S. aureus stimulation. The CXCR1 band was found specific as we compared with human polymorphonuclear neutrophils (PMNs) as a positive control (data not shown). Although, we did not show that secreted IL-8 from S. aureus-infected macrophages promotes migration of PMNs. Blocking of cell surface CXCR1 decreases the macrophage's ability to clear staphylococcal infection, attenuates proinflammatory cytokine production and the increased catalase and decreased superoxide dismutase (SOD) enzymes of the bacteria might indicate their role in scavenging macrophage derived hydrogen peroxide (H2O2). The decreased levels of cytokines due to CXCR1 blockade before S. aureus infection appear to regulate the killing of bacteria by destroying H2O2 and nitric oxide (NO). Moreover, functional importance of macrophage subpopulation heterogeneity might be important in designing new effective approaches to limit S. aureus infection induced inflammation and cytotoxicity.
Collapse
Affiliation(s)
- Biswadev Bishayi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India.
| | - Ajeya Nandi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India
| | - Rajen Dey
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India
| | - Rana Adhikary
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India
| |
Collapse
|
177
|
Kaplan A, Lee MW, Wolf AJ, Limon JJ, Becker CA, Ding M, Murali R, Lee EY, Liu GY, Wong GCL, Underhill DM. Direct Antimicrobial Activity of IFN-β. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:4036-4045. [PMID: 28411186 PMCID: PMC5469413 DOI: 10.4049/jimmunol.1601226] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 03/10/2017] [Indexed: 01/08/2023]
Abstract
Type I IFNs are a cytokine family essential for antiviral defense. More recently, type I IFNs were shown to be important during bacterial infections. In this article, we show that, in addition to known cytokine functions, IFN-β is antimicrobial. Parts of the IFN-β molecular surface (especially helix 4) are cationic and amphipathic, both classic characteristics of antimicrobial peptides, and we observed that IFN-β can directly kill Staphylococcus aureus Further, a mutant S. aureus that is more sensitive to antimicrobial peptides was killed more efficiently by IFN-β than was the wild-type S. aureus, and immunoblotting showed that IFN-β interacts with the bacterial cell surface. To determine whether specific parts of IFN-β are antimicrobial, we synthesized IFN-β helix 4 and found that it is sufficient to permeate model prokaryotic membranes using synchrotron x-ray diffraction and that it is sufficient to kill S. aureus These results suggest that, in addition to its well-known signaling activity, IFN-β may be directly antimicrobial and be part of a growing family of cytokines and chemokines, called kinocidins, that also have antimicrobial properties.
Collapse
Affiliation(s)
- Amber Kaplan
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Michelle W Lee
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095
| | - Andrea J Wolf
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Jose J Limon
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Courtney A Becker
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Minna Ding
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Ramachandran Murali
- Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| | - Ernest Y Lee
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095
| | - George Y Liu
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095
- Division of Pediatric Infectious Diseases, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Gerard C L Wong
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095;
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095
| | - David M Underhill
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048;
- Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| |
Collapse
|
178
|
Sampedro GR, Bubeck Wardenburg J. Staphylococcus aureus in the Intensive Care Unit: Are These Golden Grapes Ripe for a New Approach? J Infect Dis 2017; 215:S64-S70. [PMID: 28003353 DOI: 10.1093/infdis/jiw581] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Staphylococcus aureus is the leading cause of infection in the setting of critical illness and injury. This pathogen causes life-threatening infection in otherwise healthy individuals and also complicates the clinical course of patients requiring intensive care as a result of their primary medical or surgical disease processes. S. aureus infection in the intensive care unit (ICU) most commonly manifests as sepsis, ventilator-associated pneumonia, and infection of surgical sites and indwelling medical devices. With the epidemic spread of methicillin-resistant S. aureus, many cases of staphylococcal infection in the ICU are now classified as drug resistant, prompting hospital-based screening for methicillin-resistant S. aureus and implementation of both isolation practices and decolonization strategies in ICU patients. The genetic adaptability of S. aureus, heterogeneity of disease presentation, clinical course, and outcome between individual S. aureus-infected ICU patients remains enigmatic, suggesting a need to define disease classification subtypes that inform disease progression and therapy. We propose that S. aureus infection in the ICU now presents a unique opportunity for individualized risk stratification coupled with the investigation of novel approaches to mitigate disease. Given our increasing knowledge of the molecular pathogenesis of S. aureus disease, we suggest that the application of molecular pathological epidemiology to S. aureus infection can usher in a new era of highly focused personalized therapy that may be particularly beneficial in the setting of critical illness and injury.
Collapse
Affiliation(s)
- Georgia R Sampedro
- Departments of 1 Microbiology and.,Pediatrics, University of Chicago, Illinois
| | | |
Collapse
|
179
|
Gough P, Ganesan S, Datta SK. IL-20 Signaling in Activated Human Neutrophils Inhibits Neutrophil Migration and Function. THE JOURNAL OF IMMUNOLOGY 2017; 198:4373-4382. [PMID: 28424238 DOI: 10.4049/jimmunol.1700253] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 03/23/2017] [Indexed: 12/23/2022]
Abstract
Neutrophils possess multiple antimicrobial mechanisms that are critical for protection of the host against infection with extracellular microbes, such as the bacterial pathogen Staphylococcus aureus Recruitment and activation of neutrophils at sites of infection are driven by cytokine and chemokine signals that directly target neutrophils via specific cell surface receptors. The IL-20 subfamily of cytokines has been reported to act at epithelial sites and contribute to psoriasis, wound healing, and anti-inflammatory effects during S. aureus infection. However, the ability of these cytokines to directly affect neutrophil function remains incompletely understood. In this article, we show that human neutrophils altered their expression of IL-20R chains upon migration and activation in vivo and in vitro. Such activation of neutrophils under conditions mimicking infection with S. aureus conferred responsiveness to IL-20 that manifested as modification of actin polymerization and inhibition of a broad range of actin-dependent functions, including phagocytosis, granule exocytosis, and migration. Consistent with the previously described homeostatic and anti-inflammatory properties of IL-20 on epithelial cells, the current study provides evidence that IL-20 directly targets and inhibits key inflammatory functions of neutrophils during infection with S. aureus.
Collapse
Affiliation(s)
- Portia Gough
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Sundar Ganesan
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Sandip K Datta
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| |
Collapse
|
180
|
Landrum ML, Lalani T, Niknian M, Maguire JD, Hospenthal DR, Fattom A, Taylor K, Fraser J, Wilkins K, Ellis MW, Kessler PD, Fahim REF, Tribble DR. Safety and immunogenicity of a recombinant Staphylococcus aureus α-toxoid and a recombinant Panton-Valentine leukocidin subunit, in healthy adults. Hum Vaccin Immunother 2017; 13:791-801. [PMID: 28010246 PMCID: PMC5404372 DOI: 10.1080/21645515.2016.1248326] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/29/2016] [Accepted: 10/11/2016] [Indexed: 01/01/2023] Open
Abstract
We conducted a randomized, double-blind, placebo-controlled dose-escalation study in healthy adults to evaluate the safety and immunogenicity of recombinant Staphylococcus aureus candidate vaccine antigens, recombinant α-toxoid (rAT) and a sub-unit of Panton-Valentine leukocidin (rLukS-PV). 176 subjects were enrolled and randomized within 1 of 11 treatment cohorts: monovalent rAT or rLukS-PV dosages of 10, 25, 50, and 100 μg; bivalent rAT:rLukS dosages of 10:10, 25:25, and 50:50 μg; and alum or saline placebo. All subjects were assessed at Days 0, 7, 14, 28, and 84. Subjects in the 50:50 μg bivalent cohort received a second injection on Day 84 and were assessed on Days 98 and 112. Incidence and severity of reactogenicity and adverse events (AEs) were compared. Geometric mean serum concentrations (GMC) and neutralizing activity of anti-rAT and anti-rLukS-PV IgG were assessed. Reactogenicity incidence was significantly higher in vaccine than placebo recipients (77% versus 55%, respectively; p = 0.006). However, 77% of reactogenicity events were mild and 19% were moderate in severity. The AE incidence and severity were similar between the cohorts. All monovalent and bivalent rAT dosages resulted in a significant increase in the anti-rAT IgG and anti- rLukS-PV GMCs between day 0 and 28 compared with placebo, and persisted through Day 84. Exploratory subgroup analyses suggested a higher GMC and neutralizing antibody titers for the 50 μg monovalent or bivalent rAT and rLukS-PV dose as compared to the other doses. No booster effect was observed after administration of the second dose. We conclude that the rAT and rLukS-PV vaccine formulations were well-tolerated and had a favorable immunogenicity profile, producing antibody with neutralizing activity through day 84. There was no benefit observed with a booster dose of the vaccine.
Collapse
Affiliation(s)
- Michael L. Landrum
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Rockville, MD, USA
- Division of Infectious Diseases, San Antonio Military Medical Center, Fort Sam Houston, TX, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Tahaniyat Lalani
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Rockville, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Division of Infectious Diseases, Naval Medical Center Portsmouth, Portsmouth, VA, USA
| | | | - Jason D. Maguire
- Division of Infectious Diseases, Naval Medical Center Portsmouth, Portsmouth, VA, USA
| | - Duane R. Hospenthal
- Division of Infectious Diseases, San Antonio Military Medical Center, Fort Sam Houston, TX, USA
| | - Ali Fattom
- Nabi Biopharmaceuticals, Rockville, MD, USA
| | | | - Jamie Fraser
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Rockville, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Kenneth Wilkins
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Rockville, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Michael W. Ellis
- Infectious Diseases Division, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | | | | | - David R. Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Rockville, MD, USA
| |
Collapse
|
181
|
Bagnoli F. Staphylococcus aureus toxin antibodies: Good companions of antibiotics and vaccines. Virulence 2017; 8:1037-1042. [PMID: 28267417 DOI: 10.1080/21505594.2017.1295205] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
182
|
Wang L, Si W, Xue H, Zhao X. A fibronectin-binding protein (FbpA) of Weissella cibaria inhibits colonization and infection of Staphylococcus aureus in mammary glands. Cell Microbiol 2017; 19. [PMID: 28125161 DOI: 10.1111/cmi.12731] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/24/2017] [Accepted: 01/25/2017] [Indexed: 12/17/2022]
Abstract
Staphylococcus aureus (S. aureus) is a frequent cause of infections in both humans and animals. Probiotics are known to inhibit colonization of pathogens on host tissues. However, mechanisms for the inhibition are still elusive due to complex host-microbe and microbe-microbe interactions. Here, we show that reduced abilities of S. aureus to infect mammary glands in the presence of Weissella cibaria (W. cibaria) were correlated with its poor adherence to mammary epithelial cells. Such inhibition by W. cibaria isolates was at least partially attributed to a fibronectin-binding protein (FbpA) on this lactic acid bacterium. Three W. cibaria isolates containing fbpA had higher inhibitory abilities than other three LAB isolates without the gene. The fbpA-deficient mutant of W. cibaria isolate LW1, LW1ΔfbpA, lost the inhibitory activity to reduce the adhesion of S. aureus to mammary epithelial cells and was less able to reduce the colonization of S. aureus in mammary glands. Expression of FbpA to the surface of LW1ΔfbpA reversed its inhibitory activities. Furthermore, addition of purified FbpA inhibited S. aureus biofilm formation. Our results suggest that W. cibaria FbpA hinders S. aureus colonization and infection through interfering with the S. aureus invasion pathway mediated by fibronectin-binding proteins and inhibiting biofilm formation of S. aureus.
Collapse
Affiliation(s)
- Liangliang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, People's Republic of China.,School of Pharmaceutical Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Wei Si
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, People's Republic of China.,Department of Animal Science, McGill University, Quebec, Canada
| | - Huping Xue
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, People's Republic of China
| | - Xin Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, People's Republic of China.,Department of Animal Science, McGill University, Quebec, Canada
| |
Collapse
|
183
|
Nakatsuji T, Chen TH, Narala S, Chun KA, Two AM, Yun T, Shafiq F, Kotol PF, Bouslimani A, Melnik AV, Latif H, Kim JN, Lockhart A, Artis K, David G, Taylor P, Streib J, Dorrestein PC, Grier A, Gill SR, Zengler K, Hata TR, Leung DYM, Gallo RL. Antimicrobials from human skin commensal bacteria protect against Staphylococcus aureus and are deficient in atopic dermatitis. Sci Transl Med 2017; 9:eaah4680. [PMID: 28228596 PMCID: PMC5600545 DOI: 10.1126/scitranslmed.aah4680] [Citation(s) in RCA: 694] [Impact Index Per Article: 86.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/08/2016] [Accepted: 01/17/2017] [Indexed: 12/21/2022]
Abstract
The microbiome can promote or disrupt human health by influencing both adaptive and innate immune functions. We tested whether bacteria that normally reside on human skin participate in host defense by killing Staphylococcus aureus, a pathogen commonly found in patients with atopic dermatitis (AD) and an important factor that exacerbates this disease. High-throughput screening for antimicrobial activity against S. aureus was performed on isolates of coagulase-negative Staphylococcus (CoNS) collected from the skin of healthy and AD subjects. CoNS strains with antimicrobial activity were common on the normal population but rare on AD subjects. A low frequency of strains with antimicrobial activity correlated with colonization by S. aureus The antimicrobial activity was identified as previously unknown antimicrobial peptides (AMPs) produced by CoNS species including Staphylococcus epidermidis and Staphylococcus hominis These AMPs were strain-specific, highly potent, selectively killed S. aureus, and synergized with the human AMP LL-37. Application of these CoNS strains to mice confirmed their defense function in vivo relative to application of nonactive strains. Strikingly, reintroduction of antimicrobial CoNS strains to human subjects with AD decreased colonization by S. aureus These findings show how commensal skin bacteria protect against pathogens and demonstrate how dysbiosis of the skin microbiome can lead to disease.
Collapse
Affiliation(s)
- Teruaki Nakatsuji
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92092, USA
| | - Tiffany H Chen
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92092, USA
| | - Saisindhu Narala
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92092, USA
| | - Kimberly A Chun
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92092, USA
| | - Aimee M Two
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92092, USA
| | - Tong Yun
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92092, USA
| | - Faiza Shafiq
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92092, USA
| | - Paul F Kotol
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92092, USA
| | - Amina Bouslimani
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92092, USA
| | - Alexey V Melnik
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92092, USA
| | - Haythem Latif
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92092, USA
| | - Ji-Nu Kim
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92092, USA
| | | | - Keli Artis
- Rho Federal Systems Division Inc., Chapel Hill, NC 27517, USA
| | - Gloria David
- Rho Federal Systems Division Inc., Chapel Hill, NC 27517, USA
| | - Patricia Taylor
- Division of Allergy and Immunology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA
| | - Joanne Streib
- Division of Allergy and Immunology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA
| | - Pieter C Dorrestein
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92092, USA
- Departments of Chemistry, Biochemistry and Pharmacology, University of California, San Diego, La Jolla, CA 92092, USA
| | - Alex Grier
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14611, USA
| | - Steven R Gill
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14611, USA
| | - Karsten Zengler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92092, USA
| | - Tissa R Hata
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92092, USA
| | - Donald Y M Leung
- Division of Allergy and Immunology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92092, USA.
| |
Collapse
|
184
|
Kruglikov IL, Wollina U. Local effects of adipose tissue in psoriasis and psoriatic arthritis. PSORIASIS-TARGETS AND THERAPY 2017; 7:17-25. [PMID: 29387604 PMCID: PMC5774600 DOI: 10.2147/ptt.s122959] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The structure and physiological state of the local white adipose tissue (WAT) located underneath the lesional psoriatic skin and inside of the joints affected by psoriatic arthritis play an important role in the pathophysiology of these diseases. WAT pads associated with inflammatory sites in psoriasis and psoriatic arthritis are, correspondingly, dermal WAT and articular adipose tissue; these pads demonstrate inflammatory phenotypes in both diseases. Such local WAT inflammation could be the primary effect in the pathophysiology of psoriasis leading to the modification of the local expression of adipokines, a change in the structure of the basement membrane and the release of keratinocytes with consequent epidermal hyperproliferation during psoriasis. Similar articular adipose tissue inflammation can lead to the induction of structural modifications and synovial inflammation in the joints of patients with psoriatic arthritis.
Collapse
Affiliation(s)
| | - Uwe Wollina
- Department of Dermatology and Allergology, Hospital Dresden-Friedrichstadt Academic Teaching Hospital of the Technical University of Dresden, Dresden, Germany
| |
Collapse
|
185
|
Innate Immune Memory Contributes to Host Defense against Recurrent Skin and Skin Structure Infections Caused by Methicillin-Resistant Staphylococcus aureus. Infect Immun 2017; 85:IAI.00876-16. [PMID: 27849182 DOI: 10.1128/iai.00876-16] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/09/2016] [Indexed: 12/31/2022] Open
Abstract
Staphylococcus aureus is the leading cause of skin and skin structure infections (SSSI). The high frequency of recurring SSSI due to S. aureus, including methicillin-resistant S. aureus (MRSA) strains, despite high titers of specific antibodies and circulating T cells, implies that traditional adaptive immunity imparts incomplete protection. We hypothesized that innate immune memory contributes to the protective host defense against recurring MRSA infection. To test this hypothesis, SSSI was induced in wild-type and rag1-/- mice in the BALB/c and C57BL/6 backgrounds. Prior infection (priming) of wild-type and rag1-/- mice of either background afforded protection against repeat infection, as evidenced by reduced abscess severities and decreased CFU densities compared to those in naive controls. Interestingly, protection was greater on the previously infected flank than on the naive flank for wild-type and rag1-/- mice. For wild-type mice, protective efficacy corresponded to increased infiltration of neutrophils (polymorphonuclear leukocytes [PMN]), macrophages (MΦ), Langerin+ dendritic cells (LDC), and natural killer (NK) cells. Protection was associated with the induction of interleukin-17A (IL-17A), IL-22, and gamma interferon (IFN-γ) as well as the antimicrobial peptides CRAMP and mβD-3. Priming also protected rag1-/- mice against recurring SSSI, with increased MΦ and LDC infiltration and induction of IL-22, CRAMP, and mβD-3. These findings suggest that innate immune memory, mediated by specific cellular and molecular programs, likely contributes to the localized host defense in recurrent MRSA SSSI. These insights support the development of targeted immunotherapeutic strategies to address the challenge of MRSA infection.
Collapse
|
186
|
Mayer-Barber KD, Yan B. Clash of the Cytokine Titans: counter-regulation of interleukin-1 and type I interferon-mediated inflammatory responses. Cell Mol Immunol 2017; 14:22-35. [PMID: 27264686 PMCID: PMC5214938 DOI: 10.1038/cmi.2016.25] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/25/2016] [Accepted: 04/26/2016] [Indexed: 02/07/2023] Open
Abstract
Over the past decades the notion of 'inflammation' has been extended beyond the original hallmarks of rubor (redness), calor (heat), tumor (swelling) and dolor (pain) described by Celsus. We have gained a more detailed understanding of the cellular players and molecular mediators of inflammation which is now being applied and extended to areas of biomedical research such as cancer, obesity, heart disease, metabolism, auto-inflammatory disorders, autoimmunity and infectious diseases. Innate cytokines are often central components of inflammatory responses. Here, we discuss how the type I interferon and interleukin-1 cytokine pathways represent distinct and specialized categories of inflammatory responses and how these key mediators of inflammation counter-regulate each other.
Collapse
Affiliation(s)
- Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bo Yan
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
187
|
Hong SN, Kim J, Sung HH. A Study on Changes in Antimicrobial Resistant Staphylococcus aureus from Wound Isolates in a South Korean University Hospital for the Past 10 Years (2006, 2016). KOREAN JOURNAL OF CLINICAL LABORATORY SCIENCE 2016. [DOI: 10.15324/kjcls.2016.48.4.335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Seong-No Hong
- Department of Clinical Laboratory Science, Dongnam Health University, Suwon 16328, Korea
| | - Joon Kim
- Department of Laboratory Medicine, Ajou University Hospital, Suwon 16499, Korea
| | - Hyun-Ho Sung
- Department of Clinical Laboratory Science, Dongnam Health University, Suwon 16328, Korea
| |
Collapse
|
188
|
Dejani NN, Brandt SL, Piñeros A, Glosson-Byers NL, Wang S, Son YM, Medeiros AI, Serezani CH. Topical Prostaglandin E Analog Restores Defective Dendritic Cell-Mediated Th17 Host Defense Against Methicillin-Resistant Staphylococcus Aureus in the Skin of Diabetic Mice. Diabetes 2016; 65:3718-3729. [PMID: 27605625 PMCID: PMC5127243 DOI: 10.2337/db16-0565] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 08/30/2016] [Indexed: 12/17/2022]
Abstract
People with diabetes are more prone to Staphylococcus aureus skin infection than healthy individuals. Control of S. aureus infection depends on dendritic cell (DC)-induced T-helper 17 (Th17)-mediated neutrophil recruitment and bacterial clearance. DC ingestion of infected apoptotic cells (IACs) drive prostaglandin E2 (PGE2) secretion to generate Th17 cells. We speculated that hyperglycemia inhibits skin DC migration to the lymph nodes and impairs the Th17 differentiation that accounts for poor skin host defense in diabetic mice. Diabetic mice showed increased skin lesion size and bacterial load and decreased PGE2 secretion and Th17 cells compared with nondiabetic mice after methicillin-resistant S. aureus (MRSA) infection. Bone marrow-derived DCs (BMDCs) cultured in high glucose (25 mmol/L) exhibited decreased Ptges mRNA expression, PGE2 production, lower CCR7-dependent DC migration, and diminished maturation after recognition of MRSA-IACs than BMDCs cultured in low glucose (5 mmol/L). Similar events were observed in DCs from diabetic mice infected with MRSA. Topical treatment of diabetic mice with the PGE analog misoprostol improved host defense against MRSA skin infection by restoring DC migration to draining lymph nodes, Th17 differentiation, and increased antimicrobial peptide expression. These findings identify a novel mechanism involved in poor skin host defense in diabetes and propose a targeted strategy to restore skin host defense in diabetes.
Collapse
Affiliation(s)
- Naiara N Dejani
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
- University of São Paulo, Ribeirão Preto, Brazil
- Departamento de Ciências Biológicas, Faculdade de Ciências Farmacêuticas, Universidade Estadual Paulista "Júlio de Mesquita Filho," Araraquara, Brazil
| | - Stephanie L Brandt
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Annie Piñeros
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
- University of São Paulo, Ribeirão Preto, Brazil
| | - Nicole L Glosson-Byers
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Sue Wang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Young Min Son
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| | - Alexandra I Medeiros
- University of São Paulo, Ribeirão Preto, Brazil
- Departamento de Ciências Biológicas, Faculdade de Ciências Farmacêuticas, Universidade Estadual Paulista "Júlio de Mesquita Filho," Araraquara, Brazil
| | - C Henrique Serezani
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
189
|
West EE, Spolski R, Kazemian M, Yu ZX, Kemper C, Leonard WJ. A TSLP-complement axis mediates neutrophil killing of methicillin-resistant Staphylococcus aureus. Sci Immunol 2016; 1:eaaf8471. [PMID: 28783679 PMCID: PMC8530006 DOI: 10.1126/sciimmunol.aaf8471] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/03/2016] [Indexed: 09/29/2023]
Abstract
Community-acquired Staphylococcus aureus infections often present as serious skin infections in otherwise healthy individuals and have become a worldwide epidemic problem fueled by the emergence of strains with antibiotic resistance, such as methicillin-resistant S. aureus (MRSA). The cytokine thymic stromal lymphopoietin (TSLP) is highly expressed in the skin and in other barrier surfaces and plays a deleterious role by promoting T helper cell type 2 (TH2) responses during allergic diseases; however, its role in host defense against bacterial infections has not been well elucidated. We describe a previously unrecognized non-TH2 role for TSLP in enhancing neutrophil killing of MRSA during an in vivo skin infection. Specifically, we demonstrate that TSLP acts directly on both mouse and human neutrophils to augment control of MRSA. Additionally, we show that TSLP also enhances killing of Streptococcus pyogenes, another clinically important cause of human skin infections. Unexpectedly, TSLP mechanistically mediates its antibacterial effect by directly engaging the complement C5 system to modulate production of reactive oxygen species by neutrophils. Thus, TSLP increases MRSA killing in a neutrophil- and complement-dependent manner, revealing a key connection between TSLP and the innate complement system, with potentially important therapeutic implications for control of MRSA infection.
Collapse
Affiliation(s)
- Erin E West
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1674, USA.
| | - Rosanne Spolski
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1674, USA
| | - Majid Kazemian
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1674, USA
| | - Zu Xi Yu
- Pathology Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1674, USA
| | - Claudia Kemper
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1674, USA
- Division of Transplant Immunology and Mucosal Biology, King's College London, Great Maze Pond, London SE1 9RT, U.K
| | - Warren J Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1674, USA.
| |
Collapse
|
190
|
Bäsler K, Brandner JM. Tight junctions in skin inflammation. Pflugers Arch 2016; 469:3-14. [DOI: 10.1007/s00424-016-1903-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/01/2016] [Accepted: 11/07/2016] [Indexed: 12/27/2022]
|
191
|
Hatta-Kobayashi Y, Toyama-Shirai M, Yamanaka T, Takamori M, Wakabayashi Y, Naora Y, Kunieda T, Fukazawa T, Kubo T. Acute phase response in amputated tail stumps and neural tissue-preferential expression in tail bud embryos of the Xenopus neuronal pentraxin I gene. Dev Growth Differ 2016; 58:688-701. [PMID: 27804121 DOI: 10.1111/dgd.12326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 09/25/2016] [Accepted: 09/25/2016] [Indexed: 12/26/2022]
Abstract
Regeneration of lost organs involves complex processes, including host defense from infection and rebuilding of lost tissues. We previously reported that Xenopus neuronal pentraxin I (xNP1) is expressed preferentially in regenerating Xenopus laevis tadpole tails. To evaluate xNP1 function in tail regeneration, and also in tail development, we analyzed xNP1 expression in tailbud embryos and regenerating/healing tails following tail amputation in the 'regeneration' period, as well as in the 'refractory' period, when tadpoles lose their tail regenerative ability. Within 10 h after tail amputation, xNP1 was induced at the amputation site regardless of the tail regenerative ability, suggesting that xNP1 functions in acute phase responses. xNP1 was widely expressed in regenerating tails, but not in the tail buds of tailbud embryos, suggesting its possible role in the immune response/healing after an injury. xNP1 expression was also observed in neural tissues/primordia in tailbud embryos and in the spinal cord in regenerating/healing tails in both periods, implying its possible roles in neural development or function. Moreover, during the first 48 h after amputation, xNP1 expression was sustained at the spinal cord of tails in the 'regeneration' period tadpoles, but not in the 'refractory' period tadpoles, suggesting that xNP1 expression at the spinal cord correlates with regeneration. Our findings suggest that xNP1 is involved in both acute phase responses and neural development/functions, which is unique compared to mammalian pentraxins whose family members are specialized in either acute phase responses or neural functions.
Collapse
Affiliation(s)
- Yuko Hatta-Kobayashi
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Mie Toyama-Shirai
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Takehiro Yamanaka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Mayuko Takamori
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yoko Wakabayashi
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yuko Naora
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Takekazu Kunieda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Taro Fukazawa
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Takeo Kubo
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
192
|
Malhotra N, Yoon J, Leyva-Castillo JM, Galand C, Archer N, Miller LS, Geha RS. IL-22 derived from γδ T cells restricts Staphylococcus aureus infection of mechanically injured skin. J Allergy Clin Immunol 2016; 138:1098-1107.e3. [PMID: 27543072 PMCID: PMC5056816 DOI: 10.1016/j.jaci.2016.07.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 06/27/2016] [Accepted: 07/01/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Staphylococcus aureus is an opportunistic pathogen that colonizes the skin of patients with atopic dermatitis (AD) and aggravates their disease. Neutrophils and the cytokines IL-17A and IL-17F, which drive the expression of the neutrophil-attracting chemokines, are important for the clearance of S aureus infection. The cytokine IL-22 is often coproduced by IL-17-secreting cells. The levels of IL-22 are elevated in AD skin lesions. OBJECTIVE We sought to determine the role of IL-22 in the clearance of S aureus infection of mouse skin subjected to tape stripping, a surrogate for scratching, a cardinal feature of AD. METHODS S aureus was applied to the tape-stripped skin of wild-type and Il22-/- mice. Bacterial burden was evaluated by enumerating colony-forming units. Quantitative PCR and ELISA were performed to quantify Il22 mRNA and IL-22 protein in mouse and human skin. Flow cytometry was used to enumerate neutrophils in the skin. RESULTS Scratching the skin of healthy adults and tape stripping of mouse skin induced local expression of Il22 mRNA and IL-22 protein. Induction of Il22 expression by tape stripping was dependent on IL-23 and γδ T cells. Clearance of S aureus from tape-stripped skin was significantly impaired in Il22-/- mice. Neutrophil infiltration and upregulation of expression of genes encoding the antimicrobial peptides antigen-6/urokinase-type plasminogen activator receptor related protein-1 and β-DEFENSIN 14 and the chemokine (C-X-C motif) ligand following tape stripping were significantly impaired in Il22-/- mice. CONCLUSIONS These findings show that IL-22 is important for limiting the growth of S aureus on mechanically injured skin and caution that IL-23 and IL-22 blockade in patients with AD may enhance susceptibility to staphylococcal skin infection.
Collapse
Affiliation(s)
- Nidhi Malhotra
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Juhan Yoon
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Juan Manuel Leyva-Castillo
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Claire Galand
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Nathan Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Raif S Geha
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, Mass.
| |
Collapse
|
193
|
Mathew SM, Suchithra TV. Zymogram profiling of myeloperoxidase in association with increased risk of infection susceptibility in diabetic foot ulcer. Int J Diabetes Dev Ctries 2016. [DOI: 10.1007/s13410-016-0518-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
194
|
Serum neutrophil gelatinase-associated lipocalin and resistin are associated with dengue infection in adults. BMC Infect Dis 2016; 16:441. [PMID: 27549428 PMCID: PMC4994210 DOI: 10.1186/s12879-016-1759-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 08/07/2016] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Dengue is a major health problem in tropical areas, including Taiwan. Dengue virus infection affects various types of cells and results in elevation of serum inflammatory molecules. Because these molecules may be associated with dengue virus infection, the aim of this study was to identify novel molecules in febrile patients with dengue infection. In addition, we determined whether these molecules were correlated with the count of leukocytes and platelets. METHODS Febrile adults (Age >18 years old) who presented to the emergency department and were confirmed dengue virus infection were enrolled in this study. Serum from dengue patients and healthy controls was collected and serum level of sepsis-associated inflammatory molecules was measured by Luminex assay. RESULTS Elevated level of macrophage migration inhibitory factor, soluble vascular cell adhesion molecule-1, sFasL, resistin and interferon-γ were detected in patients' serum. Higher levels of neutrophil gelatinase-associated lipocalin (NGAL) and resistin were detected in dengue patients with normal leukocyte count and all dengue patients, respectively. Furthermore, the serum level of NGAL, but not resistin, was correlated with cell count in dengue patients. CONCLUSION Our results revealed that resistin and NGAL are novel dengue-associated molecules. These results may help elucidate the regulatory mechanisms of anti-dengue immune responses.
Collapse
|
195
|
Hill PB, Imai A. The immunopathogenesis of staphylococcal skin infections - A review. Comp Immunol Microbiol Infect Dis 2016; 49:8-28. [PMID: 27865269 DOI: 10.1016/j.cimid.2016.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 07/22/2016] [Accepted: 08/10/2016] [Indexed: 12/20/2022]
Abstract
Staphylococcus aureus and S. pseudintermedius are the major causes of bacterial skin disease in humans and dogs. These organisms can exist as commensals on the skin, but they can also cause severe or even devastating infections. The immune system has evolved mechanisms to deal with pathogenic microorganisms and has strategies to combat bacteria of this type. What emerges is a delicate "peace" between the opposing sides, but this balance can be disrupted leading to a full blown "war". In the ferocious battle that ensues, both sides attempt to get the upper hand, using strategies that are comparable to those used by modern day armies. In this review article, the complex interactions between the immune system and the organisms are described using such military analogies. The process is described in a sequential manner, starting with the invasion itself, and progressing to the eventual battlezone in which there are heavy casualties on both sides. By the end, the appearance of a simple pustule on the skin surface will take on a whole new meaning.
Collapse
Affiliation(s)
- P B Hill
- Companion Animal Health Centre, School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy Campus, Roseworthy SA 5371, Australia.
| | - A Imai
- Dermatology resident, Synergy Animal General Hospital, 815 Kishigami Kawaguchi, Saitama, 333-0823, Japan
| |
Collapse
|
196
|
Necroptosis Promotes Staphylococcus aureus Clearance by Inhibiting Excessive Inflammatory Signaling. Cell Rep 2016; 16:2219-2230. [PMID: 27524612 DOI: 10.1016/j.celrep.2016.07.039] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 06/09/2016] [Accepted: 07/17/2016] [Indexed: 12/24/2022] Open
Abstract
Staphylococcus aureus triggers inflammation through inflammasome activation and recruitment of neutrophils, responses that are critical for pathogen clearance but are associated with substantial tissue damage. We postulated that necroptosis, cell death mediated by the RIPK1/RIPK3/MLKL pathway, would function to limit pathological inflammation. In models of skin infection or sepsis, Mlkl-/- mice had high bacterial loads, an inability to limit interleukin-1b (IL-1b) production, and excessive inflammation. Similarly, mice treated with RIPK1 or RIPK3 inhibitors had increased bacterial loads in a model of sepsis. Ripk3-/- mice exhibited increased staphylococcal clearance and decreased inflammation in skin and systemic infection, due to direct effects of RIPK3 on IL-1b activation and apoptosis. In contrast to Casp1/4-/- mice with defective S. aureus killing, the poor outcomes of Mlkl-/- mice could not be attributed to impaired phagocytic function. We conclude that necroptotic cell death limits the pathological inflammation induced by S. aureus.
Collapse
|
197
|
Proteome-wide antigen discovery of novel protective vaccine candidates against Staphylococcus aureus infection. Vaccine 2016; 34:4602-4609. [PMID: 27496278 DOI: 10.1016/j.vaccine.2016.07.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 07/02/2016] [Accepted: 07/12/2016] [Indexed: 01/15/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a rapidly growing problem, especially in hospitals where MRSA cause increased morbidity and mortality and a significant rise in health expenditures. As many strains of MRSA are resistant to other antimicrobials in addition to methicillin, there is an urgent need to institute non-antimicrobial measures, such as vaccination, against the spread of MRSA. With the aim of finding new protective antigens for vaccine development, this study used a proteome-wide in silico antigen prediction platform to screen the proteome of S. aureus strain MRSA252. Thirty-five different S. aureus proteins were identified, recombinantly expressed, and tested for protection in a lethal sepsis mouse model using S. aureus strain MRSA252 as the challenge organism. We found that 13 of the 35 recombinant peptides yielded significant protection and that 12 of these antigens were highly conserved across 70 completely sequenced S. aureus strains. Thus, this in silico platform was capable of identifying novel candidates for inclusion in future vaccines against MRSA.
Collapse
|
198
|
Collaborative Interferon-γ and Interleukin-17 Signaling Protects the Oral Mucosa from Staphylococcus aureus. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2337-52. [PMID: 27470712 DOI: 10.1016/j.ajpath.2016.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Infections with Staphylococcus aureus are a continuing and growing problem in community and hospital settings. Preclinical animal modeling of S. aureus relies on experimental infection, which carries some limitations. We describe here a novel, spontaneous model of oral staphylococcal infection in double knockout mice, deficient in the receptors for IL-17 (IL-17RA) and interferon (IFN)-γ (IFNγRI), beginning at 6 to 8 weeks of age. IFNγRI(-/-)IL17RA(-/-) (GRAKO) mice developed progressive oral abscesses. Cytometric methods revealed extensive neutrophilic infiltration of oral tissues in GRAKO mice; further investigation evidenced that IL-17 predominated neutrophil defects in these mice. To investigate the contribution of IFN-γ signaling to this native host defense to S. aureus, we observed perturbations of monocyte recruitment and macrophage differentiation in the oral tissues of GRAKO mice, and CXCL9/chemokine ligand receptor (CXCR)3-driven recruitment of T-cell oral tissues and draining lymph nodes. To address the former finding, we depleted macrophages and monocytes in vivo from IL17RA(-/-) mice using liposomes loaded with clodronate. This treatment elicited oral abscesses, recapitulating the phenotype of GRAKO mice. From these findings, we propose novel collaborative functions of IL-17 and IFN-γ, acting through neutrophils and macrophages, respectively, in native mucocutaneous host defenses to S. aureus.
Collapse
|
199
|
Mohamed MF, Abdelkhalek A, Seleem MN. Evaluation of short synthetic antimicrobial peptides for treatment of drug-resistant and intracellular Staphylococcus aureus. Sci Rep 2016; 6:29707. [PMID: 27405275 PMCID: PMC4942614 DOI: 10.1038/srep29707] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/21/2016] [Indexed: 02/07/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections present a serious challenge because of the emergence of resistance to numerous conventional antibiotics. Due to their unique mode of action, antimicrobial peptides are novel alternatives to traditional antibiotics for tackling the issue of bacterial multidrug resistance. Herein, we investigated the antibacterial activity of two short novel peptides (WR12, a 12 residue peptide composed exclusively of arginine and tryptophan, and D-IK8, an eight residue β-sheet peptide) against multidrug resistant staphylococci. In vitro, both peptides exhibited good antibacterial activity against MRSA, vancomycin-resistant S. aureus, linezolid-resistant S. aureus, and methicillin-resistant S. epidermidis. WR12 and D-IK8 were able to eradicate persisters, MRSA in stationary growth phase, and showed significant clearance of intracellular MRSA in comparison to both vancomycin and linezolid. In vivo, topical WR12 and D-IK8 significantly reduced both the bacterial load and the levels of the pro-inflammatory cytokines including tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in MRSA-infected skin lesions. Moreover, both peptides disrupted established in vitro biofilms of S. aureus and S. epidermidis significantly more so than traditional antimicrobials tested. Taken together, these results support the potential of WR12 and D-IK8 to be used as a topical antimicrobial agent for the treatment of staphylococcal skin infections.
Collapse
Affiliation(s)
- Mohamed F. Mohamed
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Ahmed Abdelkhalek
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Mohamed N. Seleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
200
|
Type 2 Interleukin-4 Receptor Signaling in Neutrophils Antagonizes Their Expansion and Migration during Infection and Inflammation. Immunity 2016; 45:172-84. [DOI: 10.1016/j.immuni.2016.06.025] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 03/29/2016] [Accepted: 06/23/2016] [Indexed: 12/23/2022]
|