1
|
Zhao Y, Liu B, Zhang SY, Wang YF, Hasi SR, Qian YH, Gong ZG, Zhao JM, Yang XL, Bai YT, Cao JS, Mao W. Egg yolk-derived low-density lipoprotein: A potential drug delivery system to eradicate intracellular bacteria. Int J Biol Macromol 2025; 306:141291. [PMID: 39984077 DOI: 10.1016/j.ijbiomac.2025.141291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
Antibiotics have limited capacities to penetrate and eliminate intracellular bacteria. This study developed a drug delivery system to surmount the cell-membrane barrier and achieve efficient intracellular antibiotic accumulation for intracellular-bacterial eradication. Ceftiofur (CEF) was encapsulated in hen egg-yolk-extracted low-density lipoproteins (heLDLs) to generate CEF-heLDLs. Based on preliminary research, the drug-loading efficiency was approximately 44.48 % ± 2.35 % (encapsulation rate, approximately 99.31 % ± 0.63 %). CEF-heLDLs exhibited smaller particle sizes and higher absolute zeta potentials than heLDLs, indicating improved dispersibility and stability. In-vitro analyses demonstrated receptor-mediated uptake of CEF-heLDL, with lysosome colocalization. In-vivo localization analyses in mice showed multiorgan distribution characteristics. In-vitro and in-vivo antibacterial experiments showed that CEF-heLDLs displayed superior antibacterial effects against intracellular Staphylococcus aureus compared with free CEF, significantly damaging bacterial cell walls and decreasing intracellular-bacterial survival rates (P < 0.001). CEF-heLDLs significantly reduced mortality in methicillin-resistant S. aureus-infected mice (P < 0.001) compared with free CEF and improved bacterial-induced leukocytosis (P < 0.001). The CEF-heLDL synthesized in this study effectively delivers CEF into cells. Compared to free CEF, it has significantly enhanced efficacy in eliminating intracellular S. aureus, offering a promising novel approach for eradication of intracellular bacteria.
Collapse
Affiliation(s)
- Yi Zhao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Bo Liu
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Shuang-Yi Zhang
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Yong-Fei Wang
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Inner Mongolia Medical University, Hohhot 010030, China
| | - Su-Rong Hasi
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Ying-Hong Qian
- Inner Mongolia Academy of Agricultural & Animal Husbandry Science, Hohhot 010010, China
| | - Zhi-Guo Gong
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Jia-Min Zhao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Xiao-Lin Yang
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Yu-Ting Bai
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China
| | - Jin-Shan Cao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China.
| | - Wei Mao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot City, China.
| |
Collapse
|
2
|
Li N, Deshmukh MV, Sahin F, Hafza N, Ammanath AV, Ehnert S, Nüssler A, Weber ANR, Jin T, Götz F. Staphylococcus aureus thermonuclease NucA is a key virulence factor in septic arthritis. Commun Biol 2025; 8:598. [PMID: 40210969 PMCID: PMC11986129 DOI: 10.1038/s42003-025-07920-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 03/11/2025] [Indexed: 04/12/2025] Open
Abstract
Septic arthritis, primarily caused by Staphylococcus aureus, poses a significant risk of both mortality and morbidity due to its aggressive nature. The nuc1-encoded thermonuclease NucA of S. aureus degrades extracellular DNA/RNA, allowing the pathogen to escape neutrophil extracellular traps (NETs) and maintain the infection unabated. Here we show that in the mouse model for hematogenous septic arthritis, the Δnuc1 mutant is much less pathogenic and the severity of clinical septic arthritis is markedly reduced, including decreased weight loss, lower kidney bacterial load, reduced bone erosion, and much less IL-6 production. In vitro, S. aureus genomic DNA induces a robust TNF-α response in macrophage-like RAW 264.7 cells abrogated when the DNA is degraded by NucA. Moreover, the wild type induces high levels of TNF-α, IL-10, and IL-6 in neutrophils and osteoblast-like SAOS-2 cells, respectively. NucA exacerbates septic arthritis by increasing extracellular and intracellular survival of bacteria.
Collapse
Affiliation(s)
- Ningna Li
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Meghshree Vinod Deshmukh
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Filiz Sahin
- Siegfried Weller Institute for trauma research, BG Unfallklinik Tübingen, University of Tübingen, Tübingen, Germany
| | - Nourhane Hafza
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | | | - Sabrina Ehnert
- Siegfried Weller Institute for trauma research, BG Unfallklinik Tübingen, University of Tübingen, Tübingen, Germany
| | - Andreas Nüssler
- Siegfried Weller Institute for trauma research, BG Unfallklinik Tübingen, University of Tübingen, Tübingen, Germany
| | - Alexander N R Weber
- Interfaculty Institute for Cell Biology, Department of Immunology, Section Innate Immunity, University of Tübingen, Tübingen, Germany
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Friedrich Götz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
3
|
Bergersen KV, Zheng Y, Rossetti M, Ruffin F, Pickering H, Parmar R, Sunga G, Chan LC, Gjertson D, Fowler VG, Yeaman MR, Reed EF. Early cytokine signatures and clinical phenotypes discriminate persistent from resolving MRSA bacteremia. BMC Infect Dis 2025; 25:231. [PMID: 39966757 PMCID: PMC11834594 DOI: 10.1186/s12879-025-10620-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Staphylococcus aureus bacteremia (SAB) is a prevalent life-threatening infection often caused by methicillin-resistant S. aureus (MRSA). Up to 30% of SAB patients fail to clear infection even with gold-standard anti-MRSA antibiotics. This phenomenon is termed antibiotic-persistent MRSA bacteremia (APMB). The mechanisms driving APMB are complex and involve host phenotypes significantly impacting the immune response. Thus, defining early immune signatures and clinical phenotypes that differentiate APMB from antibiotic resolving (AR)MB could aid therapeutic success. METHODS We assessed 38 circulating cytokines and chemokines using affinity proteomics in 74 matched pairs of vancomycin-treated SAB cases identified as ARMB or APMB after 5 days of blood culture. RESULTS Unsupervised hierarchical clustering segregated APMB from ARMB based on differential levels of IL-10, IL-12p40, IL-13, CCL4, and TGFα. Additionally, CXCL1, CCL22 and IL-17A significantly differed between APMB and ARMB when correlated with diabetes, dialysis, metastatic infection, or cardiac vegetation. Combining immune signatures with these relevant clinical phenotypes sharply increased accuracy of discriminating APMB outcome to 79.1% via logistic regression modeling. Finally, classification-regression tree analysis revealed explicit analyte thresholds associated with APMB outcome at presentation especially in patients with metastatic infection. CONCLUSIONS Collectively, this study identifies previously unrecognized cytokine and chemokine signatures that distinguish APMB and ARMB at presentation and in the context of host clinical characteristics associated with increased disease severity. Validation of a biomarker signature that accurately predicts outcomes could guide early therapeutic strategies and interventions to reduce risks of persistent SAB that are associated with worsened morbidity and mortality.
Collapse
Affiliation(s)
- Kristina V Bergersen
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA
| | - Ying Zheng
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Maura Rossetti
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Felicia Ruffin
- Division of Infectious Diseases, Duke University School of Medicine, 2301 Erwin Road, Durham, NC, 27710, USA
| | - Harry Pickering
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Rajesh Parmar
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Gemalene Sunga
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA
| | - Liana C Chan
- Institute for Infection and Immunity, Lundquist Institute at Harbor UCLA Medical Center, Torrance, CA, USA
- Division of Molecular Medicine, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, USA
| | - David Gjertson
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Vance G Fowler
- Division of Infectious Diseases, Duke University School of Medicine, 2301 Erwin Road, Durham, NC, 27710, USA.
- Duke Clinical Research Institute, Duke University, Durham, NC, USA.
| | - Michael R Yeaman
- Institute for Infection and Immunity, Lundquist Institute at Harbor UCLA Medical Center, Torrance, CA, USA.
- Division of Molecular Medicine, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, USA.
- Division of Infectious Diseases, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, USA.
- Divisions of Molecular Medicine and Infectious Diseases, David Geffen School of Medicine and Harbor-UCLA Medical Center, 1124 West Carson Street, Building MRL / 250, Torrance, CA, 90502, USA.
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA.
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Hajam IA, Tsai CM, Gonzalez C, Caldera JR, Lázaro Díez M, Du X, Aralar A, Lin B, Duong W, Liu GY. Pathobiont-induced suppressive immune imprints thwart T cell vaccine responses. Nat Commun 2024; 15:10335. [PMID: 39681568 PMCID: PMC11649901 DOI: 10.1038/s41467-024-54644-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Pathobionts have evolved many strategies to coexist with the host, but how immune evasion mechanisms contribute to the difficulty of developing vaccines against pathobionts is unclear. Meanwhile, Staphylococcus aureus (SA) has resisted human vaccine development to date. Here we show that prior SA exposure induces non-protective CD4+ T cell imprints, leading to the blunting of protective IsdB vaccine responses. Mechanistically, these SA-experienced CD4+ T cells express IL-10, which is further amplified by vaccination and impedes vaccine protection by binding with IL-10Rα on CD4+ T cell and inhibit IL-17A production. IL-10 also mediates cross-suppression of IsdB and sdrE multi-antigen vaccine. By contrast, the inefficiency of SA IsdB, IsdA and MntC vaccines can be overcome by co-treatment with adjuvants that promote IL-17A and IFN-γ responses. We thus propose that IL-10 secreting, SA-experienced CD4+ T cell imprints represent a staphylococcal immune escaping mechanism that needs to be taken into consideration for future vaccine development.
Collapse
Affiliation(s)
- Irshad Ahmed Hajam
- Department of Pediatrics, University of California San Diego, San Diego, CA, 92093, USA
| | - Chih-Ming Tsai
- Department of Pediatrics, University of California San Diego, San Diego, CA, 92093, USA
| | - Cesia Gonzalez
- Department of Pediatrics, University of California San Diego, San Diego, CA, 92093, USA
| | - Juan Raphael Caldera
- Department of Pediatrics, University of California San Diego, San Diego, CA, 92093, USA
- Quest Diagnostics, 33608 Ortega Hwy., San Juan Capistrano, CA, 92675, USA
| | - María Lázaro Díez
- Department of Pediatrics, University of California San Diego, San Diego, CA, 92093, USA
- AIDS Research Institute (IrsiCaixa). VIRus Immune Escape and VACcine Design (VIRIEVAC) Universitary Hospital German Trias i Pujol Crta Canyet s/n 08916, Badalona, Barcelona, Spain
| | - Xin Du
- Department of Pediatrics, University of California San Diego, San Diego, CA, 92093, USA
| | - April Aralar
- Department of Pediatrics, University of California San Diego, San Diego, CA, 92093, USA
| | - Brian Lin
- Department of Pediatrics, University of California San Diego, San Diego, CA, 92093, USA
| | - William Duong
- Department of Pediatrics, University of California San Diego, San Diego, CA, 92093, USA
| | - George Y Liu
- Department of Pediatrics, University of California San Diego, San Diego, CA, 92093, USA.
- Division of Infectious Diseases, Rady Children's Hospital, San Diego, CA, 92123, USA.
| |
Collapse
|
5
|
Manasherob R, Warren SI, Arora P, Heo L, Haddock NL, Koliesnik I, Furukawa D, Otieno-Ayayo ZN, Maloney WJ, Lowenberg DW, Goodman SB, Amanatullah DF. The mononuclear phagocyte system obscures the accurate diagnosis of infected joint replacements. J Transl Med 2024; 22:1041. [PMID: 39563367 PMCID: PMC11575056 DOI: 10.1186/s12967-024-05866-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
INTRODUCTION Diagnosing infected joint replacements relies heavily on assessing the neutrophil response to bacteria. Bacteria form biofilms on joint replacements. Biofilms are sessile bacterial communities encased in a protective extracellular matrix, making them notoriously difficult to culture, remarkably tolerant to antibiotics, and able to evade phagocytosis. Phagocytized bacteria dramatically alter cytokine production and compromise macrophage antigen presentation. We hypothesize that a subset of joint replacements have a dormant infection that suppresses the neutrophil response to bacteria but can be distinguished from uninfected joint replacements by the response of the mononuclear phagocyte system (MPS) within periarticular tissue, synovial fluid, and circulating plasma. METHODS Single cell RNASeq transcriptomic and OLink proteomic profiling was performed on matched whole blood, synovial fluid, and periarticular tissue samples collected from 4 joint replacements with an active infection and 3 joint replacements without infection as well as 6 joint replacements with a prior infection deemed "infection-free" by the 2018 Musculoskeletal Infection Society criteria (follow-up of 26 ± 3 months). RESULTS The MPS and neutrophil responses differ by infected state; the cellular distribution of the MPS response in the subset of joints with dormant infections resembled actively infected joints (p = 0.843, Chi-square test) but was significantly different from uninfected joints (p < 0.001, Chi-square test) despite the absence of systemic acute phase reactants and recruitment of neutrophils (p < 0.001, t-test). When compared to no infection, the cellular composition of dormant infection was distinct. There was reduction in classically activated M1 macrophages (p < 0.001, Fischer's test) and alternatively activated M2 macrophages coupled with an increase in classical monocytes (p < 0.001, Fischer's test), myeloid dendritic cells (p < 0.001, Fischer's test), regulatory T-cells (p < 0.001, Fischer's test), natural killer cells (p = 0.009, Fischer's test), and plasmacytoid dendritic cells (p = 0.005, Fischer's test). Hierarchical cluster analysis and single-cell gene expression revealed that classically M1 and alternatively M2 activated macrophages as well as myeloid dendritic cells can independently distinguish the dormant and uninfected patient populations suggesting that a process that modulates neutrophil recruitment (C1QA, C1QB, LY86, SELL, CXCL5, CCL20, CD14, ITGAM), macrophage polarization (FOSB, JUN), immune checkpoint regulation (IFITM2, IFITM3, CST7, THBS1), and T-cell response (VISIG4, CD28, FYN, LAT2, FCGR3A, CD52) was occurring during dormant infection. Gene set variation analysis suggested that activation of the TNF (FDR < 0.01) and IL17 (FDR < 0.01) pathways may distinguish dormant infections from the active and uninfected populations, while an inactivation of neutrophil extracellular traps (NETs) may be involved in the lack of a clinical response to a dormant infection using established diagnostic criteria. Synovial inflammatory proteomics show an increase in synovial CXCL5 associated with dormant infection (p = 0.011, t-test), suggesting the establishment of a chronic inflammatory state by the MPS during a dormant infection involved in neutrophil inhibition. Plasma inflammatory proteomics also support a chronic inflammatory state (EGF, GZMN, FGF2, PTN, MMP12) during dormant infection that involves a reduction in neutrophil recruitment (CXCL5, p = 0.006, t-test), antigen presentation (LAMP3, p = 0.047, t-test), and T-cell function (CD28, p = 0.045, t-test; CD70, p = 0.002, t-test) that are also seen during the development of bacterial tolerance. DISCUSSION All current diagnostic criteria assume each patient can mount the same neutrophil response to an implant-associated infection. However, the state of the MPS is of critical importance to accurate diagnosis of an implant-associated infection. A reduction in neutrophil recruitment and function mediated by the MPS may allow joint replacements with a dormant infection to be mischaracterized as uninfected, thus limiting the prognostic capabilities of all current diagnostic tests.
Collapse
Affiliation(s)
- Robert Manasherob
- Department of Orthopaedic Surgery, Stanford University, 450 Broadway Street, Redwood City, CA, 94025, USA
- Department of Orthopaedic Surgery, Stanford School of Medicine, Biomedical Innovations Building, 240 Pasteur Drive, Palo Alto, CA, 94304, USA
| | - Shay I Warren
- Department of Orthopaedic Surgery, Stanford University, 450 Broadway Street, Redwood City, CA, 94025, USA
| | - Prerna Arora
- Department of Orthopaedic Surgery, Stanford University, 450 Broadway Street, Redwood City, CA, 94025, USA
| | - Lyong Heo
- Genetics and Bioinformatics Service Center (GBSC), Stanford University, 3165 Porter Drive, Palo Alto, CA, 94304, USA
| | - Naomi L Haddock
- School of Medicine, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Ievgen Koliesnik
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, 211 Quarry Road, Palo Alto, CA, 94305, USA
| | - Diasuke Furukawa
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, 211 Quarry Road, Palo Alto, CA, 94305, USA
| | - Z Ngalo Otieno-Ayayo
- School of Science, Agriculture, and Environmental Studies, Rongo University, P.O. Box 103, Rongo, 40404, Kenya
| | - William J Maloney
- Department of Orthopaedic Surgery, Stanford University, 450 Broadway Street, Redwood City, CA, 94025, USA
| | - David W Lowenberg
- Department of Orthopaedic Surgery, Stanford University, 450 Broadway Street, Redwood City, CA, 94025, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, 450 Broadway Street, Redwood City, CA, 94025, USA
- Department of Orthopaedic Surgery, Stanford School of Medicine, Biomedical Innovations Building, 240 Pasteur Drive, Palo Alto, CA, 94304, USA
| | - Derek F Amanatullah
- Department of Orthopaedic Surgery, Stanford University, 450 Broadway Street, Redwood City, CA, 94025, USA.
- Department of Orthopaedic Surgery, Stanford School of Medicine, Biomedical Innovations Building, 240 Pasteur Drive, Palo Alto, CA, 94304, USA.
| |
Collapse
|
6
|
Liu C, Wang X, Cao X. IL-10: A Key Regulator and potential therapeutic target in uveitis. Cell Immunol 2024; 405-406:104885. [PMID: 39447525 DOI: 10.1016/j.cellimm.2024.104885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
Uveitis is a prevalent inflammatory eye disease that primarily affects working-age individuals and can lead to blindness if untreated. Interleukin-10 (IL-10) is a multifunctional cytokine with broad immunosuppressive properties and plays a significant role in various pathological and physiological processes. However, its specific role and underlying mechanisms in uveitis remain incompletely understood. This review aims to shed light on the biological characteristics of IL-10, its involvement in the uveitis pathophysiology, and its potential as a novel therapeutic target. By examining existing literature, the review analyzes IL-10 expression levels and regulatory mechanisms in different types of uveitis, discussing its role in immune regulation. Despite IL-10 being expressed variably across various forms of autoimmune uveitis, studies consistently highlight its protective role, prompting research into ways to enhance its bioavailability in the eye. IL-10 is often upregulated in infectious uveitis, contributing to pathogen immune evasion. Furthermore, primary intraocular lymphoma (PIOL), which shares clinical similarities with uveitis, also shows upregulated IL-10 levels, whereas IL-6 is more commonly elevated in uveitis. This differential expression suggests that IL-6 and IL-10 could be diagnostic markers to distinguish between PIOL and uveitis. Future research should continue to focus on elucidating the molecular mechanisms of IL-10 in uveitis, exploring its potential therapeutic applications, and developing targeted treatments that leverage the immunomodulatory effects of IL-10 to prevent and manage this sight-threatening condition.
Collapse
Affiliation(s)
- Chengzhi Liu
- Institution: Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xinyu Wang
- Institution: Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xusheng Cao
- Institution: Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China.
| |
Collapse
|
7
|
Rich HE, Bhutia S, Gonzales de Los Santos F, Entrup GP, Warheit-Niemi HI, Gurczynski SJ, Bame M, Douglas MT, Morris SB, Zemans RL, Lukacs NW, Moore BB. RSV enhances Staphylococcus aureus bacterial growth in the lung. Infect Immun 2024; 92:e0030424. [PMID: 39150268 PMCID: PMC11475690 DOI: 10.1128/iai.00304-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 07/23/2024] [Indexed: 08/17/2024] Open
Abstract
Patients coinfected with respiratory syncytial virus (RSV) and bacteria have longer hospital stays, higher risk of intensive care unit admission, and worse outcomes. We describe a model of RSV line 19F/methicillin-resistant Staphylococcus aureus (MRSA) USA300 coinfection that does not impair viral clearance, but prior RSV infection enhances USA300 MRSA bacterial growth in the lung. The increased bacterial burden post-RSV correlates with reduced accumulation of neutrophils and impaired bacterial killing by alveolar macrophages. Surprisingly, reduced neutrophil accumulation is likely not explained by reductions in phagocyte-recruiting chemokines or alterations in proinflammatory cytokine production compared with mice infected with S. aureus alone. Neutrophils from RSV-infected mice retain their ability to migrate toward chemokine signals, and neutrophils from the RSV-infected lung are better able to phagocytize and kill S. aureus ex vivo on a per cell basis. In contrast, while alveolar macrophages could ingest USA300 post-RSV, intracellular bacterial killing was impaired. The RSV/S. aureus coinfected lung promotes a state of overactivation in neutrophils, demonstrated by increased production of reactive oxygen species (ROS) that can drive formation of neutrophil extracellular traps (NETs), resulting in cell death. Mice with RSV/S. aureus coinfection had increased extracellular DNA and protein in bronchoalveolar lavage fluid and histological evidence confirmed NETosis in vivo. Taken together, these data highlight that prior RSV infection can prime the overactivation of neutrophils leading to cell death that impairs neutrophil accumulation in the lung. Additionally, alveolar macrophage killing of bacteria is impaired post-RSV. Together, these defects enhance USA300 MRSA bacterial growth in the lung post-RSV.
Collapse
Affiliation(s)
- Helen E. Rich
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Simran Bhutia
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Gabrielle P. Entrup
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Helen I. Warheit-Niemi
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Stephen J. Gurczynski
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Monica Bame
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael T. Douglas
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Susan B. Morris
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Rachel L. Zemans
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicholas W. Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
8
|
Vozza EG, Kelly AM, Daly CM, O'Rourke SA, Carlile SR, Morris B, Dunne A, McLoughlin RM. Type 1 interferons promote Staphylococcus aureus nasal colonization by inducing phagocyte apoptosis. Cell Death Discov 2024; 10:403. [PMID: 39271670 PMCID: PMC11399434 DOI: 10.1038/s41420-024-02173-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/23/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Staphylococcus aureus is an important human commensal which persistently colonizes up to 30% of the human population, predominantly within the nasal cavity. The commensal lifestyle of S. aureus is complex, and the mechanisms underpinning colonization are not fully understood. S. aureus can induce an immunosuppressive environment in the nasal tissue (NT) by driving IL-10 and IL-27 to facilitate nasal colonization, indicating that S. aureus has the capacity to modulate the local immune environment for its commensal habitation. Mounting evidence suggests commensal bacteria drive type 1 interferons (IFN-I) to establish an immunosuppressive environment and whilst S. aureus can induce IFN-I during infection, its role in colonization has not yet been examined. Here, we show that S. aureus preferentially induces IFN signaling in macrophages. This IFN-I in turn upregulates expression of proapoptotic genes within macrophages culminating in caspase-3 cleavage. Importantly, S. aureus was found to drive phagocytic cell apoptosis in the nasal tissue during nasal colonization in an IFN-I dependent manner with colonization significantly reduced under caspase-3 inhibition. Overall, loss of IFN-I signaling significantly diminished S. aureus nasal colonization implicating a pivotal role for IFN-I in controlling S. aureus persistence during colonization through its ability to induce phagocyte apoptosis. Together, this study reveals a novel strategy utilized by S. aureus to circumvent host immunity in the nasal mucosa to facilitate nasal colonization.
Collapse
Affiliation(s)
- Emilio G Vozza
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Alanna M Kelly
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Clíodhna M Daly
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Sinead A O'Rourke
- Molecular Immunology Group, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Simon R Carlile
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Brenda Morris
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Aisling Dunne
- Molecular Immunology Group, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Rachel M McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
9
|
Guzylack-Piriou L, Gausseres B, Tasca C, Hassel C, Tabouret G, Foucras G. A loss of function mutation in SOCS2 results in increased inflammatory response of macrophages to TLR ligands and Staphylococcus aureus. Front Immunol 2024; 15:1397330. [PMID: 39185412 PMCID: PMC11341364 DOI: 10.3389/fimmu.2024.1397330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/19/2024] [Indexed: 08/27/2024] Open
Abstract
Introduction The role of suppressor of cytokine signaling (SOCS)2 in anti-infective bacterial immunity has been poorly investigated compared to other members of the SOCS family. Methods We characterized the previously identified loss of function R96C point mutation of SOCS2 using a genome-edited mouse model that resumes the phenotype of Socs2 knockout mice. The response of macrophages to TLR-ligands and Staphylococcus aureus was examined. Results and discussion Conversely to previously published data using human monocyte-derived macrophages, the stimulation of bone-marrow-derived macrophages with various TLR ligands did not show any difference according to the SOCS2 variant. Upregulation of IL-6 and TNF-α pro-inflammatory cytokines production was only seen when the SOCS2 expression was promoted by the culture of macrophages in the presence of GM-CSF. Furthermore, we showed that the SOCS2 point mutation is associated with heightened STAT5 phosphorylation in a short time frame upon GM-CSF incubation. In mice, recruitment of neutrophil and F4/80int Ly6C+ inflammatory macrophage, as well as IFN-γ and IL-10 concentrations, are significantly increased upon S. aureus peritoneal infection. Altogether, these data support the idea that by lowering the pro-inflammatory environment, SOCS2 favors better control of bacterial burden during a systemic infection caused by S. aureus.
Collapse
|
10
|
Nagarajan A, Scoggin K, Adams LG, Threadgill D, Andrews-Polymenis H. Identification of a genetic region linked to tolerance to MRSA infection using Collaborative Cross mice. PLoS Genet 2024; 20:e1011378. [PMID: 39178306 PMCID: PMC11407622 DOI: 10.1371/journal.pgen.1011378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 09/17/2024] [Accepted: 07/29/2024] [Indexed: 08/25/2024] Open
Abstract
Staphylococcus aureus (S. aureus) colonizes humans asymptomatically but can also cause opportunistic infections, ranging from mild skin infections to severe life-threatening conditions. Resistance and tolerance are two ways a host can survive an infection. Resistance is limiting the pathogen burden, while tolerance is limiting the health impact of a given pathogen burden. In previous work, we established that collaborative cross (CC) mouse line CC061 is highly susceptible to Methicillin-resistant S. aureus infection (MRSA, USA300), while CC024 is tolerant. To identify host genes involved in tolerance after S. aureus infection, we crossed CC061 mice and CC024 mice to generate F1 and F2 populations. Survival after MRSA infection in the F1 and F2 generations was 65% and 55% and followed a complex dominant inheritance pattern for the CC024 increased survival phenotype. Colonization in F2 animals was more extreme than in their parents, suggesting successful segregation of genetic factors. We identified a Quantitative Trait Locus (QTL) peak on chromosome 7 for survival and weight change after infection. In this QTL, the WSB/EiJ (WSB) allele was present in CC024 mice and contributed to their MRSA tolerant phenotype. Two genes, C5ar1 and C5ar2, have high-impact variants in this region. C5ar1 and C5ar2 are receptors for the complement factor C5a, an anaphylatoxin that can trigger a massive immune response by binding to these receptors. We hypothesize that C5a may have altered binding to variant receptors in CC024 mice, reducing damage caused by the cytokine storm and resulting in the ability to tolerate a higher pathogen burden and longer survival.
Collapse
Affiliation(s)
- Aravindh Nagarajan
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| | - Kristin Scoggin
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| | - L Garry Adams
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - David Threadgill
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas, United States of America
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, Texas, United States of America
- Department of Biochemistry & Biophysics and Department of Nutrition, Texas A&M University, College Station, Texas, United States of America
| | - Helene Andrews-Polymenis
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
11
|
Kelly AM, McCarthy KN, Claxton TJ, Carlile SR, O'Brien EC, Vozza EG, Mills KH, McLoughlin RM. IL-10 inhibition during immunization improves vaccine-induced protection against Staphylococcus aureus infection. JCI Insight 2024; 9:e178216. [PMID: 38973612 PMCID: PMC11383370 DOI: 10.1172/jci.insight.178216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/22/2024] [Indexed: 07/09/2024] Open
Abstract
Staphylococcus aureus is a major human pathogen. An effective anti-S. aureus vaccine remains elusive as the correlates of protection are ill-defined. Targeting specific T cell populations is an important strategy for improving anti-S. aureus vaccine efficacy. Potential bottlenecks that remain are S. aureus-induced immunosuppression and the impact this might have on vaccine-induced immunity. S. aureus induces IL-10, which impedes effector T cell responses, facilitating persistence during both colonization and infection. Thus, it was hypothesized that transient targeting of IL-10 might represent an innovative way to improve vaccine efficacy. In this study, IL-10 expression was elevated in the nares of persistent carriers of S. aureus, and this was associated with reduced systemic S. aureus-specific Th1 responses. This suggests that systemic responses are remodeled because of commensal exposure to S. aureus, which negatively implicates vaccine function. To provide proof of concept that targeting immunosuppressive responses during immunization may be a useful approach to improve vaccine efficacy, we immunized mice with T cell-activating vaccines in combination with IL-10-neutralizing antibodies. Blocking IL-10 during vaccination enhanced effector T cell responses and improved bacterial clearance during subsequent systemic and subcutaneous infection. Taken together, these results reveal a potentially novel strategy for improving anti-S. aureus vaccine efficacy.
Collapse
Affiliation(s)
| | - Karen N McCarthy
- Host-Pathogen Interactions Group and
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | | - Kingston Hg Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
12
|
Parmar R, Pickering H, Ahn R, Rossetti M, Gjertson DW, Ruffin F, Chan LC, Fowler VG, Yeaman MR, Reed EF. Integrated transcriptomic analysis reveals immune signatures distinguishing persistent versus resolving outcomes in MRSA bacteremia. Front Immunol 2024; 15:1373553. [PMID: 38846955 PMCID: PMC11153731 DOI: 10.3389/fimmu.2024.1373553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/02/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Staphylococcus aureus bacteremia (SAB) is a life-threatening infection particularly involving methicillin-resistant S. aureus (MRSA). In contrast to resolving MRSA bacteremia (RB), persistent MRSA bacteremia (PB) blood cultures remain positive despite appropriate antibiotic treatment. Host immune responses distinguishing PB vs. RB outcomes are poorly understood. Here, integrated transcriptomic, IL-10 cytokine levels, and genomic analyses sought to identify signatures differentiating PB vs. RB outcomes. Methods Whole-blood transcriptomes of propensity-matched PB (n=28) versus RB (n=30) patients treated with vancomycin were compared in one independent training patient cohort. Gene expression (GE) modules were analyzed and prioritized relative to host IL-10 cytokine levels and DNA methyltransferase-3A (DNMT3A) genotype. Results Differential expression of T and B lymphocyte gene expression early in MRSA bacteremia discriminated RB from PB outcomes. Significant increases in effector T and B cell signaling pathways correlated with RB, lower IL-10 cytokine levels and DNMT3A heterozygous A/C genotype. Importantly, a second PB and RB patient cohort analyzed in a masked manner demonstrated high predictive accuracy of differential signatures. Discussion Collectively, the present findings indicate that human PB involves dysregulated immunity characterized by impaired T and B cell responses associated with excessive IL-10 expression in context of the DNMT3A A/A genotype. These findings reveal distinct immunologic programs in PB vs. RB outcomes, enable future studies to define mechanisms by which host and/or pathogen drive differential signatures and may accelerate prediction of PB outcomes. Such prognostic assessment of host risk could significantly enhance early anti-infective interventions to avert PB and improve patient outcomes.
Collapse
Affiliation(s)
- Rajesh Parmar
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Harry Pickering
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Richard Ahn
- Department of Microbiology, Immunology, & Molecular Genetics, University of California Los Angeles, Los Angeles, CA, United States
| | - Maura Rossetti
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - David W. Gjertson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Felicia Ruffin
- Division of Infectious Diseases, Duke University, Durham, NC, United States
| | - Liana C. Chan
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
- Divisions of Molecular Medicine and Infectious Diseases, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, United States
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Vance G. Fowler
- Division of Infectious Diseases, Duke University, Durham, NC, United States
| | - Michael R. Yeaman
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
- Divisions of Molecular Medicine and Infectious Diseases, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, United States
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
13
|
Harris JR, Zoccoli-Rodriguez V, Delaney MS, Cruz TN, Gaudette BT, Wilmore JR. Gut commensals require Peyer's patches to induce protective systemic IgA responses. RESEARCH SQUARE 2024:rs.3.rs-4220532. [PMID: 38798510 PMCID: PMC11118714 DOI: 10.21203/rs.3.rs-4220532/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Gut educated IgA secreting plasma cells that disseminate beyond the mucosa and into systemic tissues have been described as providing beneficial effects from disease in several contexts. Several bacteria have been implicated in the induction of systemic IgA, however the mechanisms that result in differential levels of induction by each bacterial species are still unknown. Here we show, the commensal bacteria, Bacteroides fragilis (Bf), is an efficient inducer of systemic IgA responses. The ability of Bf to induce the production of bone marrow IgA plasma cells and high levels of serum IgA relied on high levels of gut colonization in a dose-dependent manner. Colonization induced Bf-specific IgA responses were severely diminished in the absence of Peyer's patches, but not the murine cecal patch. Colonization of mice with Bf, a natural human commensal, resulted in few changes within the microbiome and the host transcriptional profile in the gut, suggesting a commensal relationship with the host. Bf colonization did benefit the mice by inducing systemic IgA that led to increased protection in a bowel perforation model resulting in lower peritoneal abscess formation. These findings demonstrate a critical role for bacterial colonization and Peyer's patches in the induction of robust systemic IgA responses that confer protection from bacterial dissemination outside of the gut.
Collapse
Affiliation(s)
- Joshua R. Harris
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY
| | | | - Mara S. Delaney
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY
| | - Tania N. Cruz
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY
| | - Brian T. Gaudette
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Joel R. Wilmore
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY
- Sepsis Interdisciplinary Research Center, SUNY Upstate Medical University, Syracuse, NY
| |
Collapse
|
14
|
Hajam IA, Liu GY. Linking S. aureus Immune Evasion Mechanisms to Staphylococcal Vaccine Failures. Antibiotics (Basel) 2024; 13:410. [PMID: 38786139 PMCID: PMC11117348 DOI: 10.3390/antibiotics13050410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
Vaccination arguably remains the only long-term strategy to limit the spread of S. aureus infections and its related antibiotic resistance. To date, however, all staphylococcal vaccines tested in clinical trials have failed. In this review, we propose that the failure of S. aureus vaccines is intricately linked to prior host exposure to S. aureus and the pathogen's capacity to evade adaptive immune defenses. We suggest that non-protective immune imprints created by previous exposure to S. aureus are preferentially recalled by SA vaccines, and IL-10 induced by S. aureus plays a unique role in shaping these non-protective anti-staphylococcal immune responses. We discuss how S. aureus modifies the host immune landscape, which thereby necessitates alternative approaches to develop successful staphylococcal vaccines.
Collapse
Affiliation(s)
- Irshad Ahmed Hajam
- Department of Pediatrics, University of California San Diego, San Diego, CA 92093, USA;
| | - George Y. Liu
- Department of Pediatrics, University of California San Diego, San Diego, CA 92093, USA;
- Division of Infectious Diseases, Rady Children’s Hospital, San Diego, CA 92123, USA
| |
Collapse
|
15
|
Taha SM, Abd El-Aziz NK, Abdelkhalek A, Pet I, Ahmadi M, El-Nabtity SM. Chitosan-Loaded Lagenaria siceraria and Thymus vulgaris Potentiate Antibacterial, Antioxidant, and Immunomodulatory Activities against Extensive Drug-Resistant Pseudomonas aeruginosa and Vancomycin-Resistant Staphylococcus aureus: In Vitro and In Vivo Approaches. Antioxidants (Basel) 2024; 13:428. [PMID: 38671876 PMCID: PMC11047512 DOI: 10.3390/antiox13040428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/17/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Antimicrobial resistance poses considerable issues for current clinical care, so the modified use of antimicrobial agents and public health initiatives, coupled with new antimicrobial approaches, may help to minimize the impact of multidrug-resistant (MDR) bacteria in the future. This study aimed to evaluate the antimicrobial, antioxidant, and immunomodulatory activities of Lagenaria siceraria, Thymus vulgaris, and their chitosan nanocomposites against extensive drug-resistant (XDR) Pseudomonas aeruginosa and vancomycin-resistant Staphylococcus aureus (VRSA) using both in vitro and in vivo assays. The in vitro antimicrobial susceptibilities of P. aeruginosa and VRSA strains revealed 100% sensitivity to imipenem (100%). All P. aeruginosa strains were resistant to cefoxitin, cefepime, trimethoprim + sulfamethoxazole, and fosfomycin. However, S. aureus strains showed a full resistance to cefoxitin, amoxicillin, ampicillin, erythromycin, chloramphenicol, and fosfomycin (100% each). Interestingly, all S. aureus strains were vancomycin-resistant (MIC = 32-512 μg/mL), and 90% of P. aeruginosa and S. aureus strains were XDR. The antimicrobial potential of Lagenaria siceraria and Thymus vulgaris nanocomposites with chitosan nanoparticles demonstrated marked inhibitory activities against XDR P. aeruginosa and VRSA strains with inhibition zones' diameters up to 50 mm and MIC values ranging from 0.125 to 1 μg/mL and 1 to 8 μg/mL, respectively. The results of the in vivo approach in male Sprague Dawley rats revealed that infection with P. aeruginosa and S. aureus displayed significant changes in biochemical, hematological, and histopathological findings compared to the negative control group. These values returned to the normal range after treatment by chitosan nanoparticles, either loaded with Lagenaria siceraria or Thymus vulgaris. Real-time quantitative polymerase chain reaction (RT-qPCR) findings presented significant upregulation of the relative expression of the IL10 gene and downregulation of the IFNG gene throughout the experimental period, especially after treatment with chitosan nanoparticles loaded either with Lagenaria siceraria or Thymus vulgaris in comparison to the positive control groups. In conclusion, this is the first report suggesting the use of Lagenaria siceraria and Thymus vulgaris nanocomposites with chitosan nanoparticles as a promising contender for combating XDR P. aeruginosa and VRSA infections as well as a manager for inflammatory situations and oxidative stress-related disorders.
Collapse
Affiliation(s)
- Selwan M Taha
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Norhan K Abd El-Aziz
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Adel Abdelkhalek
- Food Safety, Hygiene and Technology Department, Faculty of Veterinary Medicine, Badr University in Cairo (BUC), Badr City 11829, Egypt
| | - Ioan Pet
- Department of Biotechnology, Faculty of Bioengineering of Animals Resources, University of Life Sciences "King Mihai I" from Timisoara, 300645 Timisoara, Romania
| | - Mirela Ahmadi
- Department of Biotechnology, Faculty of Bioengineering of Animals Resources, University of Life Sciences "King Mihai I" from Timisoara, 300645 Timisoara, Romania
| | - Sameh M El-Nabtity
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| |
Collapse
|
16
|
Bertrand BP, Shinde D, Thomas VC, Whiteley M, Ibberson CB, Kielian T. Metabolic diversity of human macrophages: potential influence on Staphylococcus aureus intracellular survival. Infect Immun 2024; 92:e0047423. [PMID: 38179975 PMCID: PMC10863412 DOI: 10.1128/iai.00474-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/04/2023] [Indexed: 01/06/2024] Open
Abstract
Staphylococcus aureus is a leading cause of medical device-associated biofilm infections. This is influenced by the ability of S. aureus biofilm to evade the host immune response, which is partially driven by the anti-inflammatory cytokine interleukin-10 (IL-10). Here, we show that treatment of human monocyte-derived macrophages (HMDMs) with IL-10 enhanced biofilm formation, suggesting that macrophage anti-inflammatory programming likely plays an important role during the transition from planktonic to biofilm growth. To identify S. aureus genes that were important for intracellular survival in HMDMs and how this was affected by IL-10, transposon sequencing was performed. The size of the S. aureus essential genome was similar between unstimulated HMDMs and the outgrowth control (18.5% vs 18.4%, respectively, with 54.4% overlap) but increased to 22.5% in IL-10-treated macrophages, suggesting that macrophage polarization status exerts differential pressure on S. aureus. Essential genes for S. aureus survival within IL-10-polarized HMDMs were dominated by negative regulatory pathways, including nitrogen and RNA metabolism, whereas S. aureus essential genes within untreated HMDMs were enriched in biosynthetic pathways such as purine and pyrimidine biosynthesis. To explore how IL-10 altered the macrophage intracellular metabolome, targeted metabolomics was performed on HMDMs from six individual donors. IL-10 treatment led to conserved alterations in distinct metabolites that were increased (dihydroxyacetone phosphate, glyceraldehyde-3-phosphate, and acetyl-CoA) or reduced (fructose-6-phosphate, aspartic acid, and ornithine) across donors, whereas other metabolites were variable. Collectively, these findings highlight an important aspect of population-level heterogeneity in human macrophage responsiveness that should be considered when translating results to a patient population.IMPORTANCEOne mechanism that Staphylococcus aureus biofilm elicits in the host to facilitate infection persistence is the production of the anti-inflammatory cytokine interleukin-10 (IL-10). Here, we show that exposure of human monocyte-derived macrophages (HMDMs) to IL-10 promotes S. aureus biofilm formation and programs intracellular bacteria to favor catabolic pathways. Examination of intracellular metabolites in HMDMs revealed heterogeneity between donors that may explain the observed variability in essential genes for S. aureus survival based on nutrient availability for bacteria within the intracellular compartment. Collectively, these studies provide novel insights into how IL-10 polarization affects S. aureus intracellular survival in HMDMs and the importance of considering macrophage heterogeneity between human donors as a variable when examining effector mechanisms.
Collapse
Affiliation(s)
- Blake P. Bertrand
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Dhananjay Shinde
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Vinai C. Thomas
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Marvin Whiteley
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Carolyn B. Ibberson
- School of Biological Sciences, University of Oklahoma, Norman, Oklahoma, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
17
|
De M, Serpa G, Zuiker E, Hisert KB, Liles WC, Manicone AM, Hemann EA, Long ME. MEK1/2 inhibition decreases pro-inflammatory responses in macrophages from people with cystic fibrosis and mitigates severity of illness in experimental murine methicillin-resistant Staphylococcus aureus infection. Front Cell Infect Microbiol 2024; 14:1275940. [PMID: 38352056 PMCID: PMC10861668 DOI: 10.3389/fcimb.2024.1275940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 01/09/2024] [Indexed: 02/16/2024] Open
Abstract
Chronic pulmonary bacterial infections and associated inflammation remain a cause of morbidity and mortality in people with cystic fibrosis (PwCF) despite new modulator therapies. Therapies targeting host factors that dampen detrimental inflammation without suppressing immune responses critical for controlling infections remain limited, while the development of lung infections caused by antimicrobial resistant bacteria is an increasing global problem, and a significant challenge in CF. Pharmacological compounds targeting the mammalian MAPK proteins MEK1 and MEK2, referred to as MEK1/2 inhibitor compounds, have potential combined anti-microbial and anti-inflammatory effects. Here we examined the immunomodulatory properties of MEK1/2 inhibitor compounds PD0325901, trametinib, and CI-1040 on CF innate immune cells. Human CF macrophage and neutrophil phagocytic functions were assessed by quantifying phagocytosis of serum opsonized pHrodo red E. coli, Staphylococcus aureus, and zymosan bioparticles. MEK1/2 inhibitor compounds reduced CF macrophage pro-inflammatory cytokine production without impairing CF macrophage or neutrophil phagocytic abilities. Wild-type C57BL6/J and Cftr tm1kth (F508del homozygous) mice were used to evaluate the in vivo therapeutic potential of PD0325901 compared to vehicle treatment in an intranasal methicillin-resistant Staphylococcus aureus (MRSA) infection with the community-acquired MRSA strain USA300. In both wild-type and CF mice, PD0325901 reduced inflammation associated body mass loss. Wild-type mice treated with PD0325901 had significant reduction in neutrophil-mediated inflammation compared to vehicle treatment groups, with preserved clearance of bacteria in lung, liver, or spleen 1 day after infection in either wild-type or CF mouse models. In summary, this study provides the first data evaluating the therapeutic potential of MEK1/2 inhibitor to modulate CF immune cells and demonstrates that MEK1/2 inhibitors diminish pro-inflammatory responses without impairing host defense mechanisms required for acute pathogen clearance.
Collapse
Affiliation(s)
- Mithu De
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Gregory Serpa
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Eryn Zuiker
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | | | - W. Conrad Liles
- Department of Medicine, Division of Infectious Diseases, University of Washington, Seattle, WA, United States
- Center for Lung Biology, University of Washington, Seattle, WA, United States
| | - Anne M. Manicone
- Center for Lung Biology, University of Washington, Seattle, WA, United States
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, WA, United States
| | - Emily A. Hemann
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Matthew E. Long
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
18
|
Vozza EG, Daly CM, O'Rourke SA, Fitzgerald HK, Dunne A, McLoughlin RM. Staphylococcus aureus suppresses the pentose phosphate pathway in human neutrophils via the adenosine receptor A2aR to enhance intracellular survival. mBio 2024; 15:e0257123. [PMID: 38108639 PMCID: PMC10790693 DOI: 10.1128/mbio.02571-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 12/19/2023] Open
Abstract
IMPORTANCE Staphylococcus aureus is one of the leading causes of antimicrobial-resistant infections whose success as a pathogen is facilitated by its massive array of immune evasion tactics, including intracellular survival within critical immune cells such as neutrophils, the immune system's first line of defense. In this study, we describe a novel pathway by which intracellular S. aureus can suppress the antimicrobial capabilities of human neutrophils by using the anti-inflammatory adenosine receptor, adora2a (A2aR). We show that signaling through A2aR suppresses the pentose phosphate pathway, a metabolic pathway used to fuel the antimicrobial NADPH oxidase complex that generates reactive oxygen species (ROS). As such, neutrophils show enhanced ROS production and reduced intracellular S. aureus when treated with an A2aR inhibitor. Taken together, we identify A2aR as a potential therapeutic target for combatting intracellular S. aureus infection.
Collapse
Affiliation(s)
- Emilio G. Vozza
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Clíodhna M. Daly
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Sinead A. O'Rourke
- Molecular Immunology Group, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Hannah K. Fitzgerald
- Molecular Immunology Group, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Aisling Dunne
- Molecular Immunology Group, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Rachel M. McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
19
|
Horn KJ, Fulte S, Yang M, Lorenz BP, Clark SE. Neutrophil responsiveness to IL-10 impairs clearance of Streptococcus pneumoniae from the lungs. J Leukoc Biol 2024; 115:4-15. [PMID: 37381945 PMCID: PMC10768920 DOI: 10.1093/jleuko/qiad070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/25/2023] [Accepted: 06/20/2023] [Indexed: 06/30/2023] Open
Abstract
The early immune response to bacterial pneumonia requires a careful balance between pathogen clearance and tissue damage. The anti-inflammatory cytokine interleukin (IL)-10 is critical for restraining otherwise lethal pulmonary inflammation. However, pathogen-induced IL-10 is associated with bacterial persistence in the lungs. In this study, we used mice with myeloid cell specific deletion of IL-10R to investigate the cellular targets of IL-10 immune suppression during infection with Streptococcus pneumoniae, the most common bacterial cause of pneumonia. Our findings suggest that IL-10 restricts the neutrophil response to S. pneumoniae, as neutrophil recruitment to the lungs was elevated in myeloid IL-10 receptor (IL-10R)-deficient mice and neutrophils in the lungs of these mice were more effective at killing S. pneumoniae. Improved killing of S. pneumoniae was associated with increased production of reactive oxygen species and serine protease activity in IL-10R-deficient neutrophils. Similarly, IL-10 suppressed the ability of human neutrophils to kill S. pneumoniae. Burdens of S. pneumoniae were lower in myeloid IL-10R-deficient mice compared with wild-type mice, and adoptive transfer of IL-10R-deficient neutrophils into wild-type mice significantly improved pathogen clearance. Despite the potential for neutrophils to contribute to tissue damage, lung pathology scores were similar between genotypes. This contrasts with total IL-10 deficiency, which is associated with increased immunopathology during S. pneumoniae infection. Together, these findings identify neutrophils as a critical target of S. pneumoniae-induced immune suppression and highlight myeloid IL-10R abrogation as a mechanism to selectively reduce pathogen burdens without exacerbating pulmonary damage.
Collapse
Affiliation(s)
- Kadi J Horn
- Department of Otolaryngology, University of Colorado School of Medicine, 12700 East 19th Avenue, Aurora, CO 80045, United States
| | - Sam Fulte
- Department of Otolaryngology, University of Colorado School of Medicine, 12700 East 19th Avenue, Aurora, CO 80045, United States
| | - Michael Yang
- Department of Pathology, University of Colorado School of Medicine, 12631 East 17th Avenue, Aurora, CO80045, United States
| | - Brian P Lorenz
- Department of Otolaryngology, University of Colorado School of Medicine, 12700 East 19th Avenue, Aurora, CO 80045, United States
| | - Sarah E Clark
- Department of Otolaryngology, University of Colorado School of Medicine, 12700 East 19th Avenue, Aurora, CO 80045, United States
| |
Collapse
|
20
|
Xu X, Zhou T, Fang X, Hu L, Zhu J, Zheng F. Biological characteristics and pathogenicity of a Staphylococcus aureus strain with an incomplete hemolytic phenotype isolated from bovine milk. Microbiol Immunol 2024; 68:6-14. [PMID: 37985160 DOI: 10.1111/1348-0421.13102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/21/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023]
Abstract
Staphylococcus aureus is a common pathogen capable of infecting both humans and animals and causing various severe diseases. Here, we aimed to determine the biological features and pathogenicity of S. aureus strain Sa9, of the incomplete hemolysis phenotype, isolated from bovine milk. Sa9 was classified as ST97 by multilocus sequence typing, and it showed increased β-hemolysin expression and lower Hla and Hld expression levels compared with that in the S. aureus USA300 strain LAC. RT-PCR and ELISA results showed that the expression levels of inflammatory cytokines were higher in Sa9-induced mouse primary peritoneal macrophages compared with those induced by the LAC strain. However, the Sa9 strain also mediated anti-inflammatory effects by upregulating IL-10 and IFN-β in macrophages, which were not apparently induced by S. aureus culture supernatants. Phagocytosis and whole-blood survival assays were also performed to assess the in vitro survival of bacteria, and the virulence was evaluated in mice. Although the Sa9 strain showed lower ability of intracellular survival in macrophages than LAC, similar multiplication in human whole blood and pathogenicity toward mice were observed. Taken together, we report that the distinctive immune response induced by the S. aureus strain with an incomplete hemolysis phenotype occurs in cattle, and its potential pathogenicity and risk of transmission to humans require attention.
Collapse
Affiliation(s)
- Xiuhua Xu
- Department of Microbiology, Huadong Medical Institute of Biotechniques, Nanjing, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tingting Zhou
- Department of Microbiology, Huadong Medical Institute of Biotechniques, Nanjing, China
| | - Xueyao Fang
- Key Laboratory of Medicine in Jiangxi Province, Department of Clinical Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Longhua Hu
- Key Laboratory of Medicine in Jiangxi Province, Department of Clinical Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jin Zhu
- Department of Microbiology, Huadong Medical Institute of Biotechniques, Nanjing, China
| | - Feng Zheng
- Department of Microbiology, Huadong Medical Institute of Biotechniques, Nanjing, China
| |
Collapse
|
21
|
Chou LF, Yang HY, Hung CC, Tian YC, Hsu SH, Yang CW. Leptospirosis kidney disease: Evolution from acute to chronic kidney disease. Biomed J 2023; 46:100595. [PMID: 37142093 PMCID: PMC10345244 DOI: 10.1016/j.bj.2023.100595] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 04/17/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023] Open
Abstract
Leptospirosis is a neglected bacterial disease caused by leptospiral infection that carries a substantial mortality risk in severe cases. Research has shown that acute, chronic, and asymptomatic leptospiral infections are closely linked to acute and chronic kidney disease (CKD) and renal fibrosis. Leptospires affect renal function by infiltrating kidney cells via the renal tubules and interstitium and surviving in the kidney by circumventing the immune system. The most well-known pathogenic molecular mechanism of renal tubular damage caused by leptospiral infection is the direct binding of the bacterial outer membrane protein LipL32 to toll-like receptor-2 expressed in renal tubular epithelial cells (TECs) to induce intracellular inflammatory signaling pathways. These pathways include the production of tumor necrosis factor (TNF)-α and nuclear factor kappa activation, resulting in acute and chronic leptospirosis-related kidney injury. Few studies have investigated the relationship between acute and chronic renal diseases and leptospirosis and further evidence is necessary. In this review, we intend to discuss the roles of acute kidney injury (AKI) to/on CKD in leptospirosis. This study reviews the molecular pathways underlying the pathogenesis of leptospirosis kidney disease, which will assist in concentrating on potential future research directions.
Collapse
Affiliation(s)
- Li-Fang Chou
- Kidney Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan; Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, Department of Biochemistry, Chang Gung University, Taoyuan, Taiwan
| | - Huang-Yu Yang
- Kidney Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan; Department of Nephrology, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Chieh Hung
- Kidney Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan; Department of Nephrology, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ya-Chung Tian
- Kidney Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan; Department of Nephrology, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shen-Hsing Hsu
- Kidney Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan
| | - Chih-Wei Yang
- Kidney Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan; Department of Nephrology, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
22
|
Yamashita Y, Yasuda I, Tanaka T, Ikeda T, Terada M, Takaki M, Tsuchihashi Y, Asoh N, Ohara Y, Enany S, Kobayashi H, Matsumoto S, Morimoto K. Antigen-specific cytokine profiles for pulmonary Mycobacterium avium complex disease stage diagnosis. Front Immunol 2023; 14:1222428. [PMID: 37520555 PMCID: PMC10380938 DOI: 10.3389/fimmu.2023.1222428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 06/23/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Controlling pulmonary Mycobacterium avium complex (MAC) disease is difficult because there is no way to know the clinical stage accurately. There have been few attempts to use cell-mediated immunity for diagnosing the stage. The objective of this study was to characterize cytokine profiles of CD4+T and CD19+B cells that recognize various Mycobacterium avium-associated antigens in different clinical stages of MAC. Methods A total of 47 MAC patients at different stages based on clinical information (14 before-treatment, 16 on-treatment, and 17 after-treatment) and 17 healthy controls were recruited. Peripheral blood mononuclear cells were cultured with specific antigens (MAV0968, 1160, 1276, and 4925), and the cytokine profiles (IFN-γ, TNF-α, IL-2, IL-10, IL-13, and IL-17) of CD4+/CD3+ and CD19+ cells were analyzed by flow cytometry. Results The response of Th1 cytokines such as IFN-γ and TNF-α against various antigens was significantly higher in both the on-treatment and after-treatment groups than in the before-treatment group and control (P < 0.01-0.0001 and P < 0.05-0.0001). An analysis of polyfunctional T cells suggested that the presence of IL-2 is closely related to the stage after the start of treatment (P = 0.0309-P < 0.0001) and is involved in memory function. Non-Th1 cytokines, such as IL-10 and IL-17, showed significantly higher responses in the before-treatment group (P < 0.0001 and P < 0.01-0.0001). These responses were not observed with purified protein derivative (PPD). CD19+B cells showed a response similar to that of CD4+T cells. Conclusion There is a characteristic cytokine profile at each clinical stage of MAC.
Collapse
Affiliation(s)
- Yoshiro Yamashita
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
- Department of Respiratory Medicine, Shunkaikai Inoue Hospital, Nagasaki, Nagasaki, Japan
| | - Ikkoh Yasuda
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
- Department of General Internal Medicine and Clinical Infectious Diseases, Fukushima Medical University, Fukushima, Fukushima, Japan
| | - Takeshi Tanaka
- Infection Control and Education Center, Nagasaki University Hospital, Nagasaki, Nagasaki, Japan
| | - Toru Ikeda
- Department of Respiratory Medicine, Nagasaki Rosai Hospital, Sasebo, Nagasaki, Japan
| | - Mayumi Terada
- Department of Internal Medicine, Koseikai Nijigaoka Hospital, Nagasaki, Nagasaki, Japan
| | - Masahiro Takaki
- Department of Respiratory Medicine, Shunkaikai Inoue Hospital, Nagasaki, Nagasaki, Japan
| | - Yoshiko Tsuchihashi
- Department of Respiratory Medicine, Juzenkai Hospital, Nagasaki, Nagasaki, Japan
| | - Norichika Asoh
- Department of Respiratory Medicine, Juzenkai Hospital, Nagasaki, Nagasaki, Japan
| | - Yukiko Ohara
- Department of Bacteriology, Niigata University Graduate School of Medicine, Niigata, Niigata, Japan
| | - Shymaa Enany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
- Biomedical Research Department, Armed Force College of Medicine, Cairo, Egypt
| | - Haruka Kobayashi
- Department of Bacteriology, Niigata University Graduate School of Medicine, Niigata, Niigata, Japan
| | - Sohkichi Matsumoto
- Department of Bacteriology, Niigata University Graduate School of Medicine, Niigata, Niigata, Japan
| | - Konosuke Morimoto
- Department of Internal Medicine, Koseikai Nijigaoka Hospital, Nagasaki, Nagasaki, Japan
- Department of Respiratory Infectious Disease, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| |
Collapse
|
23
|
Chatuphonprasert W, Tatiya-aphiradee N, Sutthanut K, Thammawat S, Puthongking P, Nopwinyoowong N, Jarukamjorn K. Combinatory effects of Dipterocarpus alatus twig emulgel: Wound-restoring, antibacterial, and anti-inflammatory activities against methicillin-resistant Staphylococcus aureus-infected mouse superficial wounds. Heliyon 2023; 9:e17483. [PMID: 37416687 PMCID: PMC10320117 DOI: 10.1016/j.heliyon.2023.e17483] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/22/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023] Open
Abstract
Dipterocarpus alatus has been used for the treatment of infectious skin diseases and ulcerative wounds in Thai traditional medicine. A major pathogen in human superficial skin infections is methicillin-resistant Staphylococcus aureus (MRSA). This study determined the wound healing, antibacterial, and anti-inflammatory activities of D. alatus twig emulgel against MRSA-infected mouse superficial skin wounds. Ethyl acetate-methanol crude extract of D. alatus twig was incorporated into emulgel at concentrations of 20 and 40 mg/g (D20 and D40) and its activity was compared to tetracycline emulgel (160 μg/g, Tetra). MRSA-infected superficial wounds demonstrated decreased skin barrier strength, increased transepidermal water loss (TEWL), and mast cell accumulation. Expression of toll-like receptor 2 (TLR-2), NF-κβ, TNFα, IL-1β, IL-6 and IL-10 genes were induced after MRSA infection. Daily application of 100 μL of D20 or D40 for 9 days restored skin barrier strength and TEWL while reducing mast cell and MRSA numbers compared to the non-treated group (MRSA-NT). The wounds treated with D20 and D40 were entirely healed on day 9. Expression of TLR-2 and cytokine-related genes NF-κβ, TNFα, IL-1β, IL-6 and IL-10 were normalized by treatment with either D20 or D40. Therefore, emulgel containing 20 to 40 mg/g ethyl acetate-methanol crude D. alatus twig extract is a good candidate for development as a topical formulation for MRSA-infected ulcerated wounds.
Collapse
Affiliation(s)
- Waranya Chatuphonprasert
- Division of Pre-clinic, Faculty of Medicine, Mahasarakham University, Maha Sarakham, 44000, Thailand
- Research Group for Pharmaceutical Activities of Natural Products Using Pharmaceutical Biotechnology (PANPB), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Nitima Tatiya-aphiradee
- Research Group for Pharmaceutical Activities of Natural Products Using Pharmaceutical Biotechnology (PANPB), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Khaetthareeya Sutthanut
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002 Thailand
| | - Sutthiwan Thammawat
- Division of Pre-clinic, Faculty of Medicine, Mahasarakham University, Maha Sarakham, 44000, Thailand
| | - Ploenthip Puthongking
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002 Thailand
| | - Naroeporn Nopwinyoowong
- Research Group for Pharmaceutical Activities of Natural Products Using Pharmaceutical Biotechnology (PANPB), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Kanokwan Jarukamjorn
- Research Group for Pharmaceutical Activities of Natural Products Using Pharmaceutical Biotechnology (PANPB), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002 Thailand
| |
Collapse
|
24
|
Dutta P, Bishayi B. IL-10 in combination with IL-12 and TNF-α attenuates CXCL8/CXCR1 axis in peritoneal macrophages of mice infected with Staphylococcus aureus through the TNFR1-IL-1R-NF-κB pathway. Int Immunopharmacol 2023; 120:110297. [PMID: 37207443 DOI: 10.1016/j.intimp.2023.110297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/21/2023]
Abstract
Overexpression of Staphylococcus aureus mediated CXCL8/CXCR1 axis is a major cause of sepsis and severe inflammatory diseases. This chemokine acts conjointly with various pro-inflammatory and anti-inflammatory cytokines that govern the severity of inflammation. The effects of different combinations of exogenous cytokines on CXCR1 expression in macrophages remain undetermined. Exogenous cytokine and anti-inflammatory cytokine therapy had been used to modulate CXCL8 and CXCR1 expression in peritoneal macrophages. Male Swiss albino mice were inoculated with live S. aureus (106 cells/ mouse) for the development of infection. Exogenous cytokines (TNF-α, IL-12, IFN-γ and IL-10) were administered intraperitoneally (single or combination) 24 h post S. aureus infection. The mice were sacrificed and peritoneal macrophages were isolated three days post infection. CXCL8, IL-12, IL-10 secretion, ROS generation and the bacterial phagocytic process had been evaluated. Western blot was used to study the expressions of TNFR1, IL-1R, CXCR1 and NF-κB. TNF-α, IL-12 and IFN-γ treatments aggravated CXCL8 and CXCR1 expression in the macrophages of infected mice. TNF-α + IFN-γ treatment was a major inducer of nitric oxide release and mediated maximum bacterial killing. IL-12 + TNF-α treatment was most potent in increasing ROS, CXCL8/CXCR1 expression through increased levels of TNFR1, IL-1R and NF-κB activation. IL-10 reversed the effects of exogenous cytokines but also impaired the bacterial clearance phenomenon in peritoneal lavage. Treatment with IL-12 + TNF-α + IL-10 was most effective in ameliorating oxidative stress, reduced CXCL8 release and expression levels of TNFR1, IL-1R, and NF-κB. Concludingly, IL-12 + TNF-α + IL-10 treatment mitigated CXCL8/CXCR1 expression and inflammatory signalling via downregulation of TNFR1-IL-1R-NF-κB pathway in peritoneal macrophages and inflammatory sequelae during S. aureus infection.
Collapse
Affiliation(s)
- Puja Dutta
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India.
| |
Collapse
|
25
|
Kak G, Van Roy Z, Heim CE, Fallet RW, Shi W, Roers A, Duan B, Kielian T. IL-10 production by granulocytes promotes Staphylococcus aureus craniotomy infection. J Neuroinflammation 2023; 20:114. [PMID: 37179295 PMCID: PMC10183138 DOI: 10.1186/s12974-023-02798-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Treatment of brain tumors, epilepsy, or hemodynamic abnormalities requires a craniotomy to access the brain. Nearly 1 million craniotomies are performed in the US annually, which increase to ~ 14 million worldwide and despite prophylaxis, infectious complications after craniotomy range from 1 to 3%. Approximately half are caused by Staphylococcus aureus (S. aureus), which forms a biofilm on the bone flap that is recalcitrant to antibiotics and immune-mediated clearance. However, the mechanisms responsible for the persistence of craniotomy infection remain largely unknown. The current study examined the role of IL-10 in promoting bacterial survival. METHODS A mouse model of S. aureus craniotomy infection was used with wild type (WT), IL-10 knockout (KO), and IL-10 conditional KO mice where IL-10 was absent in microglia and monocytes/macrophages (CX3CR1CreIL-10 fl/fl) or neutrophils and granulocytic myeloid-derived suppressor cells (G-MDSCs; Mrp8CreIL-10 fl/fl), the major immune cell populations in the infected brain vs. subcutaneous galea, respectively. Mice were examined at various intervals post-infection to quantify bacterial burden, leukocyte recruitment, and inflammatory mediator production in the brain and galea to assess the role of IL-10 in craniotomy persistence. In addition, the role of G-MDSC-derived IL-10 on neutrophil activity was examined. RESULTS Granulocytes (neutrophils and G-MDSCs) were the major producers of IL-10 during craniotomy infection. Bacterial burden was significantly reduced in IL-10 KO mice in the brain and galea at day 14 post-infection compared to WT animals, concomitant with increased CD4+ and γδ T cell recruitment and cytokine/chemokine production, indicative of a heightened proinflammatory response. S. aureus burden was reduced in Mrp8CreIL-10 fl/fl but not CX3CR1CreIL-10 fl/fl mice that was reversed following treatment with exogenous IL-10, suggesting that granulocyte-derived IL-10 was important for promoting S. aureus craniotomy infection. This was likely due, in part, to IL-10 production by G-MDSCs that inhibited neutrophil bactericidal activity and TNF production. CONCLUSION Collectively, these findings reveal a novel role for granulocyte-derived IL-10 in suppressing S. aureus clearance during craniotomy infection, which is one mechanism to account for biofilm persistence.
Collapse
Affiliation(s)
- Gunjan Kak
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Zachary Van Roy
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Cortney E Heim
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Rachel W Fallet
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Wen Shi
- Mary and Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Axel Roers
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Bin Duan
- Mary and Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA.
| |
Collapse
|
26
|
Peng J, Lu Q, Yuan L, Zhang H. Synthetic Cationic Lipopeptide Can Effectively Treat Mouse Mastitis Caused by Staphylococcus aureus. Biomedicines 2023; 11:biomedicines11041188. [PMID: 37189805 DOI: 10.3390/biomedicines11041188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Mastitis caused by Staphylococcus aureus (S. aureus) in dairy cows is one of the most common clinical diseases in dairy cattle. Unfortunately, traditional antibiotic treatment has resulted in the emergence of drug-resistant strains of bacteria, making this disease more difficult to treat. Therefore, novel lipopeptide antibiotics are becoming increasingly important in treating bacterial diseases, and developing novel antibiotics is critical in controlling mastitis in dairy cows. We designed and synthesized three cationic lipopeptides with palmitic acid, all with two positive charges and dextral amino acids. The lipopeptides' antibacterial activity against S. aureus was determined using MIC and scanning electron microscopy. The safety concentration range of lipopeptides for clinical usage was then estimated using the mouse erythrocyte hemolysis assay and CCK8 cytotoxicity. Finally, lipopeptides with high antibacterial activity and minimal cytotoxicity were selected for the treatment experiments regarding mastitis in mice. The observation of histopathological changes, bacterial tissue load and expression of inflammatory factors determined the therapeutic effects of lipopeptides on mastitis in mice. The results showed that all three lipopeptides displayed some antibacterial activity against S. aureus, with C16dKdK having a strong antibacterial impact and being able to treat the mastitis induced by S. aureus infection in mice within a safe concentration range. The findings of this study can be used as a starting point for the development of new medications for the treatment of mastitis in dairy cows.
Collapse
Affiliation(s)
- Jie Peng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Qiangsheng Lu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Lvfeng Yuan
- Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou 730000, China
| | - Hecheng Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
27
|
Ramata-Stunda A, Boroduskis M, Kaktina E, Patetko L, Kalnenieks U, Lasa Z, Rubina M, Strazdina I, Kalnins G, Rutkis R. Comparative Evaluation of Existing and Rationally Designed Novel Antimicrobial Peptides for Treatment of Skin and Soft Tissue Infections. Antibiotics (Basel) 2023; 12:antibiotics12030551. [PMID: 36978418 PMCID: PMC10044245 DOI: 10.3390/antibiotics12030551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/01/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Skin and soft tissue infections (SSTIs) and acne are among the most common skin conditions in primary care. SSTIs caused by ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter sp.) can range in severity, and treating them is becoming increasingly challenging due to the growing number of antibiotic-resistant pathogens. There is also a rise in antibiotic-resistant strains of Cutibacterium acne, which plays a role in the development of acne. Antimicrobial peptides (AMPs) are considered to be a promising solution to the challenges posed by antibiotic resistance. In this study, six new AMPs were rationally designed and compared to five existing peptides. The MIC values against E. coli, P. aeruginosa, K. pneumoniae, E. faecium, S. aureus, and C. acnes were determined, and the peptides were evaluated for cytotoxicity using Balb/c 3T3 cells and dermal fibroblasts, as well as for hemolytic activity. The interaction with bacterial membranes and the effect on TNF-α and IL-10 secretion were also evaluated for selected peptides. Of the tested peptides, RP556 showed high broad-spectrum antibacterial activity without inducing cytotoxicity or hemolysis, and it stimulated the production of IL-10 in LPS-stimulated peripheral blood mononuclear cells. Four of the novel AMPs showed pronounced specificity against C. acnes, with MIC values (0.3–0.5 μg/mL) below the concentrations that were cytotoxic or hemolytic.
Collapse
Affiliation(s)
- Anna Ramata-Stunda
- Alternative Plants Ltd., 2 Podraga Str., LV-1007 Riga, Latvia
- Correspondence:
| | | | - Elza Kaktina
- Alternative Plants Ltd., 2 Podraga Str., LV-1007 Riga, Latvia
| | - Liene Patetko
- Laboratory of Bioanalytical and Biodosimetry Methods, Faculty of Biology, University of Latvia, 3 Jelgavas Str., LV-1004 Riga, Latvia
| | - Uldis Kalnenieks
- Alternative Plants Ltd., 2 Podraga Str., LV-1007 Riga, Latvia
- Institute of Microbiology and Biotechnology, University of Latvia, 1 Jelgavas Str., LV-1004 Riga, Latvia
| | - Zane Lasa
- Institute of Microbiology and Biotechnology, University of Latvia, 1 Jelgavas Str., LV-1004 Riga, Latvia
| | - Marta Rubina
- Institute of Microbiology and Biotechnology, University of Latvia, 1 Jelgavas Str., LV-1004 Riga, Latvia
| | - Inese Strazdina
- Institute of Microbiology and Biotechnology, University of Latvia, 1 Jelgavas Str., LV-1004 Riga, Latvia
| | - Gints Kalnins
- Latvian Biomedical Research and Study Centre, 1 Ratsupites Str., LV-1067 Riga, Latvia
| | - Reinis Rutkis
- Institute of Microbiology and Biotechnology, University of Latvia, 1 Jelgavas Str., LV-1004 Riga, Latvia
| |
Collapse
|
28
|
Staphylococcus aureus-induced immunosuppression mediated by IL-10 and IL-27 facilitates nasal colonisation. PLoS Pathog 2022; 18:e1010647. [PMID: 35776778 PMCID: PMC9282462 DOI: 10.1371/journal.ppat.1010647] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/14/2022] [Accepted: 06/06/2022] [Indexed: 11/19/2022] Open
Abstract
Staphylococcus aureus persistently colonises the anterior nares of a significant proportion of the healthy population, however the local immune response elicited during S. aureus nasal colonisation remains ill-defined. Local activation of IL-17/IL-22 producing T cells are critical for controlling bacterial clearance from the nasal cavity. However, recurrent and long-term colonisation is commonplace indicating efficient clearance does not invariably occur. Here we identify a central role for the regulatory cytokine IL-10 in facilitating bacterial persistence during S. aureus nasal colonisation in a murine model. IL-10 is produced rapidly within the nasal cavity following S. aureus colonisation, primarily by myeloid cells. Colonised IL-10-/- mice demonstrate enhanced IL-17+ and IL-22+ T cell responses and more rapidly clear bacteria from the nasal tissues as compared with wild-type mice. S. aureus also induces the regulatory cytokine IL-27 within the nasal tissue, which acts upstream of IL-10 promoting its production. IL-27 blockade reduces IL-10 production within the nasal cavity and improves bacterial clearance. TLR2 signalling was confirmed to be central to controlling the IL-10 response. Our findings conclude that during nasal colonisation S. aureus creates an immunosuppressive microenvironment through the local induction of IL-27 and IL-10, to dampen protective T cell responses and facilitate its persistence. Nasal colonisation by the bacterium Staphylococcus aureus is a very common occurrence in the human population. However there is a lack of knowledge on the immune response that controls nasal colonisation. It is known that a local pro-inflammatory immune response is important for bacterial clearance, however sustained colonisation is commonplace suggesting efficient clearance may not be occurring. Here we demonstrate for the first time that S. aureus is manipulating the host immune response by promoting immunosuppression in the nasal cavity which enables bacterial survival. We found that the regulatory proteins IL-10 and IL-27 are central to this suppressive response and result in reduced protective T cell responses. We also demonstrate that S. aureus is inducing IL-27 production to enhance IL-10 production in order to prolong bacterial colonisation. Our findings show that the host-pathogen interaction during nasal colonisation is more complex than previously described and that S. aureus is capable of manipulating the regulatory immune response of the host for its’ own benefit.
Collapse
|
29
|
Kim SY, Lee JE. Resolvin D1 Inhibits Corneal Inflammation in Staphylococcus Aureus Keratitis. Ocul Immunol Inflamm 2022:1-8. [PMID: 35522258 DOI: 10.1080/09273948.2022.2070504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE To investigate the role of lipid mediator, resolvin D1 (RvD1), in corneal inflammation. METHODS The anti-inflammatory effect of RvD1 on stimulated human corneal epithelial cells (HCECs) was assessed. C57BL/6 mice corneas were abraded and treated with RvD1 after stimulation with Staphylococcus aureus. Cytokine levels in the corneas, cervical drainage lymph nodes (DLNs), and spleens were measured. Anterior segment photography and optical coherence tomography quantified the changes in corneal thickness and haziness. Neutrophil infiltration in the cornea was examined by haematoxylin and eosin (H&E) staining and immunohistochemistry. RESULTS RvD1 significantly inhibited cytokine production in HCECs and mouse corneas, cervical DLNs, and spleens while stimulating interleukin-10 (IL-10) production. Corneal opacity development, thickening, and neutrophil infiltration significantly reduced in response to RvD1 stimulation in the S. aureus-infected mice corneas. CONCLUSION RvD1 inhibited S. aureus-induced corneal inflammation. These results potentiate RvD1 as an anti-inflammatory therapy for patients with corneal inflammation induced by bacterial keratitis.
Collapse
Affiliation(s)
- Sang Yoon Kim
- Department of Ophthalmology, Pusan National University School of Medicine, Yangsan, South Korea
| | - Ji Eun Lee
- Department of Ophthalmology, Pusan National University School of Medicine, Yangsan, South Korea.,Department of Ophthalmology, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| |
Collapse
|
30
|
Fujii K. Pathogenesis of cutaneous T cell lymphoma: Involvement of Staphylococcus aureus. J Dermatol 2021; 49:202-209. [PMID: 34927279 DOI: 10.1111/1346-8138.16288] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 12/30/2022]
Abstract
Mycosis fungoides (MF) and Sézary syndrome (SS) are representative cutaneous lymphomas. In their early stage, a small number of tumor cells and a large number of non-malignant cells form a Th1-dominant tumor microenvironment. Increase in malignant T cells is accompanied by a decrease in CD8-positive T cells, with a shift toward a Th2-dominant milieu in advanced-stage lesions. The etiologies of MF/SS are diverse, and the underlying pathogenetic mechanisms are yet to be elucidated. Advanced MF/SS is known to be highly sensitive to Staphylococcus aureus, and the majority of deaths are caused by severe infections. The susceptibility to infection is associated with barrier dysfunction and immunosuppression, which are the main symptoms of MF. In recent years, skin-colonizing S. aureus has been identified to not only cause severe infections but also play an important role in the pathogenesis of MF/SS. Staphylococcal superantigens activate the proliferation of tumor cells and induce CD25 upregulation, FOXP3 expression, IL-17 expression, and miR-155 expression. Alpha-toxin eliminates non-neoplastic CD4-positive cells and CD8-positive cells and plays a major role in tumor cell selection. Lipoprotein may also be associated with the induction of Th2-dominant milieu. Antibiotic therapy for S. aureus eradication has been reported to cause considerable clinical improvement in the majority of individuals with advanced cutaneous T-cell lymphoma. Therefore, S. aureus may be a novel target for the treatment of advanced-stage MF/SS in the future.
Collapse
Affiliation(s)
- Kazuyasu Fujii
- Department of Dermatology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
31
|
Kwiatkowski P, Kurzawski M, Łopusiewicz Ł, Pruss A, Sienkiewicz M, Wojciechowska-Koszko I, Dołęgowska B. Preliminary evaluation of selected inflammatory cytokine gene expression in lymphocytes isolated from whole human blood infected with trans-anethole-treated Staphylococcus aureus Newman strain. Lett Appl Microbiol 2021; 74:513-518. [PMID: 34904269 DOI: 10.1111/lam.13632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 11/28/2022]
Abstract
In our previous study based on a whole-blood model of sepsis infected with trans-anethole (TA)-treated Staphylococcus aureus, we have found that innate immune response was more effective in comparison to non-treated cells. Due to the previous observation, in the current preliminary study, a primary adaptive immune response was analysed. This study was conducted to evaluate the expression of selected cytokine (IL1B, IL2, IL6, IL10, TNF, TGFB1, IFNG) and Toll-like receptor (TLR2) genes in lymphocytes isolated from whole human blood infected with S. aureus Newman strain treated with TA. The lymphocytes were isolated by density gradient centrifugation from blood samples infected with S. aureus, as well as from non-infected samples. Gene expression was measured using quantitative real-time PCR. The lymphocytes isolated from the blood infected with TA-treated staphylococcal cells demonstrated significantly greater IL10, IL1B, IL6, TNF and TLR2 expression. Hence, it is possible that the previously observed changes in the surface structure of TA-treated S. aureus Newman strain may significantly increase the relative expression of IL10, IL1B, IL6, TNF and TLR2 genes in lymphocytes; however, further studies are needed.
Collapse
Affiliation(s)
- P Kwiatkowski
- Department of Diagnostic Immunology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - M Kurzawski
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Ł Łopusiewicz
- Center of Bioimmobilisation and Innovative Packaging Materials, Faculty of Food Sciences and Fisheries, West Pomeranian University of Technology Szczecin, Szczecin, Poland
| | - A Pruss
- Department of Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - M Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostic, Medical University of Lodz, Lodz, Poland
| | - I Wojciechowska-Koszko
- Department of Diagnostic Immunology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - B Dołęgowska
- Department of Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
32
|
Muniz IPR, Galantini MPL, Ribeiro IS, Gonçalves CV, Dos Santos DP, Moura TC, Silva ES, Silva NR, Cipriano BP, Correia TML, de Jesus Soares T, de Freitas LM, Costa DJ, da Silva RAA. Antimicrobial photodynamic therapy (aPDT) with curcumin controls intradermal infection by Staphylococcus aureus in mice with type 1 diabetes mellitus: a pilot study. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2021; 224:112325. [PMID: 34598018 DOI: 10.1016/j.jphotobiol.2021.112325] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is one of the main pathogens that cause infections in diabetic individuals. In this paper, we report the outcomes of our investigation on the intradermal application of antimicrobial photodynamic therapy (PDT) with curcumin in an infection induced by MRSA ATCC 43300 strain in the ear of mice with Type 1 Diabetes Mellitus (T1DM). A solution containing 100 μg of curcumin was photoactivated ex vivo with a LED light (450 nm) delivering a fluency of 13.5 J/cm3. This solution was administered in the ear intradermally, at the same inoculum site as the MRSA ATCC 43300 strain (PDT Group). This study also included the use of two control groups (both infected): One was treated with saline and the other was treated with non-photoactivated curcumin. The animals were euthanized 24 h after these treatments and samples of draining lymph node and treated ear were collected for examination. The PDT group showed lower bacterial load in the draining lymph node when compared to the saline and curcumin groups (p-value <0.05) 24 h after treatment. In addition to bacterial load, the PDT group presented a higher concentration of nitrates and nitrites in the draining lymph node when compared to the saline and curcumin groups (p-value <0.001). Examining the infectious site, despite apparently having similar inflammatory cell recruitment compared with the control groups, the PDT group showed a profile with less intense activity in the myeloperoxidase expression when compared to the saline group (p-value <0.001). Additionally, the detected concentration of cytokines such as IL-1β, IL-12, and IL-10 was significantly lower in the PDT group when compared to the saline group (p-value <0.01; p-value <0.05; p-value <0.05, respectively), thus presenting a less intense inflammatory response during infection resolution. Our pilot study showed for the first time the therapeutic potential of PDT using curcumin when administered intradermally in the treatment of infections caused by S. aureus in mice with T1DM.
Collapse
Affiliation(s)
- Igor Pereira Ribeiro Muniz
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Maria Poliana Leite Galantini
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Israel Souza Ribeiro
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil; Universidade Federal do Sul da Bahia, Campus Paulo Freire, 250 Praça Joana Angélica, Bairro São José, 45.988-058, Teixeira de Freitas, Bahia, Brazil
| | - Caroline Vieira Gonçalves
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Denisar Palmito Dos Santos
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Tatyana Chagas Moura
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Emely Soares Silva
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Nathalia Rosa Silva
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Barbara Porto Cipriano
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Thiago Macêdo Lopes Correia
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Telma de Jesus Soares
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Leandro Martins de Freitas
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Dirceu Joaquim Costa
- Universidade Estadual do Sudoeste da Bahia, Campus Vitória da Conquista, Av. Edmundo Silveira Flores, 27-43 - Lot, Alto da Boa Vista, CEP: 45029-066 Vitória da Conquista, Bahia, Brasil
| | - Robson Amaro Augusto da Silva
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil.
| |
Collapse
|
33
|
Eltwisy HO, Abdel-Fattah M, Elsisi AM, Omar MM, Abdelmoteleb AA, El-Mokhtar MA. Pathogenesis of Staphylococcus haemolyticus on primary human skin fibroblast cells. Virulence 2021; 11:1142-1157. [PMID: 32799619 PMCID: PMC7549902 DOI: 10.1080/21505594.2020.1809962] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
STAPHYLOCOCCUS HAEMOLYTICUS (S. haemolyticus) is one of the Coagulase-negative staphylococci (CoNS) that inhabits the skin as a commensal. It is increasingly implicated in opportunistic infections, including diabetic foot ulcer (DFU) infections. In contrast to the abundance of information available for S. aureus and S. epidermidis, little is known about the pathogenicity of S. haemolyticus, despite the increased prevalence of this pathogen in hospitalized patients. We described, for the first time, the pathogenesis of different clinical isolates of S. haemolyticus isolated from DFU on primary human skin fibroblast (PHSF) cells. Virulence-related genes were investigated, adhesion and invasion assays were carried out using Giemsa stain, transmission electron microscopy (TEM), MTT and flowcytometry assays. Our results showed that most S. haemolyticus carried different sets of virulence-related genes. S. haemolyticus adhered to the PHSF cells to variable degrees. TEM showed that the bacteria were engulfed in a zipper-like mechanism into a vacuole inside the cell. Bacterial internalization was confirmed using flowcytometry and achieved high intracellular levels. PHSF cells infected with S.haemolyticus suffered from amarked decrease in viability and increased apoptosis when treated with whole bacterial suspensions or cell-free supernatants but not with heat-treated cells. After co-culture with PBMCs, S. haemolyticus induced high levels of pro-inflammatory cytokines. This study highlights the significant development of S. haemolyticus, which was previously considered a contaminant when detected in cultures of clinical samples. Their high ability to adhere, invade and kill the PHSF cells illustrate the severe damage associated with DFU infections. ABBREVIATIONS CoNS, coagulase-negative staphylococci; DFU, diabetic foot ulcer; DM, diabetes mellitus; DMEM, Dulbecco's Modified Eagle Medium; MTT, 3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide; PBMCs,peripheral blood mononuclear cells; PHSF, primary human skin fibroblast; CFU, colony-forming unit.
Collapse
Affiliation(s)
- Hala O Eltwisy
- Department of Microbiology, Faculty of Science, Beni-Suef University , Beni-Suef, Egypt
| | - Medhat Abdel-Fattah
- Department of Microbiology and Botany, Faculty of Science, Beni-Suef University , Beni-Suef, Egypt
| | - Amani M Elsisi
- Department of Pharmaceutics and Industrial Pharmacy, Beni-Suef University , Beni-Suef, Egypt
| | - Mahmoud M Omar
- Department of Pharmaceutics and Industrial Pharmacy, Deraya University , El-Minia, Egypt
| | | | - Mohamed A El-Mokhtar
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University , Assiut, Egypt
| |
Collapse
|
34
|
Pacheco GA, Gálvez NMS, Soto JA, Andrade CA, Kalergis AM. Bacterial and Viral Coinfections with the Human Respiratory Syncytial Virus. Microorganisms 2021; 9:microorganisms9061293. [PMID: 34199284 PMCID: PMC8231868 DOI: 10.3390/microorganisms9061293] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/05/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
The human respiratory syncytial virus (hRSV) is one of the leading causes of acute lower respiratory tract infections in children under five years old. Notably, hRSV infections can give way to pneumonia and predispose to other respiratory complications later in life, such as asthma. Even though the social and economic burden associated with hRSV infections is tremendous, there are no approved vaccines to date to prevent the disease caused by this pathogen. Recently, coinfections and superinfections have turned into an active field of study, and interactions between many viral and bacterial pathogens have been studied. hRSV is not an exception since polymicrobial infections involving this virus are common, especially when illness has evolved into pneumonia. Here, we review the epidemiology and recent findings regarding the main polymicrobial infections involving hRSV and several prevalent bacterial and viral respiratory pathogens, such as Staphylococcus aureus, Pseudomonas aeruginosa, Streptococcus pneumoniae, Haemophilus influenzae, Moraxella catarrhalis, Klebsiella pneumoniae, human rhinoviruses, influenza A virus, human metapneumovirus, and human parainfluenza viruses. As reports of most polymicrobial infections involving hRSV lack a molecular basis explaining the interaction between hRSV and these pathogens, we believe this review article can serve as a starting point to interesting and very much needed research in this area.
Collapse
Affiliation(s)
- Gaspar A. Pacheco
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute of Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (G.A.P.); (N.M.S.G.); (J.A.S.); (C.A.A.)
| | - Nicolás M. S. Gálvez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute of Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (G.A.P.); (N.M.S.G.); (J.A.S.); (C.A.A.)
| | - Jorge A. Soto
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute of Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (G.A.P.); (N.M.S.G.); (J.A.S.); (C.A.A.)
| | - Catalina A. Andrade
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute of Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (G.A.P.); (N.M.S.G.); (J.A.S.); (C.A.A.)
| | - Alexis M. Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute of Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (G.A.P.); (N.M.S.G.); (J.A.S.); (C.A.A.)
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
- Correspondence: ; Tel.: +56-2-686-2842; Fax: +56-2-222-5515
| |
Collapse
|
35
|
Lakshmaiah Narayana J, Golla R, Mishra B, Wang X, Lushnikova T, Zhang Y, Verma A, Kumar V, Xie J, Wang G. Short and Robust Anti-Infective Lipopeptides Engineered Based on the Minimal Antimicrobial Peptide KR12 of Human LL-37. ACS Infect Dis 2021; 7:1795-1808. [PMID: 33890759 DOI: 10.1021/acsinfecdis.1c00101] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This study aims to push the frontier of the engineering of human cathelicidin LL-37, a critical antimicrobial innate immune peptide that wards off invading pathogens. By sequential truncation of the smallest antibacterial peptide (KR12) of LL-37 and conjugation with fatty acids, with varying chain lengths, a library of lipopeptides is generated. These peptides are subjected to antibacterial activity and hemolytic assays. Candidates (including both forms made of l- and d-amino acids) with the optimal cell selectivity are subsequently fed to the second layer of in vitro filters, including salts, pH, serum, and media. These practices lead to the identification of a miniature LL-37 like peptide (d-form) with selectivity, stability, and robust antimicrobial activity in vitro against both Gram-positive and negative bacteria. Proteomic studies reveal far fewer serum proteins that bind to the d-form than the l-form peptide. C10-KR8d targets bacterial membranes to become helical, making it difficult for bacteria to develop resistance in a multiple passage experiment. In vivo, C10-KR8d is able to reduce bacterial burden of methicillin-resistant Staphylococcus aureus (MRSA) USA300 LAC in neutropenic mice. In addition, this designer peptide prevents bacterial biofilm formation in a catheter-associated mouse model. Meanwhile, C10-KR8d also recruits cytokines to the vicinity of catheters to clear infection. Thus, based on the antimicrobial region of LL-37, this study succeeds in identifying the smallest anti-infective peptide C10-KR8d with both robust antimicrobial, antibiofilm, and immune modulation activities.
Collapse
Affiliation(s)
- Jayaram Lakshmaiah Narayana
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Radha Golla
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Biswajit Mishra
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Xiuqing Wang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Tamara Lushnikova
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Yingxia Zhang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Atul Verma
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Vikas Kumar
- Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68130, United States
| | - Guangshun Wang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| |
Collapse
|
36
|
Botheras CL, Bowe SJ, Cowan R, Athan E. C-reactive protein predicts complications in community-associated S. aureus bacteraemia: a cohort study. BMC Infect Dis 2021; 21:312. [PMID: 33794783 PMCID: PMC8015062 DOI: 10.1186/s12879-021-05962-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/07/2021] [Indexed: 12/12/2022] Open
Abstract
Background Staphylococcus aureus (S. aureus) bacteraemia is increasingly acquired from community settings and is associated with a mortality rate of up to 40% following complications. Identifying risk factors for complicated S. aureus bacteraemia would aid clinicians in targeting patients that benefit from expedited investigations and escalated care. Methods In this prospective observational cohort study, we aimed to identify risk factors associated with a complicated infection in community-onset S. aureus bacteraemia. Potential risk factors were collected from electronic medical records and included: - patient demographics, symptomology, portal of entry, and laboratory results. Results We identified several potential risk factors using univariate analysis. In a multiple logistic regression model, age, haemodialysis, and entry point from a diabetic foot ulcer were all significantly protective against complications. Conversely, an unknown entry point of infection, an entry point from an indwelling medical device, and a C-reactive protein concentration of over 161 mg/L on the day of admission were all significantly associated with complications. Conclusions We conclude that several factors are associated with complications including already conducted laboratory investigations and portal of entry of infection. These factors could aid the triage of at-risk patients for complications of S. aureus bacteraemia.
Collapse
Affiliation(s)
- Carly L Botheras
- School of Medicine, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia. .,School of Medicine, Faculty of Health, Deakin University, Geelong, Australia.
| | - Steven J Bowe
- Deakin Biostatistics Unit Faculty of Health, Deakin University, Geelong, Australia
| | - Raquel Cowan
- Department of Infectious Diseases, Barwon Health, Geelong, Australia
| | - Eugene Athan
- School of Medicine, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia.,School of Medicine, Faculty of Health, Deakin University, Geelong, Australia.,Department of Infectious Diseases, Barwon Health, Geelong, Australia
| |
Collapse
|
37
|
Armentrout EI, Liu GY, Martins GA. T Cell Immunity and the Quest for Protective Vaccines against Staphylococcus aureus Infection. Microorganisms 2020; 8:microorganisms8121936. [PMID: 33291260 PMCID: PMC7762175 DOI: 10.3390/microorganisms8121936] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus is a wide-spread human pathogen, and one of the top causative agents of nosocomial infections. The prevalence of antibiotic-resistant S. aureus strains, which are associated with higher mortality and morbidity rates than antibiotic-susceptible strains, is increasing around the world. Vaccination would be an effective preventive measure against S. aureus infection, but to date, every vaccine developed has failed in clinical trials, despite inducing robust antibody responses. These results suggest that induction of humoral immunity does not suffice to confer protection against the infection. Evidence from studies in murine models and in patients with immune defects support a role of T cell-mediated immunity in protective responses against S. aureus. Here, we review the current understanding of the mechanisms underlying adaptive immunity to S. aureus infections and discuss these findings in light of the recent S. aureus vaccine trial failures. We make the case for the need to develop anti-S. aureus vaccines that can specifically elicit robust and durable protective memory T cell subsets.
Collapse
Affiliation(s)
- Erin I. Armentrout
- Lung Institute, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA;
- Division of Pulmonary and Critical Care Medicine, CSMC, Los Angeles, CA 90048, USA
| | - George Y. Liu
- Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92161, USA;
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gislâine A. Martins
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute (IBIRI), CSMC, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Research Division of Immunology, CSMC, Los Angeles, CA 90048, USA
- Department of Medicine, Division of Gastroenterology, CSMC, Los Angeles, CA 90048, USA
- Correspondence:
| |
Collapse
|
38
|
Belo VA, Pereira JA, Souza SFD, Tana FDL, Pereira BP, Lopes DDO, Ceron CS, Novaes RD, Corsetti PP, de Almeida LA. The role of IL-10 in immune responses against Pseudomonas aeruginosa during acute lung infection. Cell Tissue Res 2020; 383:1123-1133. [PMID: 33165659 DOI: 10.1007/s00441-020-03308-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 09/22/2020] [Indexed: 11/28/2022]
Abstract
Pseudomonas aeruginosa is considered an opportunistic pathogen of great clinical importance. The clearance of this bacterium occurs through recognition of the pathogen by innate immune system receptors, leading to a lung inflammatory response. However, this response must be controlled via immunoregulatory pathways. In this study, we evaluate the role of endogenous murine IL-10 after acute infection with the virulent strain P. aeruginosa PA14. To assess the role of IL-10, intratracheal infection with the PA14 strain was performed in C57BL/6 or IL-10 KO mice. The PA14 strain was recovered in both types of animals, although IL-10 KO mice presented a higher number of viable bacteria in the lung when compared to the C57BL/6 group. Histopathological and stereological analyses showed that IL-10 KO mice had higher tissue damage and inflammatory infiltrate when compared to control animals. The activity of MMP-9 but not MMP-2, as well as IL-6 and TNF-α expression, were augmented in the lungs of infected animals and was much more evident in IL-10 KO animals when compared to the other analyzed groups. This work indicates that endogenous IL-10 control P. aeruginosa infection, the expression of pro-inflammatory genes, MMP-9 activity and histopathological processes of the infectious process in question.
Collapse
Affiliation(s)
- Valéria Aparecida Belo
- Departmento de Microbiologia E Imunologia, Universidade Federal de Alfenas, Alfenas, Brazil
| | - Jéssica Assis Pereira
- Departmento de Microbiologia E Imunologia, Universidade Federal de Alfenas, Alfenas, Brazil
| | - Sara Franchin D Souza
- Departmento de Microbiologia E Imunologia, Universidade Federal de Alfenas, Alfenas, Brazil
| | - Fernanda de Lima Tana
- Departmento de Microbiologia E Imunologia, Universidade Federal de Alfenas, Alfenas, Brazil
| | - Bruna P Pereira
- Faculdade de Ciências Farmacêuticas, Universidade Federal de Alfenas, Alfenas, Brazil
| | - Débora de Oliveira Lopes
- Laboratório de Biologia Molecular, Universidade Federal de São João Del-Rei (CCO), Divinópolis, Brazil
| | - Carla S Ceron
- Faculdade de Ciências Farmacêuticas, Universidade Federal de Alfenas, Alfenas, Brazil
| | - Rômulo D Novaes
- Departamento de Biologia Estrutural, Universidade Federal de Alfenas, Alfenas, Brazil
| | - Patrícia Paiva Corsetti
- Laboratório de Imunologia das Doenças Infecciosas E Crônicas, Universidade José Do Rosário Vellano, Alfenas, Minas Gerais, Brazil
| | | |
Collapse
|
39
|
Wozniak JM, Mills RH, Olson J, Caldera JR, Sepich-Poore GD, Carrillo-Terrazas M, Tsai CM, Vargas F, Knight R, Dorrestein PC, Liu GY, Nizet V, Sakoulas G, Rose W, Gonzalez DJ. Mortality Risk Profiling of Staphylococcus aureus Bacteremia by Multi-omic Serum Analysis Reveals Early Predictive and Pathogenic Signatures. Cell 2020; 182:1311-1327.e14. [PMID: 32888495 PMCID: PMC7494005 DOI: 10.1016/j.cell.2020.07.040] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/11/2020] [Accepted: 07/29/2020] [Indexed: 12/15/2022]
Abstract
Staphylococcus aureus bacteremia (SaB) causes significant disease in humans, carrying mortality rates of ∼25%. The ability to rapidly predict SaB patient responses and guide personalized treatment regimens could reduce mortality. Here, we present a resource of SaB prognostic biomarkers. Integrating proteomic and metabolomic techniques enabled the identification of >10,000 features from >200 serum samples collected upon clinical presentation. We interrogated the complexity of serum using multiple computational strategies, which provided a comprehensive view of the early host response to infection. Our biomarkers exceed the predictive capabilities of those previously reported, particularly when used in combination. Last, we validated the biological contribution of mortality-associated pathways using a murine model of SaB. Our findings represent a starting point for the development of a prognostic test for identifying high-risk patients at a time early enough to trigger intensive monitoring and interventions.
Collapse
Affiliation(s)
- Jacob M Wozniak
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robert H Mills
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joshua Olson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - J R Caldera
- Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gregory D Sepich-Poore
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Marvic Carrillo-Terrazas
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chih-Ming Tsai
- Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Fernando Vargas
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Rob Knight
- Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA
| | - George Y Liu
- Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Victor Nizet
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - George Sakoulas
- Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Warren Rose
- School of Pharmacy, School of Medicine and Public Health University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Medicine, School of Medicine and Public Health University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David J Gonzalez
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
40
|
Lactate production by Staphylococcus aureus biofilm inhibits HDAC11 to reprogramme the host immune response during persistent infection. Nat Microbiol 2020; 5:1271-1284. [PMID: 32661313 PMCID: PMC7529909 DOI: 10.1038/s41564-020-0756-3] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 06/16/2020] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus (S. aureus) is a leading cause of biofilm-associated prosthetic joint infection (PJI), resulting in significant disability and prolonged treatment. It is known that host leukocyte IL-10 production is required for S. aureus biofilm persistence in PJI. A S. aureus bursa aurealis Tn library consisting of 1,952 non-essential genes was screened for mutants that failed to induce IL-10 in myeloid-derived suppressor cells (MDSCs), which identified a critical role for bacterial lactic acid biosynthesis. We generated a S. aureus ddh/ldh1/ldh2 triple Tn mutant that cannot produce D- or L-lactate. Co-culture of MDSCs or macrophages with ddh/ldh1/ldh2 mutant biofilm produced substantially less IL-10 compared with wild type S. aureus, which was also observed in a mouse model of PJI and led to reduced biofilm burden. Using MDSCs recovered from the mouse PJI model and in vitro leukocyte-biofilm co-cultures we show that bacterial-derived lactate inhibits histone deacetylase 11 (HDAC11), causing unchecked HDAC6 activity and increased histone 3 acetylation at the Il-10 promoter, resulting in enhanced Il-10 transcription in MDSCs and macrophages. Finally, we show that synovial fluid of patients with PJI contains elevated amounts of D-lactate and IL-10 compared with control subjects, and bacterial lactate increases IL-10 production by human monocyte-derived macrophages. Biofilms are bacterial communities that are difficult to treat because of their tolerance to antibiotics and ability to evade immune-mediated clearance. Prosthetic joint infection (PJI), a devastating complication of arthroplasty, is characterized by biofilm formation. The current study has discovered a central role for lactic acid biosynthesis in S. aureus biofilm formation during PJI. Mechanistically, bacterial-derived lactate inhibits histone deacetylase 11 (HDAC11) activity, which causes extensive epigenetic changes at the promoters of numerous host genes, including the key anti-inflammatory cytokine Il-10. Indeed, IL-10 production by myeloid-derived suppressor cells (MDSCs) and macrophages is critical for biofilm persistence during PJI. HDAC11 inhibition by S. aureus lactate results in unchecked HDAC6 activity, a positive regulator of IL-10, thereby increasing IL-10 production by MDSCs and macrophages in vitro and in vivo. Similarly, S. aureus lactate promotes IL-10 production in human monocyte-derived macrophages following biofilm exposure. This study highlights how bacterial metabolism can influence the host immune response to promote infection persistence.
Collapse
|
41
|
Guimaraes AO, Cao Y, Hong K, Mayba O, Peck MC, Gutierrez J, Ruffin F, Carrasco-Triguero M, Dinoso JB, Clemenzi-Allen A, Koss CA, Maskarinec SA, Chambers HF, Fowler VG, Baruch A, Rosenberger CM. A Prognostic Model of Persistent Bacteremia and Mortality in Complicated Staphylococcus aureus Bloodstream Infection. Clin Infect Dis 2020; 68:1502-1511. [PMID: 30165412 DOI: 10.1093/cid/ciy739] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/23/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Staphylococcus aureus is a leading cause of bacteremia, yet there remains a significant knowledge gap in the identification of relevant biomarkers that predict clinical outcomes. Heterogeneity in the host response to invasive S. aureus infection suggests that specific biomarker signatures could be utilized to differentiate patients prone to severe disease, thereby facilitating earlier implementation of more aggressive therapies. METHODS To further elucidate the inflammatory correlates of poor clinical outcomes in patients with S. aureus bacteremia, we evaluated the association between a panel of blood proteins at initial presentation of bacteremia and disease severity outcomes using 2 cohorts of patients with S. aureus bacteremia (n = 32 and n = 124). RESULTS We identified 13 candidate proteins that were correlated with mortality and persistent bacteremia. Prognostic modeling identified interleukin (IL)-8 and CCL2 as the strongest individual predictors of mortality, with the combination of these biomarkers classifying fatal outcome with 89% sensitivity and 77% specificity (P < .0001). Baseline IL-17A levels were elevated in patients with persistent bacteremia (P < .0001), endovascular (P = .026) and metastatic tissue infections (P = .012). CONCLUSIONS These results demonstrate the potential utility of selected biomarkers to distinguish patients with the highest risk for treatment failure and bacteremia-related complications, providing a valuable tool for clinicians in the management of S. aureus bacteremia. Additionally, these biomarkers could identify patients with the greatest potential to benefit from novel therapies in clinical trials.
Collapse
Affiliation(s)
| | - Yi Cao
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Kyu Hong
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Oleg Mayba
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Melicent C Peck
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Johnny Gutierrez
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Felicia Ruffin
- Division of Infectious Diseases, Duke University, Durham, North Carolina
| | | | - Jason B Dinoso
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Angelo Clemenzi-Allen
- Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco
| | - Catherine A Koss
- Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco
| | | | - Henry F Chambers
- Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco
| | - Vance G Fowler
- Division of Infectious Diseases, Duke University, Durham, North Carolina
| | - Amos Baruch
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | | |
Collapse
|
42
|
Jorch SK, Surewaard BG, Hossain M, Peiseler M, Deppermann C, Deng J, Bogoslowski A, van der Wal F, Omri A, Hickey MJ, Kubes P. Peritoneal GATA6+ macrophages function as a portal for Staphylococcus aureus dissemination. J Clin Invest 2020; 129:4643-4656. [PMID: 31545300 DOI: 10.1172/jci127286] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 08/01/2019] [Indexed: 12/20/2022] Open
Abstract
Essentially all Staphylococcus aureus (S. aureus) bacteria that gain access to the circulation are plucked out of the bloodstream by the intravascular macrophages of the liver - the Kupffer cells. It is also thought that these bacteria are disseminated via the bloodstream to other organs. Our data show that S. aureus inside Kupffer cells grew and escaped across the mesothelium into the peritoneal cavity and immediately infected GATA-binding factor 6-positive (GATA6+) peritoneal cavity macrophages. These macrophages provided a haven for S. aureus, thereby delaying the neutrophilic response in the peritoneum by 48 hours and allowing dissemination to various peritoneal and retroperitoneal organs including the kidneys. In mice deficient in GATA6+ peritoneal macrophages, neutrophils infiltrated more robustly and reduced S. aureus dissemination. Antibiotics administered i.v. did not prevent dissemination into the peritoneum or to the kidneys, whereas peritoneal administration of vancomycin (particularly liposomal vancomycin with optimized intracellular penetrance capacity) reduced kidney infection and mortality, even when administered 24 hours after infection. These data indicate that GATA6+ macrophages within the peritoneal cavity are a conduit of dissemination for i.v. S. aureus, and changing the route of antibiotic delivery could provide a more effective treatment for patients with peritonitis-associated bacterial sepsis.
Collapse
Affiliation(s)
- Selina K Jorch
- Department of Physiology and Pharmacology, Immunology Research Group, and Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Bas Gj Surewaard
- Department of Physiology and Pharmacology, Immunology Research Group, and Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mokarram Hossain
- Department of Physiology and Pharmacology, Immunology Research Group, and Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Moritz Peiseler
- Department of Physiology and Pharmacology, Immunology Research Group, and Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Carsten Deppermann
- Department of Physiology and Pharmacology, Immunology Research Group, and Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jennifer Deng
- Department of Physiology and Pharmacology, Immunology Research Group, and Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ania Bogoslowski
- Department of Physiology and Pharmacology, Immunology Research Group, and Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Fardau van der Wal
- Department of Physiology and Pharmacology, Immunology Research Group, and Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Abdelwahab Omri
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Melbourne, Victoria, Australia
| | - Paul Kubes
- Department of Physiology and Pharmacology, Immunology Research Group, and Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
43
|
Naik P, Singh S, Vishwakarma S, Kaur I, Dave VP, Kumar A, Joseph J. Multidrug-Resistant Pseudomonas aeruginosa Evokes Differential Inflammatory Responses in Human Microglial and Retinal Pigment Epithelial Cells. Microorganisms 2020; 8:735. [PMID: 32423093 PMCID: PMC7285153 DOI: 10.3390/microorganisms8050735] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Increasing incidences of multidrug-resistant (MDR) pathogens causing endophthalmitis threaten our ability to treat this condition, and the modulation of inflammatory responses by MDR bacteria is not known. In this study, using human microglia and retinal pigment epithelial (RPE) cells, we compare the inflammatory responses of sensitive (S-PA) and multidrug-resistant (MDR-PA) clinical isolates of Pseudomonas aeruginosa. Infected cells were subjected to qPCR analysis, enzyme-linked immunosorbent assay (ELISA), and immunostaining to assess the expression of inflammatory mediators. Both microglia and RPE cells, challenged with S-PA and MDR-PA, induced a time-dependent expression of inflammatory cytokines. Significant differences were observed in expression levels of Toll-like receptors (TLR) TLR4, TLR5, and TLR9 in microglia cells challenged with MDR-PA vs. S-PA. Similarly, mRNA levels of interleukin (IL)-6, tumor necrosis factor (TNF)-α, Interferon (IFN)-γ, and matrix metalloproteinase (MMP)-9 were also higher in MDR-PA-infected cells. At protein levels, upregulation was observed for IL-10 (p = 0.004), IL-8 (p = 0.0006), IL-1β (p = 0.02), and Granulocyte-macrophage colony-stimulating factor (GM-CSF) (p = 0.0006) in cells infected MDR-PA versus S-PA in both microglia and RPE cells; however, the response was delayed in RPE cells. Heatmap and STRING analysis highlighted the existence of a cross-talk between the inflammatory and cytokine-mediated signaling pathways. Our study highlights a differential inflammatory response evoked by MDR vs. sensitive pathogens in retinal cells during endophthalmitis.
Collapse
Affiliation(s)
- Poonam Naik
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana 500034, India;
- Manipal Academy of Higher Education, Manipal, Karnataka 576104, India;
| | - Sukhvinder Singh
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, Michigan, MI 48201, USA; (S.S.); (A.K.)
| | - Sushma Vishwakarma
- Manipal Academy of Higher Education, Manipal, Karnataka 576104, India;
- Kallam Anji Reddy Molecular Genetics laboratory, Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana 500034, India;
| | - Inderjeet Kaur
- Kallam Anji Reddy Molecular Genetics laboratory, Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana 500034, India;
| | - Vivek Pravin Dave
- Smt. Kannuri Santhamma Centre for vitreoretinal diseases. L V Prasad Eye Institute, Hyderabad, Telangana 500034, India;
| | - Ashok Kumar
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, Michigan, MI 48201, USA; (S.S.); (A.K.)
| | - Joveeta Joseph
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana 500034, India;
| |
Collapse
|
44
|
Kelly AM, McLoughlin RM. Target the Host, Kill the Bug; Targeting Host Respiratory Immunosuppressive Responses as a Novel Strategy to Improve Bacterial Clearance During Lung Infection. Front Immunol 2020; 11:767. [PMID: 32425944 PMCID: PMC7203494 DOI: 10.3389/fimmu.2020.00767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022] Open
Abstract
The lung is under constant pressure to protect the body from invading bacteria. An effective inflammatory immune response must be tightly orchestrated to ensure complete clearance of any invading bacteria, while simultaneously ensuring that inflammation is kept under strict control to preserve lung viability. Chronic bacterial lung infections are seen as a major threat to human life with the treatment of these infections becoming more arduous as the prevalence of antibiotic resistance becomes increasingly commonplace. In order to survive within the lung bacteria target the host immune system to prevent eradication. Many bacteria directly target inflammatory cells and cytokines to impair inflammatory responses. However, bacteria also have the capacity to take advantage of and strongly promote anti-inflammatory immune responses in the host lung to inhibit local pro-inflammatory responses that are critical to bacterial elimination. Host cells such as T regulatory cells and myeloid-derived suppressor cells are often enhanced in number and activity during chronic pulmonary infection. By increasing suppressive cell populations and cytokines, bacteria promote a permissive environment suitable for their prolonged survival. This review will explore the anti-inflammatory aspects of the lung immune system that are targeted by bacteria and how bacterial-induced immunosuppression could be inhibited through the use of host-directed therapies to improve treatment options for chronic lung infections.
Collapse
Affiliation(s)
- Alanna M Kelly
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rachel M McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
45
|
Wu Y, Hu Y, Wang B, Li S, Ma C, Liu X, Moynagh PN, Zhou J, Yang S. Dopamine Uses the DRD5-ARRB2-PP2A Signaling Axis to Block the TRAF6-Mediated NF-κB Pathway and Suppress Systemic Inflammation. Mol Cell 2020; 78:42-56.e6. [DOI: 10.1016/j.molcel.2020.01.022] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/10/2019] [Accepted: 01/16/2020] [Indexed: 01/11/2023]
|
46
|
Huon JF, Gaborit B, Caillon J, Boutoille D, Navas D. A murine model of Staphylococcus aureus infected chronic diabetic wound: A new tool to develop alternative therapeutics. Wound Repair Regen 2020; 28:400-408. [PMID: 32134548 DOI: 10.1111/wrr.12802] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 02/05/2020] [Accepted: 02/11/2020] [Indexed: 12/11/2022]
Abstract
Diabetic wound infection is a frequent complication that may result in limb amputation. To develop new treatment strategies in response to increasing bacterial resistance, animal models are needed. We created a diabetic mouse model with chronically infected wounds. Diabetes was induced using streptozotocin, and wounds were performed using a biopsy punch, and then infected with a clinical strain of Staphylococcus aureus. Chronification was reached by delaying healing thanks to chemical products (aminotriazole and mercaptosuccinic acid). Overall survival, as well as clinical, bacteriological and immunological data in skin, blood and spleens were collected at days 1, 7, and 14 after wounding. After a transient bacteremia proved by bacteria presence in spleen and kidneys in the first days after wounding, infected mice showed a chronic infection, with a bioburden impairing the healing process, and bacteria persistence compared to control mice. Infected mice showed gradual increasing skin levels of IL-17A compared to control mice that resulted in an IL-17/IFN-γ inbalance, pointing out a localized Th17 polarization of the immune response. Whether infected or not, the skin level of IL-10 decreased dramatically at days 1 and 7 after wounding, with an increase observed only in the control mice at day 14. After a decrease at day 1 in both groups, spleen IL-10 showed a rather steady level at days 7 and 14 in the control group compared with the decrease observed in the infected group. The spleen IL-10/IFN-γ ratio showed a systemic inflammatory response with Th1 polarization. Therefore, this model provides useful data to study wound healing. It is easy to reproduce, affordable and offers clinical and biological tools to evaluate new therapeutics.
Collapse
Affiliation(s)
- Jean-François Huon
- CHU Nantes, Clinical Pharmacy Unit, Nantes, France.,Nantes University, Laboratory of Clinical and Experimental Therapeutics of Infections, Nantes, France
| | - Benjamin Gaborit
- Nantes University, Laboratory of Clinical and Experimental Therapeutics of Infections, Nantes, France.,CHU Nantes, Infectious Disease Department, Nantes, France
| | - Jocelyne Caillon
- Nantes University, Laboratory of Clinical and Experimental Therapeutics of Infections, Nantes, France.,CHU Nantes, Microbiology Laboratory, Nantes, France
| | - David Boutoille
- Nantes University, Laboratory of Clinical and Experimental Therapeutics of Infections, Nantes, France.,CHU Nantes, Infectious Disease Department, Nantes, France
| | - Dominique Navas
- CHU Nantes, Clinical Pharmacy Unit, Nantes, France.,Nantes University, Laboratory of Clinical and Experimental Therapeutics of Infections, Nantes, France
| |
Collapse
|
47
|
Flores-Kossack C, Montero R, Köllner B, Maisey K. Chilean aquaculture and the new challenges: Pathogens, immune response, vaccination and fish diversification. FISH & SHELLFISH IMMUNOLOGY 2020; 98:52-67. [PMID: 31899356 DOI: 10.1016/j.fsi.2019.12.093] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 11/29/2019] [Accepted: 12/30/2019] [Indexed: 06/10/2023]
Abstract
In Chile, the salmon and trout farmed fishing industries have rapidly grown during the last years, becoming one of the most important economic sources for the country. However, infectious diseases caused by bacteria, virus, mycoses and parasites, result in losses of up to 700 million dollars per year for the Chilean aquaculture production with the consequent increase of antibiotic and antiparasitic usage. After 30 years of its first appearance, the main salmon health problem is still the salmonid rickettsial septicaemia (SRS), which together with other disease outbreaks, reveal that vaccines do not provide acceptable levels of long-lasting immune protection in the field. On the other hand, due to the large dependence of the industry on salmonids production, the Chilean government promoted the Aquaculture diversification program by 2009, which includes new species such as Merluccius australis, Cilus gilberti and Genypterus chilensis, however, specific research regarding the immune system and vaccine development are issues that still need to be addressed and must be considered as important as the farm production technologies for new fish species. Based on the experience acquired from the salmonid fish farming, should be mandatory an effort to study the immune system of the new species to develop knowledge for vaccination approaches, aiming to protect these aquaculture species before diseases outbreaks may occur. This review focuses on the current status of the Chilean aquaculture industry, the challenges related to emerging and re-emerging microbial pathogens on salmonid fish farming, and the resulting needs in the development of immune protection by rational designed vaccines. We also discussed about what we have learn from 25 years of salmonid researches and what can be applied to the new Chilean farmed species on immunology and vaccinology.
Collapse
Affiliation(s)
- C Flores-Kossack
- Laboratorio de Inmunología Comparativa, Centro de Biotecnología Acuícola (CBA), Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile
| | - R Montero
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493, Greifswald-Insel Riems, Germany
| | - B Köllner
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493, Greifswald-Insel Riems, Germany
| | - K Maisey
- Laboratorio de Inmunología Comparativa, Centro de Biotecnología Acuícola (CBA), Universidad de Santiago de Chile, Alameda, 3363, Santiago, Chile.
| |
Collapse
|
48
|
Salazar A, Nieto JE, Velazquez-Soto H, Jiménez-Martínez MC. Activation of IL-10+ B cells: A novel immunomodulatory mechanism for therapeutic bacterial suspensions. SAGE Open Med 2020; 8:2050312120901547. [PMID: 32002185 PMCID: PMC6963315 DOI: 10.1177/2050312120901547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 12/23/2019] [Indexed: 01/22/2023] Open
Abstract
Objectives: Bacterial components are used to improve immune responses in patients with respiratory infections. Pharmacological formulations of bacterial components include a mixture of bacterial antigens, some of which are complete inactivated bacteria, that is, named bacterial suspensions; while others are fragments of bacteria, which are presented as bacterial lysates. Although bacterial lysates have been broadly used as immune-stimulators, the biological support for the therapeutic effectiveness of bacterial suspension has not yet been studied. Thus, the aim of our study was to investigate the immunological activity induced by bacterial suspension. Methods: This work was an exploratory translational study. Peripheral blood mononuclear cells were obtained from healthy donors and cultured in time–dose dependent assays with a commercial bacterial suspension. Flow cytometry was used for phenotypic analysis and for determining soluble cytokines in culture supernatants. Results: We observed that bacterial suspension activates B cells in a dose-dependent manner. Peripheral blood mononuclear cells were able to secrete IL-6 and IL-10 after 24 h of bacterial suspension stimulation. TLR2 expression was observed mainly on CD19+ CD38Lo B cells after 72 h of culture; remarkably, most of the TLR2+ CD19+ cells were also IL-10+. Conclusion: Our findings suggest that bacterial suspension induces the activation of B cell subsets as well as the secretion of IL-6 and IL-10. Expression of TLR2 on CD19+ cells could act as an activation loop of IL-10+ B regulatory cells. The clinical implications of these findings are discussed at the end of this article.
Collapse
Affiliation(s)
- Alberto Salazar
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico.,Department of Immunology and Research Unit, Institute of Ophthalmology "Conde de Valenciana," Mexico City, Mexico
| | - Jane E Nieto
- Department of Immunology and Research Unit, Institute of Ophthalmology "Conde de Valenciana," Mexico City, Mexico
| | - Henry Velazquez-Soto
- Department of Immunology and Research Unit, Institute of Ophthalmology "Conde de Valenciana," Mexico City, Mexico
| | - Maria C Jiménez-Martínez
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico.,Department of Immunology and Research Unit, Institute of Ophthalmology "Conde de Valenciana," Mexico City, Mexico
| |
Collapse
|
49
|
Cruciani M, Sandini S, Etna MP, Giacomini E, Camilli R, Severa M, Rizzo F, Bagnoli F, Hiscott J, Coccia EM. Differential Responses of Human Dendritic Cells to Live or Inactivated Staphylococcus aureus: Impact on Cytokine Production and T Helper Expansion. Front Immunol 2019; 10:2622. [PMID: 31781115 PMCID: PMC6861420 DOI: 10.3389/fimmu.2019.02622] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/22/2019] [Indexed: 12/11/2022] Open
Abstract
Understanding Staphylococcus aureus (S. aureus)-host immune system interaction is crucial to meet the tremendous medical need associated with this life-threatening bacterial infection. Given the crucial role of dendritic cells (DC) in dictating immune responses upon microbial challenge, we investigated how the bacterial viability and the conservation of structural integrity influence the response of human DC to S. aureus. To this end, human primary DC were stimulated with the methicillin-resistant S. aureus USA300 live strain, USA300 inactivated by heat (HI), ultraviolet irradiation (UVI), or paraformaldehyde treatment (PFAI) and subsequently analyzed for cell phenotype and immune-modulatory properties. Although no differences in terms of DC viability and maturation were observed when DC were stimulated with live or inactivated bacteria, the production of IL-12, IL-23, and other cytokines differed significantly. The Th1 and Th17 expansion was also more pronounced in response to live vs. inactivated S. aureus. Interestingly, cytokine production in DC treated with live and inactivated USA300 required phagocytosis, whereas blocking endosomal Toll-like receptor signaling mainly reduced the cytokine release by live and HI USA300. A further analysis of IFN-β signaling revealed the induction of a cyclic GMP-AMP synthase stimulator of interferon genes (cGAS-STING)-independent and IRF3-dependent signaling pathway(s) in UVI-stimulated DC. This study underscores the capacity of human DC to discriminate between live and inactivated S. aureus and, further, indicates that DC may represent a valuable experimental setting to test different inactivation methods with regard to the retention of S. aureus immunoregulatory properties. These and further insights may be useful for the development of novel therapeutic and prophylactic anti-S. aureus vaccine strategies.
Collapse
Affiliation(s)
- Melania Cruciani
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Silvia Sandini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Marilena P Etna
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Giacomini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Romina Camilli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Martina Severa
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Fabiana Rizzo
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - John Hiscott
- Pasteur Laboratory, Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Eliana M Coccia
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
50
|
Genetic variation of DNA methyltransferase-3A contributes to protection against persistent MRSA bacteremia in patients. Proc Natl Acad Sci U S A 2019; 116:20087-20096. [PMID: 31527248 PMCID: PMC6778225 DOI: 10.1073/pnas.1909849116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The severity and duration of methicillin-resistant Staphylococcus aureus (MRSA) bacteremia varies widely between individuals. Host factors predisposing to persistent MRSA bacteremia are poorly understood, although genetic association studies are beginning to identify potentially influential variants. We found an association between the A/C heterozygous genotype in the DNMT3A correlating with shorter time to resolution of MRSA bacteremia. Using in vitro macrophage assays and murine sepsis models, we demonstrated that DNMT3A variants may alter host response to infection through increased methylation of key regulatory genes, resulting in reduced interleukin-10 production and in turn, allowing for a more protective immune response that clears infection. An improved understanding of the factors predisposing to persistent MRSA bacteremia may help to discover better treatment options. The role of the host in development of persistent methicillin-resistant Staphylococcus aureus (MRSA) bacteremia is not well understood. A cohort of prospectively enrolled patients with persistent methicillin-resistant S. aureus bacteremia (PB) and resolving methicillin-resistant S. aureus bacteremia (RB) matched by sex, age, race, hemodialysis status, diabetes mellitus, and presence of implantable medical device was studied to gain insights into this question. One heterozygous g.25498283A > C polymorphism located in the DNMT3A intronic region of chromosome 2p with no impact in messenger RNA (mRNA) expression was more common in RB (21 of 34, 61.8%) than PB (3 of 34, 8.8%) patients (P = 7.8 × 10−6). Patients with MRSA bacteremia and g.25498283A > C genotype exhibited significantly higher levels of methylation in gene-regulatory CpG island regions (Δmethylation = 4.1%, P < 0.0001) and significantly lower serum levels of interleukin-10 (IL-10) than patients with MRSA bacteremia without DNMT3A mutation (A/C: 9.7038 pg/mL vs. A/A: 52.9898 pg/mL; P = 0.0042). Expression of DNMT3A was significantly suppressed in patients with S. aureus bacteremia and in S. aureus-challenged primary human macrophages. Small interfering RNA (siRNA) silencing of DNMT3A expression in human macrophages caused increased IL-10 response upon S. aureus stimulation. Treating macrophages with methylation inhibitor 5-Aza-2′-deoxycytidine resulted in increased levels of IL-10 when challenged with S. aureus. In the murine sepsis model, methylation inhibition increased susceptibility to S. aureus. These findings indicate that g.25498283A > C genotype within DNMT3A contributes to increased capacity to resolve MRSA bacteremia, potentially through a mechanism involving increased methylation of gene-regulatory regions and reduced levels of antiinflammatory cytokine IL-10.
Collapse
|