51
|
de Souza Feitosa Lima IM, Zagmignan A, Santos DM, Maia HS, Dos Santos Silva L, da Silva Cutrim B, Vieira SL, Bezerra Filho CM, de Sousa EM, Napoleão TH, Krogfelt KA, Løbner-Olesen A, Paiva PMG, Nascimento da Silva LC. Schinus terebinthifolia leaf lectin (SteLL) has anti-infective action and modulates the response of Staphylococcus aureus-infected macrophages. Sci Rep 2019; 9:18159. [PMID: 31796807 PMCID: PMC6890730 DOI: 10.1038/s41598-019-54616-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus is recognized as an important pathogen causing a wide spectrum of diseases. Here we examined the antimicrobial effects of the lectin isolated from leaves of Schinus terebinthifolia Raddi (SteLL) against S. aureus using in vitro assays and an infection model based on Galleria mellonella larvae. The actions of SteLL on mice macrophages and S. aureus-infected macrophages were also evaluated. SteLL at 16 µg/mL (8 × MIC) increased cell mass and DNA content of S. aureus in relation to untreated bacteria, suggesting that SteLL impairs cell division. Unlike ciprofloxacin, SteLL did not induce the expression of recA, crucial for DNA repair through SOS response. The antimicrobial action of SteLL was partially inhibited by 50 mM N-acetylglucosamine. SteLL reduced staphyloxathin production and increased ciprofloxacin activity towards S. aureus. This lectin also improved the survival of G. mellonella larvae infected with S. aureus. Furthermore, SteLL induced the release of cytokines (IL-6, IL-10, IL-17A, and TNF-α), nitric oxide and superoxide anion by macrophagens. The lectin improved the bactericidal action of macrophages towards S. aureus; while the expression of IL-17A and IFN-γ was downregulated in infected macrophages. These evidences suggest SteLL as important lead molecule in the development of anti-infective agents against S. aureus.
Collapse
Affiliation(s)
| | - Adrielle Zagmignan
- Programas de Pós-Graduação, Universidade Ceuma, São Luís, Maranhão, Brazil
| | | | | | | | | | | | | | | | | | - Karen Angeliki Krogfelt
- Department of Viral and Microbial Diagnostics, Statens Serum Institut, Copenhagen, Denmark
- Department of Science and Environment, Roskilde University, 4000, Roskilde, Denmark
| | - Anders Løbner-Olesen
- Department of Biology, Section for Functional Genomics, University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
52
|
Lesion size is associated with genetic polymorphisms in TLR1, TLR6, and TIRAP genes in patients with major abscesses and diabetic foot infections. Eur J Clin Microbiol Infect Dis 2019; 39:353-360. [PMID: 31786695 PMCID: PMC7010613 DOI: 10.1007/s10096-019-03732-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 10/02/2019] [Indexed: 11/03/2022]
Abstract
Genetic variation in Toll-like receptors (TLRs) has previously been associated with susceptibility to complicated skin and skin structure infections (cSSSIs). The aim of this study was to investigate associations between the severity of cSSSIs, i.e., major abscesses and diabetic foot infections (DFIs), and a set of genetic polymorphisms in the Toll-like receptor pathway. A total of 121 patients with major abscesses and 132 with DFIs participating in a randomized clinical trial were genotyped for 13 nonsynonymous single-nucleotide polymorphisms (SNPs) in genes coding for TLRs and the signaling adaptor molecule TIRAP. Infection severity was defined by lesion size at clinical presentation for both types of infections. The PEDIS infection score was also used to define severity of DFIs. Linear regression models were used to study factors independently associated with severity. In patients with large abscesses, hetero- or homozygosity for the allelic variant TLR6 (P249S) was associated with significantly smaller lesions while homozygosity for the allelic variant TLR1 (R80T) was associated with significantly larger lesions. PRRs genes were not significantly associated with PEDIS. However, patients with DFI hetero- or homozygous for the allelic variant TLR1 (S248N) had significantly larger lesions. Polymorphisms in TLR1 and TLR6 influence the severity of cSSSIs as assessed by the lesion size of major abscesses and DFIs. ClinicalTrial.gov Identifier: NCT00402727.
Collapse
|
53
|
Handschuh J, Amore J, Müller AJ. From the Cradle to the Grave of an Infection: Host-Pathogen Interaction Visualized by Intravital Microscopy. Cytometry A 2019; 97:458-470. [PMID: 31777152 DOI: 10.1002/cyto.a.23938] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/12/2019] [Accepted: 11/06/2019] [Indexed: 12/11/2022]
Abstract
During infections, interactions between host immune cells and the pathogen occur in distinct anatomical locations and along defined time scales. This can best be assessed in the physiological context of an infection in the living tissue. Consequently, intravital imaging has enabled us to dissect the critical phases and events throughout an infection in real time in living tissues. Specifically, advances in visualizing specific cell types and individual pathogens permitted tracking the early events of tissue invasion of the pathogen, cellular interactions involved in the induction of the immune response as well the events implicated in clearance of the infection. In this respect, two vantage points have evolved since the initial employment of this technique in the field of infection biology. On the one hand, strategies acquired by the pathogen to establish within the host and circumvent or evade the immune defenses have been elucidated. On the other hand, analyzing infections from the immune system's perspective has led to insights into the dynamic cellular interactions that are involved in the initial recognition of the pathogen, immune induction as well as effector function delivery and immunopathology. Furthermore, an increasing interest in probing functional parameters in vivo has emerged, such as the analysis of pathogen reactivity to stress conditions imposed by the host organism in order to mediate clearance upon pathogen encounter. Here, we give an overview on recent intravital microscopy findings of host-pathogen interactions along the course of an infection, from both the immune system's and pathogen's perspectives. We also discuss recent developments and future perspectives in extracting intravital information beyond the localization of pathogens and their interaction with immune cells. Such reporter systems on the pathogen's physiological state and immune cell functions may prove useful in dissecting the functional dynamics of host-pathogen interactions. © 2019 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Juliane Handschuh
- Institute of Molecular and Clinical Immunology, Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University, 39120, Magdeburg, Germany
| | - Jonas Amore
- Institute of Molecular and Clinical Immunology, Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University, 39120, Magdeburg, Germany
| | - Andreas J Müller
- Institute of Molecular and Clinical Immunology, Health Campus Immunology Infectiology and Inflammation (GC-I3), Otto-von-Guericke-University, 39120, Magdeburg, Germany.,Intravital Microscopy of Infection and Immunity, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| |
Collapse
|
54
|
Acker KP, Wong Fok Lung T, West E, Craft J, Narechania A, Smith H, O'Brien K, Moustafa AM, Lauren C, Planet PJ, Prince A. Strains of Staphylococcus aureus that Colonize and Infect Skin Harbor Mutations in Metabolic Genes. iScience 2019; 19:281-290. [PMID: 31401351 PMCID: PMC6700416 DOI: 10.1016/j.isci.2019.07.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/29/2022] Open
Abstract
Staphylococcus aureus is the most common cause of skin and soft tissue infections, yet the bacterial genetic changes associated with adaptation to human skin are not well characterized. S. aureus strains isolated from patients with chronic skin colonization and intermittent infection were used to determine the staphylococcal genotypes or phenotypes associated with adaptation to human skin. We demonstrate that polymorphisms in metabolic genes, particularly those involved in the tricarboxylic acid cycle, the fumarate-succinate axis, and the generation of terminal electron transporters, are unexpectedly common. These skin-adapted strains activated glycolysis and hypoxia-inducible factor-1α, interleukin (IL)-1β, and IL-18 release from keratinocytes and promoted dermatopathology equivalent to a methicillin-resistant Staphylococcus aureus USA300 control in a murine model of infection. However, in contrast to USA300, a skin-adapted isolate failed to generate protection from a secondary infectious challenge. Within the context of human skin, there appears to be selection for S. aureus metabolic adaptive changes that promote glycolysis and maintain pathogenicity.
Collapse
Affiliation(s)
- Karen P Acker
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Tania Wong Fok Lung
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Emily West
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Joshua Craft
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Apurva Narechania
- Sackler Institute for Comparative Genomics, American Museum of Natural History, New York, NY, USA
| | - Hannah Smith
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Kelsey O'Brien
- Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ahmed M Moustafa
- Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Christine Lauren
- Department of Dermatology and Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Paul J Planet
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Sackler Institute for Comparative Genomics, American Museum of Natural History, New York, NY, USA; Department of Pediatrics, Perelman College of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alice Prince
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
55
|
Pharmacological effects of ginseng on infectious diseases. Inflammopharmacology 2019; 27:871-883. [PMID: 31407196 DOI: 10.1007/s10787-019-00630-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/03/2019] [Indexed: 12/11/2022]
Abstract
Ginseng has been traditionally used as an herbal nutritional supplement in Asian countries, including Korea, China, Japan, and Vietnam for several millennia. Most studies have focused on the role of ginseng on anti-oxidative stress, anti-inflammatory, and anti-cancer activities. Recently, modulator activities of ginseng on the immune responses during pathogenic bacterial and viral infections and beneficial effects of ginseng in infectious diseases have been elucidated. In vivo and in vitro studies revealed the potential of ginseng extracts and ginsenosides Rg1, Rg3, Rb1, Rb2, Rb3, compound K, Re, Rd, Rh2 for treatment of several infectious diseases. The molecular mechanisms of these effects mainly involve inflammatory cytokines (TNF-α, IL-6, IL-1β, IFN-γ, IL-10), apoptotic pathway (bcl-2, bcl-xL), PI3K/Akt pathway, MAPKs pathway, JAK2/STAT5, NF-κB pathway, and the inflammasome. In this review, we will summarize the current knowledge on the effects of ginseng in the immune responses during the infections and its bioactivities on the prevention of infectious diseases as well as its underlying mechanisms. Moreover, the therapeutic potential of ginseng as an anti-bacterial and anti-viral medication and vaccine adjuvant will be discussed as well.
Collapse
|
56
|
Igawa S, Choi JE, Wang Z, Chang YL, Wu CC, Werbel T, Ishida-Yamamoto A, Nardo AD. Human Keratinocytes Use Sphingosine 1-Phosphate and its Receptors to Communicate Staphylococcus aureus Invasion and Activate Host Defense. J Invest Dermatol 2019; 139:1743-1752.e5. [PMID: 30807768 PMCID: PMC7682680 DOI: 10.1016/j.jid.2019.02.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/16/2019] [Accepted: 02/01/2019] [Indexed: 02/06/2023]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid mediator generated when a cell membrane or its components are damaged by various factors. S1P regulates diverse cell activities via S1P receptors (S1PRs). Keratinocytes express S1PR1-5. Although it is known that S1PRs control keratinocyte differentiation, apoptosis, and wound healing, S1PR functions in keratinocyte infections have not been fully elucidated. We propose that the S1P-S1PR axis in keratinocytes works as a biosensor for bacterial invasion. Indeed, in human impetigo infection, we found high epidermal expression of S1PR1 and S1PR2 in the skin. Furthermore, in normal human epidermal keratinocytes in vitro, treatment with Staphylococcus aureus bacterial supernatant not only induced S1P production but also increased the transcription of S1PR2, confirming our in vivo observation, as well as increased the levels of TNFA, IL36G, IL6, and IL8 mRNAs. However, direct treatment of normal human epidermal keratinocytes with S1P increased the expressions of IL36G, TNFA, and IL8, but not IL6. In both S1P- and S. aureus bacterial supernatant-treated normal human epidermal keratinocytes, S1PR1 knockdown reduced IL36G, TNFA, and IL8 transcription, and the S1PR2 antagonist JTE013 blocked the secretion of these cytokines. Overall, we have proven that during infections, keratinocytes communicate damage by using S1P release and tight control of S1PR1 and 2.
Collapse
Affiliation(s)
- Satomi Igawa
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Jae Eun Choi
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Zhenping Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Yu-Ling Chang
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Chia Chi Wu
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Tyler Werbel
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | | | - Anna Di Nardo
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA,Corresponding author: Anna Di Nardo, Department of Dermatology, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0869, La Jolla, CA 92093, Tel: 858-822-6712, Fax: 858-822-6985, , ORCiD: https://orcid.org/0000-0002-5575-9968
| |
Collapse
|
57
|
The good side of inflammation: Staphylococcus aureus proteins SpA and Sbi contribute to proper abscess formation and wound healing during skin and soft tissue infections. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2657-2670. [PMID: 31299217 DOI: 10.1016/j.bbadis.2019.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 06/28/2019] [Accepted: 07/08/2019] [Indexed: 01/18/2023]
Abstract
Staphylococcus aureus is the most prominent cause of skin and soft tissue infections (SSTI) worldwide. Mortality associated with invasive SSTI is a major threat to public health considering the incidence of antibiotic resistant isolates in particular methicillin resistant S. aureus both in the hospital (HA-MRSA) and in the community (CA-MRSA). To overcome the increasing difficulties in the clinical management of SSTI due to MRSA, new prophylactic and therapeutic approaches are urgently needed and a preventive vaccine would be welcome. The rational design of an anti-S. aureus vaccine requires a deep knowledge of the role that the different bacterial virulence factors play according to the type of infection. In the present study, using a set of isogenic deficient mutants and their complemented strains we determined that the staphylococcal surface proteins SpA and Sbi play an important role in the induction of inflammatory cytokines and chemokines in the skin during SSTI. SpA and Sbi initiate signaling cascades that lead to the early recruitment of neutrophils, modulate their lifespan in the skin milieu and contribute to proper abscess formation and bacterial eradication. Moreover, the expression of SpA and Sbi appear critical for skin repair and wound healing. Thus, these results indicate that SpA and Sbi can promote immune responses in the skin that are beneficial for the host and therefore, should not be neutralized with vaccine formulations designed to prevent SSTI.
Collapse
|
58
|
Nowicka D, Grywalska E. Staphylococcus aureus and Host Immunity in Recurrent Furunculosis. Dermatology 2019; 235:295-305. [DOI: 10.1159/000499184] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/28/2019] [Indexed: 11/19/2022] Open
Abstract
Staphylococcus aureus is one of the severest and most persistent bacterial pathogens. The most frequent S. aureus infections include impetigo, folliculitis, furuncles, furunculosis, abscesses, hidradenitis suppurativa, and mastitis. S. aureus produces a great variety of cellular and extracellular factors responsible for its invasiveness and ability to cause pathological lesions. Their expression depends on the growth phase, environmental factors, and location of the infection. Susceptibility to staphylococcal infections is rooted in multiple mechanisms of host immune responses and reactions to bacterial colonization. Immunological and inflammatory processes of chronic furunculosis are based on the pathogenicity of S. aureus as well as innate and acquired immunity. In-depth knowledge about them may help to discover the whole pathomechanism of the disease and to develop effective therapeutic options. In this review, we focus on the S. aureus-host immune interactions in the pathogenesis of recurrent furunculosis according to the most recent experimental and clinical findings.
Collapse
|
59
|
O’Brien EC, McLoughlin RM. Considering the ‘Alternatives’ for Next-Generation Anti-Staphylococcus aureus Vaccine Development. Trends Mol Med 2019; 25:171-184. [DOI: 10.1016/j.molmed.2018.12.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/20/2018] [Accepted: 12/28/2018] [Indexed: 12/14/2022]
|
60
|
Coates M, Blanchard S, MacLeod AS. Innate antimicrobial immunity in the skin: A protective barrier against bacteria, viruses, and fungi. PLoS Pathog 2018; 14:e1007353. [PMID: 30522130 PMCID: PMC6283644 DOI: 10.1371/journal.ppat.1007353] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Margaret Coates
- Department of Dermatology, Duke University, Durham, North Carolina, United States of America
| | - Sarah Blanchard
- Department of Dermatology, Duke University, Durham, North Carolina, United States of America
| | - Amanda S. MacLeod
- Department of Dermatology, Duke University, Durham, North Carolina, United States of America
- Department of Immunology, Duke University, Durham, North Carolina, United States of America
- Pinnell Center for Investigative Dermatology, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
61
|
Kim J, Bin BH, Choi EJ, Lee HG, Lee TR, Cho EG. Staphylococcus aureus-derived extracellular vesicles induce monocyte recruitment by activating human dermal microvascular endothelial cells in vitro. Clin Exp Allergy 2018; 49:68-81. [PMID: 30288827 DOI: 10.1111/cea.13289] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 09/20/2018] [Accepted: 09/28/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) represents the most common inflammatory skin disorder in children showing massive infiltration of immune cells. The colonization of AD-afflicted skin by Staphylococcus aureus and S. aureus-derived extracellular vesicles (SEVs) has been associated with AD pathogenesis; however, the molecular mechanism underlying SEV-mediated inflammatory responses remains unclear. OBJECTIVE We investigated how SEVs can mediate inflammatory responses in AD pathogenesis by examining the effect of SEVs on human dermal microvascular endothelia cells (HDMECs). METHODS HDMECs were treated with SEVs, and the expression of cell adhesion molecules or cytokines was assessed using RT-qPCR, Western blot or cytokine array analyses. The receptor for SEVs and related signalling molecules in HDMECs were addressed and verified via gene knockdown or inhibitor experiments. The recruitment assay of human THP-1 monocytic cells on HDMECs was performed after SEV treatment in the presence or absence of the verified receptor or signalling molecule. RESULTS SEVs, but not other gram-positive bacteria-derived extracellular vesicles, directly activated HDMECs by increasing the expression of cell adhesion molecules (E-selectin, VCAM1 and ICAM1) and that of IL-6, the inflammatory cytokine; consequently, they enhanced the recruitment of THP-1 monocytic cells to HDMECs. The SEV-induced HDMEC activation was dependent on Toll-like receptor 4 and the NF-κB signalling pathway, which was rapidly activated within 1 hour post-treatment and followed by an upregulation of cell adhesion molecules and IL-6 at later time-points. Moreover, SEV-mediated HDMEC responses were more rapid and intense than those induced by the same protein concentrations of S. aureus extracts. CONCLUSIONS & CLINICAL RELEVANCE SEVs as proinflammatory factors could mediate immune cell infiltration in AD by efficiently inducing endothelial cell activation and monocyte recruitment, which may provide insights into alleviating the S. aureus-mediated onset or progression of AD and its phenotypes.
Collapse
Affiliation(s)
- Jihye Kim
- Skincare Research Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Korea
| | - Bum-Ho Bin
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Korea
| | - Eun-Jeong Choi
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Korea
| | - Hyun Gee Lee
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Korea
| | - Tae Ryong Lee
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Korea
| | - Eun-Gyung Cho
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Korea
| |
Collapse
|
62
|
Scott WK, Medie FM, Ruffin F, Sharma-Kuinkel BK, Cyr DD, Guo S, Dykxhoorn DM, Skov RL, Bruun NE, Dahl A, Lerche CJ, Petersen A, Larsen AR, Lauridsen TK, Johansen HK, Ullum H, Sørensen E, Hassager C, Bundgaard H, Schønheyder HC, Torp-Pedersen C, Østergaard LB, Arpi M, Rosenvinge F, Erikstrup LT, Chehri M, Søgaard P, Andersen PS, Fowler VG. Human genetic variation in GLS2 is associated with development of complicated Staphylococcus aureus bacteremia. PLoS Genet 2018; 14:e1007667. [PMID: 30289878 PMCID: PMC6192642 DOI: 10.1371/journal.pgen.1007667] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 10/17/2018] [Accepted: 08/29/2018] [Indexed: 12/30/2022] Open
Abstract
The role of host genetic variation in the development of complicated Staphylococcus aureus bacteremia (SAB) is poorly understood. We used whole exome sequencing (WES) to examine the cumulative effect of coding variants in each gene on risk of complicated SAB in a discovery sample of 168 SAB cases (84 complicated and 84 uncomplicated, frequency matched by age, sex, and bacterial clonal complex [CC]), and then evaluated the most significantly associated genes in a replication sample of 240 SAB cases (122 complicated and 118 uncomplicated, frequency matched for age, sex, and CC) using targeted sequence capture. In the discovery sample, gene-based analysis using the SKAT-O program identified 334 genes associated with complicated SAB at p<3.5 x 10−3. These, along with eight biologically relevant candidate genes were examined in the replication sample. Gene-based analysis of the 342 genes in the replication sample using SKAT-O identified one gene, GLS2, significantly associated with complicated SAB (p = 1.2 x 10−4) after Bonferroni correction. In Firth-bias corrected logistic regression analysis of individual variants, the strongest association across all 10,931 variants in the replication sample was with rs2657878 in GLS2 (p = 5 x 10−4). This variant is strongly correlated with a missense variant (rs2657879, p = 4.4 x 10−3) in which the minor allele (associated here with complicated SAB) has been previously associated with lower plasma concentration of glutamine. In a microarray-based gene-expression analysis, individuals with SAB exhibited significantly lower expression levels of GLS2 than healthy controls. Similarly, Gls2 expression is lower in response to S. aureus exposure in mouse RAW 264.7 macrophage cells. Compared to wild-type cells, RAW 264.7 cells with Gls2 silenced by CRISPR-Cas9 genome editing have decreased IL1-β transcription and increased nitric oxide production after S. aureus exposure. GLS2 is an interesting candidate gene for complicated SAB due to its role in regulating glutamine metabolism, a key factor in leukocyte activation. Complications from bloodstream infection with Staphylococcus aureus (S. aureus) are important causes of hospitalization, significant illness, and death. The causes of these complications are not well understood, but likely involve genetic factors rendering people more susceptible to such infections, differences in the bacteria that cause the infection, and the interactions between them. We examined the parts of the human genome that code for proteins to find variations that were more common in people with complicated S. aureus bacteremia (SAB), and identified one gene, called GLS2, in which variation is more common in complicated SAB cases than uncomplicated cases. Expression of GLS2 is lower in people with SAB than controls and in mouse white blood cells exposed to S. aureus. GLS2 encodes a protein that regulates the metabolism of glutamine, a regulatory process that activates white blood cells. These cells are very important in the immune response to S. aureus infection, and therefore genetic variants that might influence their growth are important potential genetic risk factors for complicated SAB.
Collapse
Affiliation(s)
- William K. Scott
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, United States of America
- * E-mail: (WKS); (PSA); (VGF)
| | - Felix Mba Medie
- Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, NC, United States of America
| | - Felicia Ruffin
- Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, NC, United States of America
| | - Batu K. Sharma-Kuinkel
- Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, NC, United States of America
| | - Derek D. Cyr
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC, United States of America
| | - Shengru Guo
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America
| | - Derek M. Dykxhoorn
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States of America
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, United States of America
| | - Robert L. Skov
- Department of Bacteria, Parasites, and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Niels E. Bruun
- Department of Cardiology, Copenhagen University Hospital, Herlev-Gentofte, Denmark
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Anders Dahl
- Department of Cardiology, Copenhagen University Hospital, Herlev-Gentofte, Denmark
| | - Christian J. Lerche
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Andreas Petersen
- Department of Bacteria, Parasites, and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Anders Rhod Larsen
- Department of Bacteria, Parasites, and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | | | - Helle Krogh Johansen
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Henrik Ullum
- Department of Clinical Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Erik Sørensen
- Department of Clinical Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Christian Hassager
- Department of Cardiology, Rigshospitalet and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Rigshospitalet and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Henrik C. Schønheyder
- Department of Clinical Microbiology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | - Louise Bruun Østergaard
- Department of Cardiology, Copenhagen University Hospital, Herlev-Gentofte, Denmark
- Clinical Institute, Aalborg University, Aalborg, Denmark
| | - Magnus Arpi
- Department of Clinical Microbiology, Copenhagen University Hospital, Herlev-Gentofte, Denmark
| | - Flemming Rosenvinge
- Department of Clinical Microbiology, Odense University Hospital, Odense, Denmark
| | - Lise T. Erikstrup
- Department of Clinical Microbiology, Aarhus University Hospital, Aarhus, Denmark
| | - Mahtab Chehri
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Peter Søgaard
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Paal S. Andersen
- Department of Bacteria, Parasites, and Fungi, Statens Serum Institut, Copenhagen, Denmark
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail: (WKS); (PSA); (VGF)
| | - Vance G. Fowler
- Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, NC, United States of America
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC, United States of America
- * E-mail: (WKS); (PSA); (VGF)
| |
Collapse
|
63
|
Anderson ED, Sastalla I, Earland NJ, Mahnaz M, Moore IN, Otaizo-Carrasquero F, Myers TG, Myles CA, Datta SK, Myles IA. Prolonging culture of primary human keratinocytes isolated from suction blisters with the Rho kinase inhibitor Y-27632. PLoS One 2018; 13:e0198862. [PMID: 30208113 PMCID: PMC6135349 DOI: 10.1371/journal.pone.0198862] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/27/2018] [Indexed: 12/20/2022] Open
Abstract
Keratinocytes are the most abundant cell type in the epidermis. They prevent desiccation and provide immunological and barrier defense against potential pathogens such as Staphylococcus aureus and Candida albicans. The study of this first line of immune defense may be hindered by invasive isolation methods and/or improper culture conditions to support stem cell maintenance and other potential mechanisms contributing to long-term subcultivation in vitro. Primary keratinocytes have been successfully isolated from blister roofs induced by negative pressure, which separates the epidermis from the dermis in vivo in human subjects. This method allows collection of pure epidermal cells without dermal contamination in a minimally invasive manner. However, the isolated keratinocytes differentiate and senesce when cultured in vitro beyond five passages. Here, we present evidence that the Rho kinase (ROCK) inhibitor Y-27632 can be used to effectively increase the proliferative capabilities of keratinocytes isolated using the suction blister method, similar to what has been previously reported for primary keratinocytes isolated using alternative methods. We show that the increase in passage number is directly correlated to delayed differentiation, and that cells passaged long term with the inhibitor retain their ability to stratify in organotypic raft cultures and respond to cytokine treatment; additionally, the late passage cells have a heterogeneous mix of differentiated and non-differentiated cells which may be predicted by a ratio of select differentiation markers. The described method presents a minimally invasive procedure for keratinocyte isolation and prolonged culture that allows analysis of keratinocyte function in both healthy volunteers and patients with dermatologic diseases.
Collapse
Affiliation(s)
- Erik D. Anderson
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States of America
| | - Inka Sastalla
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States of America
| | - Noah J. Earland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States of America
| | - Minai Mahnaz
- Comparative Medicine Branch, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, United States of America
| | - Ian N. Moore
- Comparative Medicine Branch, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, United States of America
| | - Francisco Otaizo-Carrasquero
- Genomic Technologies Section, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States of America
| | - Timothy G. Myers
- Genomic Technologies Section, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States of America
| | | | - Sandip K. Datta
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States of America
| | - Ian A. Myles
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States of America
- * E-mail:
| |
Collapse
|
64
|
Rangel SM, Paller AS. Bacterial colonization, overgrowth, and superinfection in atopic dermatitis. Clin Dermatol 2018; 36:641-647. [DOI: 10.1016/j.clindermatol.2018.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
65
|
Munera-Campos M, Ballesca F, Richarz N, Ferrandiz C, Carrascosa JM. Paradoxical eczematous reaction to ixekizumab. J Eur Acad Dermatol Venereol 2018; 33:e40-e42. [PMID: 29953678 DOI: 10.1111/jdv.15156] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- M Munera-Campos
- Dermatology Department, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain
| | - F Ballesca
- Dermatology Department, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain
| | - N Richarz
- Dermatology Department, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain
| | - C Ferrandiz
- Dermatology Department, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain
| | - J M Carrascosa
- Dermatology Department, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain
| |
Collapse
|
66
|
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
|
67
|
MntC-Dependent Manganese Transport Is Essential for Staphylococcus aureus Oxidative Stress Resistance and Virulence. mSphere 2018; 3:3/4/e00336-18. [PMID: 30021878 PMCID: PMC6052334 DOI: 10.1128/msphere.00336-18] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Work outlined in this report demonstrated that MntC-dependent manganese transport is required for S. aureus virulence. These study results support the model that MntC-specific antibodies elicited by a vaccine have the potential to disrupt S. aureus manganese transport and thus abrogate to its virulence. Staphylococcus aureus is a human pathogen that has developed several approaches to evade the immune system, including a strategy to resist oxidative killing by phagocytes. This resistance is mediated by production of superoxide dismutase (SOD) enzymes which use manganese as a cofactor. S. aureus encodes two manganese ion transporters, MntABC and MntH, and a possible Nramp family manganese transporter, exemplified by S. aureus N315 SA1432. Their relative contributions to manganese transport have not been well defined in clinically relevant isolates. For this purpose, insertional inactivation mutations were introduced into mntC, mntH, and SA1432 individually and in combination. mntC was necessary for full resistance to methyl viologen, a compound that generates intracellular free radicals. In contrast, strains with an intact mntH gene had a minimal increase in resistance that was revealed only in mntC strains, and no change was observed upon mutation of SA1432 in strains lacking both mntC and mntH. Similarly, MntC alone was required for high cellular SOD activity. In addition, mntC strains were attenuated in a murine sepsis model. To further link these observations to manganese transport, an S. aureus MntC protein lacking manganese binding activity was designed, expressed, and purified. While circular dichroism experiments demonstrated that the secondary and tertiary structures of this protein were unaltered, a defect in manganese binding was confirmed by isothermal titration calorimetry. Unlike complementation with wild-type mntC, introduction of the manganese-binding defective allele into the chromosome of an mntC strain did not restore resistance to oxidative stress or virulence. Collectively, these results underscore the importance of MntC-dependent manganese transport in S. aureus oxidative stress resistance and virulence. IMPORTANCE Work outlined in this report demonstrated that MntC-dependent manganese transport is required for S. aureus virulence. These study results support the model that MntC-specific antibodies elicited by a vaccine have the potential to disrupt S. aureus manganese transport and thus abrogate to its virulence.
Collapse
|
68
|
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
|
69
|
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
|
70
|
Greenberg JA, Hrusch CL, Jaffery MR, David MZ, Daum RS, Hall JB, Kress JP, Sperling AI, Verhoef PA. Distinct T-helper cell responses to Staphylococcus aureus bacteremia reflect immunologic comorbidities and correlate with mortality. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:107. [PMID: 29695270 PMCID: PMC5916828 DOI: 10.1186/s13054-018-2025-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 04/03/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND The dysregulated host immune response that defines sepsis varies as a function of both the immune status of the host and the distinct nature of the pathogen. The degree to which immunocompromising comorbidities or immunosuppressive medications affect the immune response to infection is poorly understood because these patients are often excluded from studies about septic immunity. The objectives of this study were to determine the immune response to a single pathogen (Staphylococcus aureus) among a diverse case mix of patients and to determine whether comorbidities affect immune and clinical outcomes. METHODS Blood samples were drawn from 95 adult inpatients at multiple time points after the first positive S. aureus blood culture. Cox proportional hazards modeling was used to determine the associations between admission neutrophil counts, admission lymphocyte counts, cytokine levels, and 90-day mortality. A nested case-control flow cytometric analysis was conducted to determine T-helper type 1 (Th1), Th2, Th17, and regulatory T-cell (Treg) subsets among a subgroup of 28 patients. In a secondary analysis, we categorized patients as either having immunocompromising disorders (human immunodeficiency virus and hematologic malignancies), receiving immunosuppressive medications, or being not immunocompromised. RESULTS Higher neutrophil-to-lymphocyte count ratios and higher Th17 cytokine responses relative to Th1 cytokine responses early after infection were independently associated with mortality and did not depend on the immune state of the patient (HR 1.93, 95% CI 1.17-3.17, p = 0.01; and HR 1.13, 95% CI 1.01-1.27, p = 0.03, respectively). On the basis of flow cytometric analysis of CD4 T-helper subsets, an increasing Th17/Treg response over the course of the infection was most strongly associated with increased mortality (HR 4.41, 95% CI 1.69-11.5, p < 0.01). This type of immune response was most common among patients who were not immunocompromised. In contrast, among immunocompromised patients who died, a decreasing Th1/Treg response was most common. CONCLUSIONS The association of both increased Th17 responses and increased neutrophil counts relative to lymphocyte counts with mortality suggests that an overwhelming inflammatory response is detrimental. However, the differential responses of patients according to immune state suggest that immune status is an important clinical indicator that should be accounted for in the management of septic patients, as well as in the development of novel immunomodulatory therapies.
Collapse
Affiliation(s)
- Jared A Greenberg
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Rush University Medical Center, 1725 West Harrison Street, Suite 054, Chicago, IL, 60612, USA.
| | - Cara L Hrusch
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Mohammad R Jaffery
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Michael Z David
- Division of Infectious Disease, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert S Daum
- Section of Infectious Disease and Global Health, Department of Pediatrics, University of Chicago, Chicago, IL, USA
| | - Jesse B Hall
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - John P Kress
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Anne I Sperling
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA.,Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Philip A Verhoef
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
71
|
Pfalzgraff A, Brandenburg K, Weindl G. Antimicrobial Peptides and Their Therapeutic Potential for Bacterial Skin Infections and Wounds. Front Pharmacol 2018; 9:281. [PMID: 29643807 PMCID: PMC5882822 DOI: 10.3389/fphar.2018.00281] [Citation(s) in RCA: 284] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 03/13/2018] [Indexed: 01/10/2023] Open
Abstract
Alarming data about increasing resistance to conventional antibiotics are reported, while at the same time the development of new antibiotics is stagnating. Skin and soft tissue infections (SSTIs) are mainly caused by the so called ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) which belong to the most recalcitrant bacteria and are resistant to almost all common antibiotics. S. aureus and P. aeruginosa are the most frequent pathogens isolated from chronic wounds and increasing resistance to topical antibiotics has become a major issue. Therefore, new treatment options are urgently needed. In recent years, research focused on the development of synthetic antimicrobial peptides (AMPs) with lower toxicity and improved activity compared to their endogenous counterparts. AMPs appear to be promising therapeutic options for the treatment of SSTIs and wounds as they show a broad spectrum of antimicrobial activity, low resistance rates and display pivotal immunomodulatory as well as wound healing promoting activities such as induction of cell migration and proliferation and angiogenesis. In this review, we evaluate the potential of AMPs for the treatment of bacterial SSTIs and wounds and provide an overview of the mechanisms of actions of AMPs that contribute to combat skin infections and to improve wound healing. Bacteria growing in biofilms are more resistant to conventional antibiotics than their planktonic counterparts due to limited biofilm penetration and distinct metabolic and physiological functions, and often result in chronification of infections and wounds. Thus, we further discuss the feasibility of AMPs as anti-biofilm agents. Finally, we highlight perspectives for future therapies and which issues remain to bring AMPs successfully to the market.
Collapse
Affiliation(s)
- Anja Pfalzgraff
- Pharmacology and Toxicology, Department of Biology, Chemistry, Pharmacy, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | | | - Günther Weindl
- Pharmacology and Toxicology, Department of Biology, Chemistry, Pharmacy, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
72
|
Abstract
Barrier sites such as the skin play a critical role in immune defense. They must maintain homeostasis with commensals and rapidly detect and limit pathogen invasion. This is accomplished in part through the production of endogenous antimicrobial peptides and proteins, which can be either constitutive or inducible. Here, we focus particularly on the control of innate antiviral proteins and present the basic aspects of their regulation in the skin by interferons (IFNs), IFN-independent immunity, and environmental factors. We also discuss the activity and (dys-)function of antiviral proteins in the context of skin-tropic viruses and highlight the relevance of the innate antiviral pathway as a potential therapeutic avenue for vulnerable patient populations and skin diseases with high risk for virus infections.
Collapse
|
73
|
Ruzek MC, Huang L, Zhang TT, Bryant S, Slivka PF, Cuff CA, Tripp C, Blaich G. Dual Blockade of Interleukin-1 β 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. [PMID: 29311111 DOI: 10.1124/jpet.117.243493] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/12/2017] [Indexed: 03/08/2025] Open
Abstract
Despite the efficacy of biologics for treatment of rheumatoid arthritis (RA), many patients show inadequate responses and likely require neutralization of multiple mediators. Neutralization of both interleukin (IL)-1β and IL-17A with monoclonal antibodies showed greater efficacy than either agent alone in a mouse arthritis model with cooperative inhibition of key inflammatory factors, IL-6, granulocyte colony-stimulating factor (G-CSF), and CXC chemokine ligand (CXCL)1. Given the potential clinical benefit in RA, we generated a human dual variable domain antibody Ig, ABBV-615, capable of simultaneous binding and neutralization of IL-1β and IL-17A. ABBV-615 was characterized and evaluated in cynomolgus monkeys for pharmacokinetics and toxicity to enable clinical development. ABBV-615 exhibited affinities (KD) of 12 and 3 pM on human IL-1β and IL-17A, respectively, and potencies (IC50) of 3 and 58 pM, respectively, as well as excellent drug-like properties. ABBV-615 pharmacokinetics in cynomolgus monkeys was dose proportional from 20 to 100 mg/kg with a mean half-life of 16 days. However, a 13-week repeat-dose toxicity study in cynomolgus monkeys revealed time-dependent spontaneous infections exclusively in skin at all doses tested and not historically seen with single-agent anti-IL-1α/β or anti-IL-17A. Consistent with reduced resistance to skin infections, IL-1β- and IL-17A-stimulated human keratinocytes demonstrate cooperative or compensatory production of key antibacterial and inflammatory mediators such as lipocalin-2, G-CSF, CXCL1, IL-8, tumor necrosis factor, and IL-6, which aid in defense against skin bacterial infections. These results illustrate the skin-specific antimicrobial mechanisms of IL-1β and IL-17A and highlight the importance of understanding unique combinatorial effects of biologic agents.
Collapse
Affiliation(s)
- Melanie C Ruzek
- AbbVie Bioresearch Center, Worcester, Massachusetts (M.C.R., L.H., T.T.Z., S.B., P.F.S., C.A.C., C.T.); and AbbVie Deutschland, Ludwigshafen, Germany (G.B.)
| | - Lili Huang
- AbbVie Bioresearch Center, Worcester, Massachusetts (M.C.R., L.H., T.T.Z., S.B., P.F.S., C.A.C., C.T.); and AbbVie Deutschland, Ludwigshafen, Germany (G.B.)
| | - Ting-Ting Zhang
- AbbVie Bioresearch Center, Worcester, Massachusetts (M.C.R., L.H., T.T.Z., S.B., P.F.S., C.A.C., C.T.); and AbbVie Deutschland, Ludwigshafen, Germany (G.B.)
| | - Shaughn Bryant
- AbbVie Bioresearch Center, Worcester, Massachusetts (M.C.R., L.H., T.T.Z., S.B., P.F.S., C.A.C., C.T.); and AbbVie Deutschland, Ludwigshafen, Germany (G.B.)
| | - Peter F Slivka
- AbbVie Bioresearch Center, Worcester, Massachusetts (M.C.R., L.H., T.T.Z., S.B., P.F.S., C.A.C., C.T.); and AbbVie Deutschland, Ludwigshafen, Germany (G.B.)
| | - Carolyn A Cuff
- AbbVie Bioresearch Center, Worcester, Massachusetts (M.C.R., L.H., T.T.Z., S.B., P.F.S., C.A.C., C.T.); and AbbVie Deutschland, Ludwigshafen, Germany (G.B.)
| | - Catherine Tripp
- AbbVie Bioresearch Center, Worcester, Massachusetts (M.C.R., L.H., T.T.Z., S.B., P.F.S., C.A.C., C.T.); and AbbVie Deutschland, Ludwigshafen, Germany (G.B.)
| | - Guenter Blaich
- AbbVie Bioresearch Center, Worcester, Massachusetts (M.C.R., L.H., T.T.Z., S.B., P.F.S., C.A.C., C.T.); and AbbVie Deutschland, Ludwigshafen, Germany (G.B.)
| |
Collapse
|
74
|
Lee JK, Luchian T, Park Y. New antimicrobial peptide kills drug-resistant pathogens without detectable resistance. Oncotarget 2018; 9:15616-15634. [PMID: 29643997 PMCID: PMC5884652 DOI: 10.18632/oncotarget.24582] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 02/20/2018] [Indexed: 11/25/2022] Open
Abstract
Clavaspirin peptide (CSP) is derived from the pharyngeal tissues of the tunicate Styela clava. The 23-amino acid peptide is histidine-rich and amidated at the N-terminus. CSP possesses low antimicrobial and high hemolytic activity at pH 7.4. Therefore, we designed 4 CSP analogs with substituted hydrophobic amino acids to reduce hydrophobic amino acid interactions. These modifications reduced the aggregation and cytotoxicity of the analogs at pH 7.4. The analogs also showed potent antimicrobial activity by accumulating on bacterial cell surfaces and inducing the lytic mechanism against gram-negative and gram-positive cells at pH 5.5 and 7.4. Moreover, exposure to the CSP-4 analog for up to 29 passages did not induce drug resistance in Staphylococcus aureus. Application of CSP-4 to inflamed skin of hairless mice infected with drug-resistant S. aureus (DRSA) significantly reduced skin infections without damaging dermal collagen or elastin. Topically applied CSP-4 penetrated 25–40 µm in the dermis within 30 min, reducing the levels of Toll-like receptor-2, nuclear factor kappa B (NF-κB), and the pro-inflammatory cytokines tumor necrosis factor- α (TNF-α) and interleukin-1β (IL-1 β). These results suggest that CSP-4 could be a promising topical antimicrobial agent for skin diseases caused by DRSA such as S. aureus CCARM 0027.
Collapse
Affiliation(s)
- Jong-Kook Lee
- Research Center for Proteinaceous Materials, Chosun University, Gwangju, Korea.,Department of Biomedical Science, Chosun University, Gwangju, Korea
| | - Tudor Luchian
- Department of Physics, Alexandru I. Cuza University, Iasi, Romania
| | - Yoonkyung Park
- Research Center for Proteinaceous Materials, Chosun University, Gwangju, Korea.,Department of Biomedical Science, Chosun University, Gwangju, Korea
| |
Collapse
|
75
|
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
|
76
|
Ma J, Wei Y, Zhang L, Wang X, Yao D, Liu D, Liu W, Yu S, Yu Y, Wu Z, Yu L, Zhu Z, Cui Y. Identification of a novel linear B-cell epitope as a vaccine candidate in the N2N3 subdomain of Staphylococcus aureus fibronectin-binding protein A. J Med Microbiol 2018; 67:423-431. [PMID: 29458526 DOI: 10.1099/jmm.0.000633] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE To explore an epitope-based vaccine against Staphylococcus aureus, we screened the epitopes in the N2N3 subdomain of fibronectin-binding protein A (FnBPA) as a surface component of S. aureus. METHODOLOGY We expressed N2N3 proteins and prepared monoclonal antibodies (mAbs) against N2N3 by the hybridoma technique, before screening the B-cell epitopes in N2N3 using a phage-displayed random 12-mer peptide library with these mAbs against N2N3. Finally, we analysed the characters of the screened epitopes using immunofluorescence and an S. aureus infection assay. RESULTS In this paper, we identified a linear B-cell epitope in N2N3 through screening a phage-displayed peptide library with a 3C3 mAb against the N2N3. The 3C3 mAb recognized the 159IETFNKANNRFSH171 sequence of the N2N3 subdomain. Subsequently, site-directed mutagenic analysis demonstrated that residues F162, K164, N167, R168 and F169 formed the core of 159IETFNKANNRFSH171, and this core motif was the minimal determinant of the B-cell epitope recognized by the 3C3 mAb. The epitope 159IETFNKANNRFSH171 showed high homology among different S. aureus strains. Moreover, this epitope was exposed on the surface of the S. aureus by using an enzyme-linked immunosorbent assay (ELISA) assay and an indirect immunofluorescence assay. As expected, the epitope peptide evoked a protective immune response against S. aureus infection in immunized mice. CONCLUSION We identified a novel linear B-cell epitope, 159IETFNKANNRFSH171, in the N2N3 subdomain of S. aureus fibronectin-binding protein A that is recognized by 3C3 mAb, which will contribute to the further study of an epitope-based vaccine candidate against S. aureus.
Collapse
Affiliation(s)
- Jinzhu Ma
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Yuhua Wei
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Limeng Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Xintong Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Di Yao
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Daolong Liu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Wei Liu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Simiao Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Yongzhong Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Zhijun Wu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Liquan Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Zhanbo Zhu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Yudong Cui
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| |
Collapse
|
77
|
Li S, Zhang Y, Sun Y, Cao W, Cui L. Exposure to fermentation supernatant of Staphylococcus aureus accelerated dedifferentiation of chondrocytes and production of antimicrobial peptides. J Orthop Res 2018; 36:443-451. [PMID: 28513981 DOI: 10.1002/jor.23605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 05/10/2017] [Indexed: 02/04/2023]
Abstract
Staphylococcus aureus (S. aureus) is the most popular pathogen found in septic arthritis. Despite bacteria was eradicated from joint cavity during acute infection, destruction of articular cartilage often continues for years, leading to permanent joint damage. The mechanism responsible for this consistent catabolic reaction in septic arthritis remains unclear. Here, we found that fermentation supernatant (FS) of S. aureus accelerated dedifferentiation of chondrocytes and induced expression of catabolic factors including A Disintegrin-like and Metalloproteinase with Thrombospondin-1 motifs 5, NO synthase 2, matrix metalloproteinase-3, -13. In response to FS of S. aureus stimulation, expression of antimicrobial peptides (AMPs) including β-defensin-1, -2, -3, -4, cathelicidin antimicrobial peptide (CAMP) in dedifferentiated chondrocytes was significantly higher than that in chondrocytes which maintained their differentiated phenotype. Among AMPs detected, expression of CAMP in dedifferentiated chondrocytes was observed to increase 170 times higher than that in differentiated ones. When exposed to FS of S. aureus, expression of interleukin (IL)-1β, IL-17F, and IL-22 were remarkably increased in dedifferentiated chondrocytes. These results indicated that dedifferentiation of chondrocytes caused by exposure to S. aureus might be responsible for secondary osteoarthritis (OA) after acute S. aureus infection in joint. While, one potential benefit of dedifferentiation resulted from S. aureus exposure is that chondrocytes initiates a self-protective responsiveness by producing more AMPs against bacterial infection. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:443-451, 2018.
Collapse
Affiliation(s)
- Shuaijun Li
- Department of Orthopedics, Shanghai Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, 200065, P. R. China
| | - Yun Zhang
- Department of Orthopedics, Shanghai Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, 200065, P. R. China
| | - Yidan Sun
- Department of Orthopedics, Shanghai Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, 200065, P. R. China
| | - Weigang Cao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi-Zao-Ju Road, Shanghai, 200011, P. R. China
| | - Lei Cui
- Department of Orthopedics, Shanghai Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, 200065, P. R. China.,Department of Plastic Surgery, Beijing Shijitan Hospital affiliated to Beijing Capital Medical University, 10 Tieyi Road, Beijing, 100038, P. R. China
| |
Collapse
|
78
|
de la Fuente-Núñez C, Silva ON, Lu TK, Franco OL. Antimicrobial peptides: Role in human disease and potential as immunotherapies. Pharmacol Ther 2017; 178:132-140. [DOI: 10.1016/j.pharmthera.2017.04.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
79
|
Role of the microbiota in skin immunity and atopic dermatitis. Allergol Int 2017; 66:539-544. [PMID: 28882556 DOI: 10.1016/j.alit.2017.08.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 08/09/2017] [Indexed: 12/14/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease that affects 15-20% of children and 2-5% of adults in industrialized countries. The pathogen Staphylococcus aureus selectively colonizes the lesional skin of AD patients while this bacterium is absent in the skin of the majority of healthy individuals. However, the role of S. aureus in the pathogenesis of AD remains poorly understood. In addition to S. aureus, recent studies show a contribution of the skin microbiota to the regulation of immune responses in the skin as well as to the development of inflammatory skin disease. This review summarizes current knowledge about the role of the microbiota in skin immune responses and the role of S. aureus virulent factors in the pathogenesis of AD.
Collapse
|
80
|
Cruciani M, Etna MP, Camilli R, Giacomini E, Percario ZA, Severa M, Sandini S, Rizzo F, Brandi V, Balsamo G, Polticelli F, Affabris E, Pantosti A, Bagnoli F, Coccia EM. Staphylococcus aureus Esx Factors Control Human Dendritic Cell Functions Conditioning Th1/Th17 Response. Front Cell Infect Microbiol 2017; 7:330. [PMID: 28785545 PMCID: PMC5519619 DOI: 10.3389/fcimb.2017.00330] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 07/05/2017] [Indexed: 02/01/2023] Open
Abstract
The opportunistic pathogen Staphylococcus aureus (S. aureus) is a major cause of nosocomial- and community-acquired infections. In addition, many antibiotic-resistant strains are emerging worldwide, thus, there is an urgent unmet need to pinpoint novel therapeutic and prophylactic strategies. In the present study, we characterized the impact of infection with the pandemic methicillin-resistant USA300 S. aureus strain on human primary dendritic cells (DC), key initiators and regulators of immune responses. In particular, among staphylococcal virulence factors, the function of EsxA and EsxB, two small acidic dimeric proteins secreted by the type VII-like secretion system Ess (ESAT-6-like secretion system), was investigated in human DC setting. A comparative analysis of bacterial entry, replication rate as well as DC maturation, apoptosis, signaling pathway activation and cytokine production was performed by using wild type (wt) USA300 and three isogenic mutants carrying the deletion of esxA (ΔesxA), esxB (ΔesxB), or both genes (ΔesxAB). The S. aureus mutant lacking only the EsxA protein (ΔesxA) stimulated a stronger pro-apoptotic phenotype in infected DC as compared to wt USA300, ΔesxAB, and ΔesxB strains. When the mutant carrying the esxB deletion (ΔesxB) was analyzed, a higher production of both regulatory and pro-inflammatory mediators was found in the infected DC with respect to those challenged with the wt counterpart and the other esx mutants. In accordance with these data, supernatant derived from ΔesxB-infected DC promoted a stronger release of both IFN-γ and IL-17 from CD4+ T cells as compared with those conditioned with supernatants derived from wild type USA300-, ΔesxAB-, and ΔesxA-infected cultures. Although, the interaction of S. aureus with human DC is not yet fully understood, our data suggest that both cytokine production and apoptotic process are modulated by Esx factors, thus indicating a possible role of these proteins in the modulation of DC-mediated immunity to S. aureus.
Collapse
Affiliation(s)
- Melania Cruciani
- Department of Science, University Roma TreRome, Italy.,Department of Infectious Diseases, Istituto Superiore di SanitàRome, Italy
| | - Marilena P Etna
- Department of Infectious Diseases, Istituto Superiore di SanitàRome, Italy
| | - Romina Camilli
- Department of Infectious Diseases, Istituto Superiore di SanitàRome, Italy
| | - Elena Giacomini
- Department of Infectious Diseases, Istituto Superiore di SanitàRome, Italy
| | | | - Martina Severa
- Department of Infectious Diseases, Istituto Superiore di SanitàRome, Italy
| | - Silvia Sandini
- Department of Infectious Diseases, Istituto Superiore di SanitàRome, Italy
| | - Fabiana Rizzo
- Department of Infectious Diseases, Istituto Superiore di SanitàRome, Italy
| | | | | | - Fabio Polticelli
- Department of Science, University Roma TreRome, Italy.,National Institute of Nuclear Physics, Roma Tre UniversityRome, Italy
| | | | - Annalisa Pantosti
- Department of Infectious Diseases, Istituto Superiore di SanitàRome, Italy
| | | | - Eliana M Coccia
- Department of Infectious Diseases, Istituto Superiore di SanitàRome, Italy
| |
Collapse
|
81
|
Almeida PP, Pereira ÍS, Rodrigues KB, Leal LS, Marques AS, Rosa LP, da Silva FC, da Silva RAA. Photodynamic therapy controls of Staphylococcus aureus intradermal infection in mice. Lasers Med Sci 2017. [PMID: 28646389 DOI: 10.1007/s10103-017-2247-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Infections caused by Staphylococcus aureus lead to skin infections, as well as soft tissues and bone infections. Given the communal resistance to antibiotics developed by strains of this bacterium, photodynamic therapy emerges as a promising alternative treatment to control and cure infections. Females of the Balb/C mice were infected with 108 CFU of methicillin-resistant S. aureus (MRSA) and divided into four distinct groups: P-L- (negative control group), P+L- (group exposed only to curcumin), P-L+ (group exposed only to LED incidence of 450 nm, 75 mW/cm2, and 54 J/cm2 for 10 min), and P+L+ (group exposed to curcumin followed by 10 min of LED irradiation) (n = 24). The mice were euthanized 48 and 72 h after infection, and biologic materials were collected for analysis of the bacterial load, peripheral blood leukocyte counts, and draining lymph nodes cell counts. The normalization of data was checked and the ANOVA test was applied. The bacterial load in the draining lymph node of P+L+ group was lower when compared to the control groups 72 h post infection (p < 0.0001), indicating that the LED incidence associated with curcumin controls of the staphylococci intradermal infection. The number of the total lymph node cells shows to be lower than control groups in the two availed times (p < 0.01). The histological analysis and the counting of white blood cells did not show differences among cells in the blood and in the tissue of infection. This is the first report showing that photodynamic therapy may be effective against MRSA infection in a murine model of intradermal infection.
Collapse
Affiliation(s)
| | | | | | - Lorena Santos Leal
- Multidisciplinary Health Institute, UFBA, Vitória da Conquista, BA, Brazil
| | | | | | | | - Robson Amaro Augusto da Silva
- Multidisciplinary Health Institute, UFBA, Vitória da Conquista, BA, Brazil.,Multidisciplinary Health Institute, Federal University of Bahia, Rio de Contas Street, 58 Candeias, Vitoria da Conquista, BA, CEP 45029-094, Brazil
| |
Collapse
|
82
|
Neutrophil migration in infection and wound repair: going forward in reverse. Nat Rev Immunol 2017; 16:378-91. [PMID: 27231052 DOI: 10.1038/nri.2016.49] [Citation(s) in RCA: 710] [Impact Index Per Article: 88.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neutrophil migration and its role during inflammation has been the focus of increased interest in the past decade. Advances in live imaging and the use of new model systems have helped to uncover the behaviour of neutrophils in injured and infected tissues. Although neutrophils were considered to be short-lived effector cells that undergo apoptosis in damaged tissues, recent evidence suggests that neutrophil behaviour is more complex and, in some settings, neutrophils might leave sites of tissue injury and migrate back into the vasculature. The role of reverse migration and its contribution to resolution of inflammation remains unclear. In this Review, we discuss the different cues within tissues that mediate neutrophil forward and reverse migration in response to injury or infection and the implications of these mechanisms to human disease.
Collapse
|
83
|
Abstract
Inflammasomes are intracellular multiprotein complexes that comprise part of the
innate immune response. Since their definition, inflammasome disorders have been
linked to an increasing number of diseases. Autoinflammatory diseases refer to
disorders in which local factors lead to the activation of innate immune cells,
causing tissue damage when in the absence of autoantigens and autoantibodies.
Skin symptoms include the main features of monogenic inflammasomopathies, such
as Cryopyrin-Associated Periodic Syndromes (CAPS), Familial Mediterranean Fever
(FMF), Schnitzler Syndrome, Hyper-IgD Syndrome (HIDS), PAPA Syndrome, and
Deficiency of IL-1 Receptor Antagonist (DIRA). Concepts from other pathologies
have also been reviewed in recent years, such as psoriasis, after the
recognition of a combined contribution of innate and adaptive immunity in its
pathogenesis. Inflammasomes are also involved in the response to various
infections, malignancies, such as melanoma, autoimmune diseases, including
vitiligo and lupus erythematosus, atopic and contact dermatitis, acne,
hidradenitis suppurativa, among others. Inhibition of the inflammasome pathway
may be a target for future therapies, as already occurs in the handling of CAPS,
through the introduction of IL-1 inhibitors. This study presents a literature
review focusing on the participation of inflammasomes in skin diseases.
Collapse
Affiliation(s)
| | - Cyro Festa
- Universidade de São Paulo (USP) - São Paulo (SP), Brazil
| |
Collapse
|
84
|
Mishra B, Lushnikova T, Golla RM, Wang X, Wang G. Design and surface immobilization of short anti-biofilm peptides. Acta Biomater 2017; 49:316-328. [PMID: 27915018 PMCID: PMC5253077 DOI: 10.1016/j.actbio.2016.11.061] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/09/2016] [Accepted: 11/29/2016] [Indexed: 11/27/2022]
Abstract
Short antimicrobial peptides are essential to keep us healthy and their lasting potency can inspire the design of new types of antibiotics. This study reports the design of a family of eight-residue tryptophan-rich peptides (TetraF2W) obtained by converting the four phenylalanines in temporin-SHf to tryptophans. The temporin-SHf template was identified from the antimicrobial peptide database (http://aps.unmc.edu/AP). Remarkably, the double arginine variant (TetraF2W-RR) was more effective in killing methicillin-resistant Staphylococcus aureus (MRSA) USA300, but less cytotoxic to human skin HaCat and kidney HEK293 cells, than the lysine-containing dibasic combinations (KR, RK and KK). Killing kinetics and fluorescence spectroscopy suggest membrane targeting of TetraF2W-RR, making it more difficult for bacteria to develop resistance. Because established biofilms on medical devices are difficult to remove, we chose to covalently immobilize TetraF2W-RR onto the polyethylene terephthalate (PET) surface to prevent biofilm formation. The successful surface coating of the peptide is supported by FT-IR and XPS spectroscopies, chemical quantification, and antibacterial assays. This peptide-coated surface indeed prevented S. aureus biofilm formation with no cytotoxicity to human cells. In conclusion, TetraF2W-RR is a short Trp-rich peptide with demonstrated antimicrobial and anti-biofilm potency against MRSA in both the free and immobilized forms. Because these short peptides can be synthesized cost effectively, they may be developed into new antimicrobial agents or used as surface coating compounds. STATEMENT OF SIGNIFICANCE It is stunning that the total deaths due to methicillin-resistant Staphylococcus aureus (MRSA) infection are comparable to AIDS/HIV-1, making it urgent to explore new possibilities. This study deals with this problem by two strategies. First, we have designed a family of novel antimicrobial peptides with merely eight amino acids, making it cost effective for chemical synthesis. These peptides are potent against MRSA USA300. Our study uncovers that the high potency of the tryptophan-rich short peptide is coupled with arginines, whereas these Trp- and Arg-rich peptides are less toxic to select human cells than the lysine-containing analogs. Such a combination generates a more selective peptide. As a second strategy, we also demonstrate successful covalent immobilization of this short peptide to the polyethylene terephthalate (PET) surface by first using a chitosan linker, which is easy to obtain. Because biofilms on medical devices are difficult to remove by traditional antibiotics, we also show that the peptide coated surface can prevent biofilm formation. Although rarely demonstrated, we provide evidence that both the free and immobilized peptides target bacterial membranes, rendering it difficult for bacteria to develop resistance. Collectively, the significance of our study is the design of novel antimicrobial peptides provides a useful template for developing novel antimicrobials against MRSA. In addition, orientation-specific immobilization of the same short peptide can prevent biofilm formation on the PET surface, which is widely used in making prosthetic heart valves cuffs and other bio devices.
Collapse
Affiliation(s)
- Biswajit Mishra
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE 68198-6495, USA
| | - Tamara Lushnikova
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE 68198-6495, USA
| | - Radha M Golla
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE 68198-6495, USA
| | - Xiuqing Wang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE 68198-6495, USA; Department of Surgery, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Guangshun Wang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE 68198-6495, USA.
| |
Collapse
|
85
|
Interleukin-17A (IL-17A) and IL-17F Are Critical for Antimicrobial Peptide Production and Clearance of Staphylococcus aureus Nasal Colonization. Infect Immun 2016; 84:3575-3583. [PMID: 27736775 DOI: 10.1128/iai.00596-16] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/02/2016] [Indexed: 01/05/2023] Open
Abstract
Approximately 20% of the population is persistently colonized by Staphylococcus aureus in the nares. Th17-like immune responses mediated by the interleukin-17 (IL-17) family of cytokines and neutrophils are becoming recognized as relevant host defense mechanisms for resolution of S. aureus mucocutaneous infections. Since antimicrobial peptides are regulated by the IL-17 cytokines, we sought to determine the role of IL-17 cytokines in production of antimicrobial peptides in a murine model of S. aureus nasal carriage. We discovered that nasal tissue supernatants have antistaphylococcal activity, and mice deficient in both IL-17A and IL-17F lost the ability to clear S. aureus nasal colonization. IL-17A was found to be sufficient for nasal mBD-3 production ex vivo and was required for CRAMP, mBD-3, and mBD-14 expression in response to S. aureus colonization in vivo These data were confirmed in a clinical study of nasal secretions in which elevated levels of the human forms of these antimicrobial peptides were found in nasal secretions from healthy human subjects when they were colonized with S. aureus but not in secretions from noncolonized subjects. Together, these data provide evidence for the importance of IL-17A regulation of antimicrobial peptides and IL-17F in the clearance of S. aureus nasal carriage.
Collapse
|
86
|
Stentzel S, Gläser R, Bröker BM. Elucidating the anti-Staphylococcus aureusantibody response by immunoproteomics. Proteomics Clin Appl 2016; 10:1011-1019. [DOI: 10.1002/prca.201600050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 06/20/2016] [Accepted: 08/08/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Sebastian Stentzel
- Department of Immunology; University Medicine Greifswald; Greifswald Germany
| | - Regine Gläser
- Department of Dermatology; University Hospital Schleswig-Holstein; Kiel Germany
| | - Barbara M. Bröker
- Department of Immunology; University Medicine Greifswald; Greifswald Germany
| |
Collapse
|
87
|
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
|
88
|
Pfalzgraff A, Heinbockel L, Su Q, Gutsmann T, Brandenburg K, Weindl G. Synthetic antimicrobial and LPS-neutralising peptides suppress inflammatory and immune responses in skin cells and promote keratinocyte migration. Sci Rep 2016; 6:31577. [PMID: 27509895 PMCID: PMC4980674 DOI: 10.1038/srep31577] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/26/2016] [Indexed: 02/08/2023] Open
Abstract
The stagnation in the development of new antibiotics and the concomitant high increase of resistant bacteria emphasize the urgent need for new therapeutic options. Antimicrobial peptides are promising agents for the treatment of bacterial infections and recent studies indicate that Pep19-2.5, a synthetic anti-lipopolysaccharide (LPS) peptide (SALP), efficiently neutralises pathogenicity factors of Gram-negative (LPS) and Gram-positive (lipoprotein/-peptide, LP) bacteria and protects against sepsis. Here, we investigated the potential of Pep19-2.5 and the structurally related compound Pep19-4LF for their therapeutic application in bacterial skin infections. SALPs inhibited LP-induced phosphorylation of NF-κB p65 and p38 MAPK and reduced cytokine release and gene expression in primary human keratinocytes and dermal fibroblasts. In LPS-stimulated human monocyte-derived dendritic cells and Langerhans-like cells, the peptides blocked IL-6 secretion, downregulated expression of maturation markers and inhibited dendritic cell migration. Both SALPs showed a low cytotoxicity in all investigated cell types. Furthermore, SALPs markedly promoted cell migration via EGFR transactivation and ERK1/2 phosphorylation and accelerated artificial wound closure in keratinocytes. Peptide-induced keratinocyte migration was mediated by purinergic receptors and metalloproteases. In contrast, SALPs did not affect proliferation of keratinocytes. Conclusively, our data suggest a novel therapeutic target for the treatment of patients with acute and chronic skin infections.
Collapse
Affiliation(s)
- Anja Pfalzgraff
- Freie Universität Berlin, Institute of Pharmacy (Pharmacology and Toxicology), Berlin, Germany
| | - Lena Heinbockel
- Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Divisions of Biophysics, Borstel, Germany
| | - Qi Su
- Freie Universität Berlin, Institute of Pharmacy (Pharmacology and Toxicology), Berlin, Germany
| | - Thomas Gutsmann
- Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Divisions of Biophysics, Borstel, Germany
| | - Klaus Brandenburg
- Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Divisions of Biophysics, Borstel, Germany
| | - Günther Weindl
- Freie Universität Berlin, Institute of Pharmacy (Pharmacology and Toxicology), Berlin, Germany
| |
Collapse
|
89
|
Dennison SR, Morton LH, Harris F, Phoenix DA. Low pH Enhances the Action of Maximin H5 against Staphylococcus aureus and Helps Mediate Lysylated Phosphatidylglycerol-Induced Resistance. Biochemistry 2016; 55:3735-51. [PMID: 27336672 DOI: 10.1021/acs.biochem.6b00101] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Maximin H5 (MH5) is an amphibian antimicrobial peptide specifically targeting Staphylococcus aureus. At pH 6, the peptide showed an improved ability to penetrate (ΔΠ = 6.2 mN m(-1)) and lyse (lysis = 48%) Staphylococcus aureus membrane mimics, which incorporated physiological levels of lysylated phosphatidylglycerol (Lys-PG, 60%), compared to that at pH 7 (ΔΠ = 5.6 mN m(-1) and lysis = 40% at pH 7) where levels of Lys-PG are lower (40%). The peptide therefore appears to have optimal function at pH levels known to be optimal for the organism's growth. MH5 killed S. aureus (minimum inhibitory concentration of 90 μM) via membranolytic mechanisms that involved the stabilization of α-helical structure (approximately 45-50%) and showed similarities to the "Carpet" mechanism based on its ability to increase the rigidity (Cs(-1) = 109.94 mN m(-1)) and thermodynamic stability (ΔGmix = -3.0) of physiologically relevant S. aureus membrane mimics at pH 6. On the basis of theoretical analysis, this mechanism might involve the use of a tilted peptide structure, and efficacy was noted to vary inversely with the Lys-PG content of S. aureus membrane mimics for each pH studied (R(2) ∼ 0.97), which led to the suggestion that under biologically relevant conditions, low pH helps mediate Lys-PG-induced resistance in S. aureus to MH5 antibacterial action. The peptide showed a lack of hemolytic activity (<2% hemolysis) and merits further investigation as a potential template for development as an antistaphylococcal agent in medically and biotechnically relevant areas.
Collapse
Affiliation(s)
- Sarah R Dennison
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire , Preston PR1 2HE, U.K
| | - Leslie Hg Morton
- School of Forensic and Investigative Science, University of Central Lancashire , Preston PR1 2HE, U.K
| | - Frederick Harris
- School of Forensic and Investigative Science, University of Central Lancashire , Preston PR1 2HE, U.K
| | - David A Phoenix
- School of Applied Science, London South Bank University , 103 Borough Road, London SE1 0AA, U.K
| |
Collapse
|
90
|
|
91
|
Li XJ, Deng L, Brandt SL, Goodwin CB, Ma P, Yang Z, Mali RS, Liu Z, Kapur R, Serezani CH, Chan RJ. Role of p85α in neutrophil extra- and intracellular reactive oxygen species generation. Oncotarget 2016; 7:23096-105. [PMID: 27049833 PMCID: PMC5029613 DOI: 10.18632/oncotarget.8500] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/23/2016] [Indexed: 12/15/2022] Open
Abstract
Drug resistance is a growing problem that necessitates new strategies to combat pathogens. Neutrophil phagocytosis and production of intracellular ROS, in particular, has been shown to cooperate with antibiotics in the killing of microbes. This study tested the hypothesis that p85α, the regulatory subunit of PI3K, regulates production of intracellular ROS. Genetic knockout of p85α in mice caused decreased expression of catalytic subunits p110α, p110β, and p110δ, but did not change expression levels of the NADPH oxidase complex subunits p67phox, p47phox, and p40phox. When p85α, p55α, and p50α (all encoded by Pik3r1) were deleted, there was an increase in intracellular ROS with no change in phagocytosis in response to both Fcγ receptor and complement receptor stimulation. Furthermore, the increased intracellular ROS correlated with significantly improved neutrophil killing of both methicillin-susceptible and methicillin-resistant S. aureus. Our findings suggest inhibition of p85α as novel approach to improving the clearance of resistant pathogens.
Collapse
Affiliation(s)
- Xing Jun Li
- Department of Pediatrics, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indianapolis, IN, USA
| | - Lisa Deng
- Herman B Wells Center for Pediatric Research, Indianapolis, IN, USA
- Department of Medical & Molecular Genetics, Indianapolis, IN, USA
| | | | - Charles B. Goodwin
- Herman B Wells Center for Pediatric Research, Indianapolis, IN, USA
- Department of Medical & Molecular Genetics, Indianapolis, IN, USA
| | - Peilin Ma
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Zhenyun Yang
- Department of Pediatrics, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indianapolis, IN, USA
| | - Raghu S. Mali
- Department of Pediatrics, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indianapolis, IN, USA
| | - Ziyue Liu
- Department of Biostatistics, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
| | - Reuben Kapur
- Department of Pediatrics, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indianapolis, IN, USA
- Department of Medical & Molecular Genetics, Indianapolis, IN, USA
- Department of Microbiology & Immunology, Indianapolis, IN, USA
| | | | - Rebecca J. Chan
- Department of Pediatrics, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indianapolis, IN, USA
- Department of Medical & Molecular Genetics, Indianapolis, IN, USA
| |
Collapse
|
92
|
Simanski M, Rademacher F, Schröder L, Gläser R, Harder J. The Inflammasome and the Epidermal Growth Factor Receptor (EGFR) Are Involved in the Staphylococcus aureus-Mediated Induction of IL-1alpha and IL-1beta in Human Keratinocytes. PLoS One 2016; 11:e0147118. [PMID: 26808616 PMCID: PMC4726826 DOI: 10.1371/journal.pone.0147118] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 11/16/2015] [Indexed: 01/31/2023] Open
Abstract
Staphylococcus (S.) aureus is an important pathogen causing various infections including those of the skin. Keratinocytes are able to sense invading S. aureus and to initiate a fast defense reaction by the rapid release of innate defense mediators such as antimicrobial peptides and cytokines. There is increasing evidence that the cytokines IL-1alpha and IL-1beta, which both signal through the IL-1 receptor, play an important role in cutaneous defense against S. aureus. The aim of this study was to gain more insight into the underlying mechanisms leading to the S. aureus-induced IL-1alpha and IL-1beta expression in keratinocytes. Infection of human primary keratinocytes with S. aureus led to the induction of gene expression and protein secretion of IL-1alpha and IL-1beta. Full S. aureus-induced IL-1 protein release required the inflammasome components caspase-1 and ASC (apoptosis-associated speck-like protein containing a CARD) whereas gene induction of IL-1alpha and IL-beta by S. aureus was not dependent on caspase-1 and ASC. Since patients receiving anti-cancer therapy by inhibition of the epidermal growth factor receptor (EGFR) often suffer from skin infections caused by S. aureus we additionally evaluated whether the EGFR pathway may be involved in the IL-1alpha and IL-1beta induction by S. aureus. Inactivation of the EGFR with a blocking antibody decreased the S. aureus-mediated IL-1alpha and IL-1beta induction in primary keratinocytes. Moreover, the use of siRNA experiments revealed that ADAM17 (A Disintegrin and A Metalloprotease 17), a metalloproteinase known to mediate the shedding and release of EGFR ligands, was required for full induction of IL-1alpha and IL-1beta in keratinocytes infected with S. aureus. A failure of keratinocytes to adequately upregulate IL-1alpha and IL-1beta may promote S. aureus skin infections.
Collapse
Affiliation(s)
- Maren Simanski
- Department of Dermatology, University of Kiel, Kiel, Germany
| | | | - Lena Schröder
- Department of Dermatology, University of Kiel, Kiel, Germany
| | - Regine Gläser
- Department of Dermatology, University of Kiel, Kiel, Germany
| | - Jürgen Harder
- Department of Dermatology, University of Kiel, Kiel, Germany
- * E-mail:
| |
Collapse
|
93
|
Snook JD, Chesson CB, Peniche AG, Dann SM, Paulucci A, Pinchuk IV, Rudra JS. Peptide nanofiber–CaCO3 composite microparticles as adjuvant-free oral vaccine delivery vehicles. J Mater Chem B 2016; 4:1640-1649. [DOI: 10.1039/c5tb01623a] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
To combat mucosal pathogens that cause gastrointestinal (GI) infections, local mucosal immunity is required which is best achieved through oral vaccination.
Collapse
Affiliation(s)
- Joshua D. Snook
- Department of Pharmacology & Toxicology
- University of Texas Medical Branch
- Galveston
- USA
| | - Charles B. Chesson
- Institute for Translation Sciences
- University of Texas Medical Branch
- Galveston
- USA
- Sealy Center for Vaccine Development
| | - Alex G. Peniche
- Department of Internal Medicine-Division of Infectious Diseases
- University of Texas Medical Branch
- Galveston
- USA
| | - Sara M. Dann
- Institute for Translation Sciences
- University of Texas Medical Branch
- Galveston
- USA
- Department of Internal Medicine-Division of Infectious Diseases
| | | | - Iryna V. Pinchuk
- Institute for Translation Sciences
- University of Texas Medical Branch
- Galveston
- USA
- Department of Internal Medicine-Division of Gastroenterology
| | - Jai S. Rudra
- Department of Pharmacology & Toxicology
- University of Texas Medical Branch
- Galveston
- USA
- Sealy Center for Vaccine Development
| |
Collapse
|
94
|
Lee JK, Luchian T, Park Y. Effect of Regular Exercise on Inflammation Induced by Drug-resistant Staphylococcus aureus 3089 in ICR mice. Sci Rep 2015; 5:16364. [PMID: 26542343 PMCID: PMC4635399 DOI: 10.1038/srep16364] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 10/06/2015] [Indexed: 12/18/2022] Open
Abstract
Obesity is often associated with irregular dietary habits and reduced physical activity. Regular exercise induces a metabolic response that includes increased expression of various cytokines, signaling proteins and hormones, and reduced adipocyte size. In this study, mice performed a swimming exercise for 10 min/day, 5 days/week for 3 weeks. We then investigated the effect of this exercise regimen on inflammation induced by infection with drug-resistant Staphylococcus aureus strain 3089 (DRSA). In humans, DRSA causes dermatitis and pneumonitis. Similarly, DRSA induced inflammatory pneumonitis in both no-exercise (No-EX) and swim-trained (SW-EX) ICR mice. Regular exercise increased levels of the pro-inflammatory cytokines TNF-α and IL-1β and nitric oxide in both serum and whole lung tissue in SW-EX, as compared to No-EX control mice. Moreover, levels of the antimicrobial peptide cathelicidin were significantly increased in visceral adipose tissue and whole lung tissue in the SW-EX group, and this was accompanied by a reduction in the size of visceral adipocytes. In addition, levels of the inflammation marker peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) were not increased in the lung tissue of SW-EX mice. These findings suggest that in these model mice, regular exercise strengthens immune system responses, potentially preventing or mitigating infectious disease.
Collapse
Affiliation(s)
- Jong-Kook Lee
- Research Center for Proteinaceous Materials (RCPM), Chosun University, Gwangju, Korea
| | - Tudor Luchian
- Department of Physics, Alexandru I. Cuza University, Iasi, Romania
| | - Yoonkyung Park
- Department of Biotechnology &BK21-Plus Research Team for Bioactive Control Technology, Chosun University, Gwangju, Republic of Korea
| |
Collapse
|
95
|
Czarnowicki T, Malajian D, Khattri S, Correa da Rosa J, Dutt R, Finney R, Dhingra N, Xiangyu P, Xu H, Estrada YD, Zheng X, Gilleaudeau P, Sullivan-Whalen M, Suaréz-Fariñas M, Shemer A, Krueger JG, Guttman-Yassky E. Petrolatum: Barrier repair and antimicrobial responses underlying this "inert" moisturizer. J Allergy Clin Immunol 2015; 137:1091-1102.e7. [PMID: 26431582 DOI: 10.1016/j.jaci.2015.08.013] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 08/09/2015] [Accepted: 08/21/2015] [Indexed: 01/12/2023]
Abstract
BACKGROUND Petrolatum is a common moisturizer often used in the prevention of skin infections after ambulatory surgeries and as a maintenance therapy of atopic dermatitis (AD). However, the molecular responses induced by petrolatum in the skin have never been assessed. OBJECTIVE We sought to define the cutaneous molecular and structural effects induced by petrolatum. METHODS Thirty-six healthy subjects and 13 patients with moderate AD (mean SCORAD score, 39) were studied by using RT-PCR, gene arrays, immunohistochemistry, and immunofluorescence performed on control skin, petrolatum-occluded skin, and skin occluded with a Finn chamber only. RESULTS Significant upregulations of antimicrobial peptides (S100A8/fold change [FCH], 13.04; S100A9/FCH, 11.28; CCL20/FCH, 8.36; PI3 [elafin]/FCH, 15.40; lipocalin 2/FCH, 6.94, human β-defensin 2 [DEFB4A]/FCH, 4.96; P < .001 for all) and innate immune genes (IL6, IL8, and IL1B; P < .01) were observed in petrolatum-occluded skin compared with expression in both control and occluded-only skin. Application of petrolatum also induced expression of key barrier differentiation markers (filaggrin and loricrin), increased stratum corneum thickness, and significantly reduced T-cell infiltrates in the setting of "normal-appearing" or nonlesional AD skin, which is known to harbor barrier and immune defects. CONCLUSIONS Petrolatum robustly modulates antimicrobials and epidermal differentiation barrier measures. These data shed light on the beneficial molecular responses of petrolatum in barrier-defective states, such as AD and postoperative wound care.
Collapse
Affiliation(s)
- Tali Czarnowicki
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY
| | - Dana Malajian
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY; Columbia University College of Physicians and Surgeons, New York, NY
| | - Saakshi Khattri
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Joel Correa da Rosa
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY; Center for Clinical and Translational Science, The Rockefeller University, New York, NY
| | - Riana Dutt
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Robert Finney
- Department of Dermatology, Jefferson Medical College, Philadelphia, Pa
| | - Nikhil Dhingra
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Peng Xiangyu
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Hui Xu
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yeriel D Estrada
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Xiuzhong Zheng
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY
| | - Patricia Gilleaudeau
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY
| | - Mary Sullivan-Whalen
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY
| | - Mayte Suaréz-Fariñas
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomics Science, Icahn School of Medicine at Mount Sinai, New York, NY; Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Avner Shemer
- Department of Dermatology, Tel-Hashomer Hospital, Tel Aviv, Israel
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY
| | - Emma Guttman-Yassky
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
96
|
Thomason J, Rentsch C, Stenehjem EA, Hidron AI, Rimland D. Association between vitamin D deficiency and methicillin-resistant Staphylococcus aureus infection. Infection 2015; 43:715-22. [DOI: 10.1007/s15010-015-0815-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 06/20/2015] [Indexed: 01/18/2023]
|
97
|
Achouiti A, Van't Veer C, de Vos AF, van der Poll T. The receptor for advanced glycation end products promotes bacterial growth at distant body sites in Staphylococcus aureus skin infection. Microbes Infect 2015; 17:622-7. [PMID: 26086798 DOI: 10.1016/j.micinf.2015.06.002] [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: 01/09/2015] [Revised: 04/13/2015] [Accepted: 06/05/2015] [Indexed: 12/15/2022]
Abstract
The receptor for advanced glycation endproducts (RAGE) has been implicated in the regulation of skin inflammation. We here sought to study the role of RAGE in host defense during skin infection caused by Staphylococcus (S.) aureus, the most common pathogen in this condition. Wild-type (Wt) and RAGE deficient (rage(-/-)) mice were infected subcutaneously with S. aureus and bacterial loads and local inflammation were quantified at regular intervals up to 8 days after infection. While bacterial burdens were similar in both mouse strains at the primary site of infection, rage(-/-) mice had lower bacterial counts in lungs and liver. Skin cytokine and chemokine levels did not differ between groups. In accordance with the skin model, direct intravenous infection with S. aureus was associated with lower bacterial loads in lungs and liver of rage(-/-) mice. Together these data suggest that RAGE does not impact local host defense during S. aureus skin infection, but facilitates bacterial growth at distant body sites.
Collapse
Affiliation(s)
- Ahmed Achouiti
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Center for Infection and Immunity Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Cornelis Van't Veer
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Center for Infection and Immunity Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Alex F de Vos
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Center for Infection and Immunity Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Center for Infection and Immunity Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
98
|
Abstract
Skin is the most common site of Staphylococcus aureus infection. While most of these infections are self-limited, recurrent infections are common. Keratinocytes and recruited immune cells participate in skin defense against infection. We postulated that S. aureus is able to adapt to the milieu within human keratinocytes to avoid keratinocyte-mediated clearance. From a collection of S. aureus isolated from chronically infected patients with atopic dermatitis, we noted 22% had an agr mutant-like phenotype. Using several models of human skin infection, we demonstrate that toxin-deficient, agr mutants of methicillin-resistant S. aureus (MRSA) USA300 are able to persist within keratinocytes by stimulating autophagy and evading caspase-1 and inflammasome activation. MRSA infection induced keratinocyte autophagy, as evidenced by galectin-8 and LC3 accumulation. Autophagy promoted the degradation of inflammasome components and facilitated staphylococcal survival. The recovery of more than 58% agr or RNAIII mutants (P < 0.0001) of an inoculum of wild-type (WT) MRSA from within wortmannin-treated keratinocytes compared to control keratinocytes reflected the survival advantage for mutants no longer expressing agr-dependent toxins. Our results illustrate the dynamic interplay between S. aureus and keratinocytes that can result in the selection of mutants that have adapted specifically to evade keratinocyte-mediated clearance mechanisms. Human skin is a major site of staphylococcal infection, and keratinocytes actively participate in eradication of these pathogens. We demonstrate that methicillin-resistant Staphylococcus aureus (MRSA) is ingested by keratinocytes and activates caspase-1-mediated clearance through pyroptosis. Toxin-deficient MRSA mutants are selected within keratinocytes that fail to induce caspase-1 activity and keratinocyte-mediated clearance. These intracellular staphylococci induce autophagy that enhances their intracellular survival by diminishing inflammasome components. These findings suggest that S. aureus mutants, by exploiting autophagy, can persist within human keratinocytes.
Collapse
|
99
|
Kamenyeva O, Boularan C, Kabat J, Cheung GYC, Cicala C, Yeh AJ, Chan JL, Periasamy S, Otto M, Kehrl JH. Neutrophil recruitment to lymph nodes limits local humoral response to Staphylococcus aureus. PLoS Pathog 2015; 11:e1004827. [PMID: 25884622 PMCID: PMC4401519 DOI: 10.1371/journal.ppat.1004827] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 03/23/2015] [Indexed: 12/25/2022] Open
Abstract
Neutrophils form the first line of host defense against bacterial pathogens. They are rapidly mobilized to sites of infection where they help marshal host defenses and remove bacteria by phagocytosis. While splenic neutrophils promote marginal zone B cell antibody production in response to administered T cell independent antigens, whether neutrophils shape humoral immunity in other lymphoid organs is controversial. Here we investigate the neutrophil influx following the local injection of Staphylococcus aureus adjacent to the inguinal lymph node and determine neutrophil impact on the lymph node humoral response. Using intravital microscopy we show that local immunization or infection recruits neutrophils from the blood to lymph nodes in waves. The second wave occurs temporally with neutrophils mobilized from the bone marrow. Within lymph nodes neutrophils infiltrate the medulla and interfollicular areas, but avoid crossing follicle borders. In vivo neutrophils form transient and long-lived interactions with B cells and plasma cells, and their depletion augments production of antigen-specific IgG and IgM in the lymph node. In vitro activated neutrophils establish synapse- and nanotube-like interactions with B cells and reduce B cell IgM production in a TGF- β1 dependent manner. Our data reveal that neutrophils mobilized from the bone marrow in response to a local bacterial challenge dampen the early humoral response in the lymph node. Highly antibiotic resistant Staphylococcus aureus (S. aureus) are an important human pathogen and major cause of hospital acquired infections. An early host defense mechanism against bacterial infection is neutrophil recruitment, which helps eliminate the bacteria at the site of invasion. However, unless quickly neutralized, pathogens such as S. aureus can gain access to nearby lymph nodes via draining lymphatics. Lymph nodes protect the host by mobilizing additional resources that limit further pathogen dissemination. These include recruitment of neutrophils to the lymph node to directly target pathogens and the initiation of adaptive immune mechanisms, such as the humoral immune response, which transforms B lymphocytes capable of making pathogen specific antibodies into antibody producing plasma cells. Using a mouse model that allows direct visualization of lymphocytes, neutrophils, and fluorescently-labeled S. aureus in lymph nodes, we document the rapid appearance of bacteria in the lymph node following local S. aureus infection. We characterize the dynamic influx of neutrophils that occurs as a consequence and reveal direct B cell-neutrophil interactions within the lymph node parenchyma. We find that while lymph node neutrophils rapidly engage bacteria, they limit the subsequent humoral immune response likely by producing Transforming Growth Factor-β1, a factor known to limit B cell responses. These finding have important implication for our understanding of B cell responses against potent pathogens such as S. aureus and for the design of effective vaccines.
Collapse
Affiliation(s)
- Olena Kamenyeva
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Cedric Boularan
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Juraj Kabat
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gordon Y C Cheung
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Claudia Cicala
- Immunopathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anthony J Yeh
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - June L Chan
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Saravanan Periasamy
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John H Kehrl
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
100
|
Kobayashi SD, Malachowa N, DeLeo FR. Pathogenesis of Staphylococcus aureus abscesses. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1518-27. [PMID: 25749135 DOI: 10.1016/j.ajpath.2014.11.030] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 11/13/2014] [Accepted: 11/21/2014] [Indexed: 01/06/2023]
Abstract
Staphylococcus aureus causes many types of human infections and syndromes-most notably skin and soft tissue infections. Abscesses are a frequent manifestation of S. aureus skin and soft tissue infections and are formed, in part, to contain the nidus of infection. Polymorphonuclear leukocytes (neutrophils) are the primary cellular host defense against S. aureus infections and a major component of S. aureus abscesses. These host cells contain and produce many antimicrobial agents that are effective at killing bacteria, but can also cause non-specific damage to host tissues and contribute to the formation of abscesses. By comparison, S. aureus produces several molecules that also contribute to the formation of abscesses. Such molecules include those that recruit neutrophils, cause host cell lysis, and are involved in the formation of the fibrin capsule surrounding the abscess. Herein, we review our current knowledge of the mechanisms and processes underlying the formation of S. aureus abscesses, including the involvement of polymorphonuclear leukocytes, and provide a brief overview of therapeutic approaches.
Collapse
Affiliation(s)
- Scott D Kobayashi
- Laboratory of Human Bacterial Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Natalia Malachowa
- Laboratory of Human Bacterial Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Frank R DeLeo
- Laboratory of Human Bacterial Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana.
| |
Collapse
|