1
|
Zhao J, Gong Z, Bao W, Liu X, Yu Z, Dong YQ, Mao W, Liu B, Zhang S. Matrine alleviates Staphylococcus aureus-induced acute lung injury in mice by inhibiting MLKL and NLRP3-mediated inflammatory activity. Eur J Pharmacol 2025; 993:177385. [PMID: 39956265 DOI: 10.1016/j.ejphar.2025.177385] [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: 10/11/2024] [Revised: 02/04/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
Acute lung injury (ALI) is a serious clinical condition with high incidence and mortality. The inflammatory response induced by gram-positive bacteria, especially Staphylococcus aureus (S. aureus), is a key factor contributing to ALI progression and other infectious diseases. Matrine, known for its diverse biological and pharmacological properties, has not been fully explored for its potential to prevent or treat S. aureus-induced ALI. Our study demonstrated that matrine exerts a protective effect against lung injury in mice infected with S. aureus. Specifically, matrine reduced pulmonary edema and decreased neutrophil infiltration in the infected lungs. Furthermore, matrine significantly reduced the expression of high-mobility group box protein 1 (HMGB1) and hyaluronic acid-binding protein 2 (HABP2) in the lungs of infected mice. Additionally, matrine modulated the production of inflammatory mediators, including tumor necrosis factor-α (TNF-α); interleukin-1β (IL-1β); regulated upon activation normal T cell expressed and secreted (RANTES) and Interleukin 10 (IL-10), in both infected lungs and macrophages, suggesting a protective role against tissue damage. Moreover, matrine influenced the secretion of proinflammatory cytokines and regulated the activation of the mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) signaling pathways, along with the activation of Nod-like receptor pyrin domain-containing protein 3 (NLRP3) and Mixed-lineage kinase domain-like protein (MLKL) in macrophages. Notably, when MLKL, but not NLRP3, is deleted, the ability of matrine to regulate damage-associated proteins and prevent tissue injury is diminished. These findings suggest that matrine may be a promising therapeutic agent for the prevention and treatment of ALI.
Collapse
Affiliation(s)
- Jiamin Zhao
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Zhiguo Gong
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Wenhui Bao
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Xinyu Liu
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Zhuoya Yu
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Yan Qin Dong
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Wei Mao
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Bo Liu
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China.
| | - Shuangyi Zhang
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, 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
|
Engelhart MJ, Brock OD, Till JM, Glowacki RWP, Cantwell JW, Clarke DJ, Wesener DA, Ahern PP. BT1549 coordinates the in vitro IL-10 inducing activity of Bacteroides thetaiotaomicron. Microbiol Spectr 2025; 13:e0166924. [PMID: 39868786 PMCID: PMC11878027 DOI: 10.1128/spectrum.01669-24] [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: 07/10/2024] [Accepted: 11/27/2024] [Indexed: 01/28/2025] Open
Abstract
The intestine is home to a complex immune system that is engaged in mutualistic interactions with the microbiome that maintain intestinal homeostasis. A variety of immune-derived anti-inflammatory mediators have been uncovered and shown to be critical for maintaining these beneficial immune-microbiome relationships. Notably, the gut microbiome actively invokes the induction of anti-inflammatory pathways that limit the development of microbiome-targeted inflammatory immune responses. Despite the importance of this microbiome-driven immunomodulation, detailed knowledge of the microbial factors that promote these responses remains limited. We have previously established that the gut symbiont Bacteroides thetaiotaomicron stimulates the production of the anti-inflammatory cytokine IL-10 via soluble factors in a Toll-like receptor 2 (TLR2)-MyD88-dependent manner. Here, using TLR2 activity reporter cell lines, we show that the capacity of B. thetaiotaomicron to stimulate TLR2 activity was not critically dependent on either of the canonical heterodimeric forms of TLR2, TLR2/TLR1, or TLR2/TLR6, that typically mediate its function. Furthermore, biochemical manipulation of B. thetaiotaomicron-conditioned media suggests that IL-10 induction is mediated by a protease-resistant or non-proteogenic factor. We next uncovered that deletion of gene BT1549, a predicted secreted lipoprotein, significantly impaired the capacity of B. thetaiotaomicron to induce IL-10, while complementation in trans restored IL-10 induction, suggesting a role for BT1549 in the immunomodulatory function of B. thetaiotaomicron. Collectively, these data provide molecular insight into the pathways through which B. thetaiotaomicron operates to promote intestinal immune tolerance and symbiosis. IMPORTANCE Intestinal homeostasis requires the establishment of peaceful interactions between the gut microbiome and the intestinal immune system. Members of the gut microbiome, like the symbiont Bacteroides thetaiotaomicron, actively induce anti-inflammatory immune responses to maintain mutualistic relationships with the host. Despite the importance of such interactions, the specific microbial factors responsible remain largely unknown. Here, we show that B. thetaiotaomicron, which stimulates Toll-like receptor 2 (TLR2) to drive IL-10 production, can stimulate TLR2 independently of TLR1 or TLR6, the two known TLR that can form heterodimers with TLR2 to mediate TLR2-dependent responses. Furthermore, we show that IL-10 induction is likely mediated by a protease-resistant or non-proteogenic factor, and that this requires gene BT1549, a predicted secreted lipoprotein and peptidase. Collectively, our work provides insight into the molecular dialog through which B. thetaiotaomicron coordinates anti-inflammatory immune responses. This knowledge may facilitate future strategies to promote such responses for therapeutic purposes.
Collapse
Affiliation(s)
- Morgan J. Engelhart
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Orion D. Brock
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jessica M. Till
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Robert W. P. Glowacki
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jason W. Cantwell
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - David J. Clarke
- School of Microbiology and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Darryl A. Wesener
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Philip P. Ahern
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
4
|
May KL, Grabowicz M. Outer membrane lipoproteins: late to the party, but the center of attention. J Bacteriol 2025; 207:e0044224. [PMID: 39670753 PMCID: PMC11784454 DOI: 10.1128/jb.00442-24] [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] [Indexed: 12/14/2024] Open
Abstract
An outer membrane (OM) is the hallmark feature that is often used to distinguish "Gram-negative" bacteria. Our understanding of how the OM is built rests largely on studies of Escherichia coli. In that organism-and seemingly in all species of the Proteobacterial phyla-the essential pathways that assemble the OM each rely on one or more lipoproteins that have been trafficked to the OM. Hence, the lipoprotein trafficking pathway appeared to be foundational for the ability of these bacteria to build their OM. However, such a notion now appears to be misguided. New phylogenetic analyses now show us that lipoprotein trafficking was likely the very last of the essential OM assembly systems to have evolved. The emergence of lipoprotein trafficking must have been a powerful innovation for the ancestors of Proteobacteria, given how it assumed such a central place in OM biogenesis. In this minireview, we broadly discuss the biosynthesis and trafficking of lipoproteins and ponder why the newest OM assembly system (lipoprotein trafficking) has become so key to building the Proteobacterial OM. We examine the diversity among lipoprotein trafficking systems, noting uniting commonalities and highlighting key differences. Current novel antibiotic development is targeted against a small subset of Proteobacterial species that cause severe human diseases; several inhibitors of lipoprotein biosynthesis and OM trafficking have been recently reported that may become new antibiotics. Understanding the diversity in lipoprotein trafficking may yield selective new antibiotics that preferentially kill important human pathogens while sparing species of normal healthy flora.
Collapse
Affiliation(s)
- Kerrie L. May
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Antibiotic Resistance Center, Emory University, Atlanta, Georgia, USA
| | - Marcin Grabowicz
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Antibiotic Resistance Center, Emory University, Atlanta, Georgia, USA
- Division of Infectious Disease, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
5
|
Schultz M, Hu Z, Deshmukh M, Henning P, Lerner UH, Mohammad M, Jin T. Focal Staphylococcus Aureus Septic Arthritis Elicits Age and TLR2-Dependent Periarticular Bone Loss. J Inflamm Res 2024; 17:11901-11913. [PMID: 39758941 PMCID: PMC11699840 DOI: 10.2147/jir.s479718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/05/2024] [Indexed: 01/07/2025] Open
Abstract
Introduction Septic arthritis, primarily caused by Staphylococcus aureus (S. aureus), is a severe joint infection that leads to joint and bone damage. S. aureus lipoproteins (LPPs) bind to Toll-like Receptor 2 (TLR2), inducing arthritis and localized bone loss. Aging affects TLR2 immune response to pathogens. While intra-articular injections of S. aureus LPPs induces local bone resorption in mice, the influence of aging and TLR2 expression on bone mineral density (BMD) after S. aureus bacteremia remains unclear. Methods We analyzed distal femoral BMD in young and old TLR2 knock-out and wild-type mice following intravenous S. aureus infection. BMD was measured in both total and trabecular bone in old and young mice to determine age and TLR2-dependent responses to infection. Results In non-infected mice, BMD in both total and trabecular bone was mainly age-related and TLR2-independent. Following S. aureus bacteremia, young wild-type mice with TLR2 expression showed decreased combined cortical and trabecular BMD. This effect was absent in aged mice or TLR2 deficient mice. Focal septic arthritis, induced by S. aureus bacteremia, emerged as the primary cause to bone loss in the femur metaphysis. TLR2 appears to play a crucial role in focal septic arthritis-induced bone loss, as evidenced by in vitro findings demonstrating that staphylococcal LPPs, known TLR2 agonists, increase the Tnfsf11/Tnfrsf11b ratio in mouse pariosteal osteoblasts. Conclusion S. aureus bacteremia triggers local bone loss in murine arthritis, depending on both age and TLR2 expression.
Collapse
Affiliation(s)
- Michelle Schultz
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Zhicheng Hu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Center for Clinical Laboratories, the Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
| | - Meghshree Deshmukh
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Petra Henning
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf H Lerner
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
6
|
Gong Z, Mao W, Ren P, Hao Z, Zhao J, Yu Z, Zhao Y, Feng Y, Liu B, Zhang S. Taurochenodeoxycholic acid ameliorates the Staphylococcus aureus infection-induced acute lung injury through toll-like receptor 2 in mice. Int Immunopharmacol 2024; 142:113228. [PMID: 39317054 DOI: 10.1016/j.intimp.2024.113228] [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: 05/07/2024] [Revised: 08/15/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
Acute lung injury (ALI) is a significant clinical problem associated with high morbidity and mortality. Inflammation induced by gram-positive bacterial pathogens, specifically Staphylococcus aureus (S. aureus), plays a major role in ALI development and other infectious diseases. Taurochenodeoxycholic acid (TCDCA) exhibits diverse biological activities and pharmacological effects. Nevertheless, the potential preventive and therapeutic effects of TCDCA and the underlying mechanism in the ALI induced by S. aureus infection remain poorly understood. Our results showed that the TCDCA (0.1 μg/g) had a beneficial effect on lung damage in mice infected with S. aureus. Specifically, TCDCA could lead to a reduction in pulmonary focal or diffuse oedema and a decrease in the infiltration of neutrophils in the S. aureus-infected lungs. We observed that TCDCA could significantly down-regulate the expression of HMGB1 in lung from S. aureus-infected mice. Furthermore, TCDCA could attenuate the production of inflammatory mediators in lungs and serum from S. aureus-infected mice. This finding further supported the notion that TCDCA potentially protects against tissue injury. In addition, TCDCA regulated the secretion of the proinflammatory cytokine, the activation of MAPK and NF-κB signaling pathways, and the activation of TLR2 in macrophages. Notably, TCDCA might reduce the secretion levels of inflammatory mediators and lung damage through the TLR2 in S. aureus-infected macrophages or mice. Altogether, TCDCA shows promise as a potential drug for preventing and treating ALI by modulating or inhibiting inflammatory mediators through TLR2.
Collapse
Affiliation(s)
- Zhiguo Gong
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Wei Mao
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Peipei Ren
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Zhichao Hao
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Jiamin Zhao
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Zhuoya Yu
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Yi Zhao
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Yaya Feng
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Bo Liu
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China.
| | - Shuangyi Zhang
- 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, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China.
| |
Collapse
|
7
|
Wang X, Lee JC. Staphylococcus aureus membrane vesicles: an evolving story. Trends Microbiol 2024; 32:1096-1105. [PMID: 38677977 PMCID: PMC11511790 DOI: 10.1016/j.tim.2024.04.003] [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: 12/15/2023] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/29/2024]
Abstract
Staphylococcus aureus is an important bacterial pathogen that causes a wide variety of human diseases in community and hospital settings. S. aureus employs a diverse array of virulence factors, both surface-associated and secreted, to promote colonization, infection, and immune evasion. Over the past decade, a growing body of research has shown that S. aureus generates extracellular membrane vesicles (MVs) that package a variety of bacterial components, many of which are virulence factors. In this review, we summarize recent advances in our understanding of S. aureus MVs and highlight their biogenesis, cargo, and potential role in the pathogenesis of staphylococcal infections. Lastly, we present some emerging questions in the field.
Collapse
Affiliation(s)
- Xiaogang Wang
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA.
| | - Jean C Lee
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
8
|
Rachwalski K, Madden SJ, Ritchie N, French S, Bhando T, Girgis-Gabardo A, Tu M, Gordzevich R, Ives R, Guo AB, Johnson JW, Xu Y, Kapadia SB, Magolan J, Brown ED. A screen for cell envelope stress uncovers an inhibitor of prolipoprotein diacylglyceryl transferase, Lgt, in Escherichia coli. iScience 2024; 27:110894. [PMID: 39376497 PMCID: PMC11456916 DOI: 10.1016/j.isci.2024.110894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/25/2024] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
The increasing prevalence of antibiotic resistance demands the discovery of antibacterial chemical scaffolds with unique mechanisms of action. Phenotypic screening approaches, such as the use of reporters for bacterial cell stress, offer promise to identify compounds while providing strong hypotheses for follow-on mechanism of action studies. From a collection of ∼1,800 Escherichia coli GFP transcriptional reporter strains, we identified a reporter that is highly induced by cell envelope stress-pProm rcsA -GFP. After characterizing pProm rcsA -GFP induction, we assessed a collection of bioactive small molecules for reporter induction, identifying 24 compounds of interest. Spontaneous suppressors to one compound in particular, MAC-0452936, mapped to the gene encoding the essential prolipoprotein diacylglyceryl transferase, lgt. Lgt inhibition by MAC-0452936 inhibition was confirmed through genetic, phenotypic, and biochemical approaches. The oxime ester, MAC-0452936, represents a useful small molecule inhibitor of Lgt and highlights the potential of using pProm rcsA -GFP as a phenotypic screening tool.
Collapse
Affiliation(s)
- Kenneth Rachwalski
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Sean J. Madden
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Nicole Ritchie
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Shawn French
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Timsy Bhando
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Adele Girgis-Gabardo
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Megan Tu
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Rodion Gordzevich
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Rowan Ives
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Amelia B.Y. Guo
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Jarrod W. Johnson
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Yiming Xu
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | | | - Jakob Magolan
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Eric D. Brown
- Institute of Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
9
|
Hook JS, Matheis AD, Kavanaugh JS, Horswill AR, Moreland JG. Role for IRAK-4 and p38 in Neutrophil Signaling in Response to Bacterial Lipoproteins from Staphylococcus aureus. Inflammation 2024:10.1007/s10753-024-02147-7. [PMID: 39302496 DOI: 10.1007/s10753-024-02147-7] [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: 07/12/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Neutrophils, polymorphonuclear leukocytes (PMN), express numerous pattern recognition receptors, including TLRs, capable of recognizing a wide variety of pathogens. Receptor engagement initiates a cascade of PMN responses with some occurring in seconds, and some requiring de novo protein synthesis over the course of many hours. Although numerous species of bacteria and bacterial products have been shown to activate PMN via TLRs, the signaling intermediates required for distinct PMN responses have not been well-defined in human PMN. Given the potential for host tissue damage by overexuberant PMN activity, a better understanding of neutrophil signaling is needed to generate effective therapies. We hypothesized that PMN responses to a lipoprotein-containing cell membrane preparation from methicillin-resistant S. aureus (MRSA-CMP) would activate signaling via IRAK4 and p38, with potentially distinct pathways for early vs. late responses. Using human PMN we investigated MRSA-CMP-elicited reactive oxygen species (ROS) production, elastase activity, NET formation, IL-8 production, and the role of IRAK4 and p38 activation. MRSA-CMP elicited ROS in a concentration and lipoprotein-dependent manner. MRSA-CMP elicited phosphorylation of p38 MAPK, and MRSA-CMP-elicited ROS production was partially dependent on p38 MAPK and IRAK4 activation. Inhibition of IRAK4 resulted in a reduction of p38 phosphorylation. MRSA-CMP-elicited elastase activity and NET formation was partially dependent on p38 MAPK activation, but independent of IRAK4 activation. MRSA-CMP-elicited IL-8 production required both p38 and IRAK4 activation. In conclusion, MRSA-CMP elicits PMN responses via distinct signaling pathways. There is potential to target components of the neutrophil inflammatory response without compromising critical pathogen-specific immune functions.
Collapse
Affiliation(s)
- Jessica S Hook
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Austin D Matheis
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey S Kavanaugh
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Veterans Affairs, Eastern Colorado Healthcare System, Aurora, CO, USA
| | - Jessica G Moreland
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Microbiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390-8504, USA.
| |
Collapse
|
10
|
Giese MA, Ramakrishnan G, Steenberge LH, Dovan JX, Sauer JD, Huttenlocher A. Staphylococcus aureus lipid factors modulate melanoma cell clustering and invasion. Dis Model Mech 2024; 17:dmm050770. [PMID: 39284707 PMCID: PMC11423913 DOI: 10.1242/dmm.050770] [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: 02/28/2024] [Accepted: 07/29/2024] [Indexed: 09/25/2024] Open
Abstract
The microbiome can influence cancer development and progression. However, less is known about the role of the skin microbiota in melanoma. Here, we took advantage of a zebrafish melanoma model to probe the effects of Staphylococcus aureus on melanoma invasion. We found that S. aureus produces factors that enhance melanoma invasion and dissemination in zebrafish larvae. We used a published in vitro 3D cluster formation assay that correlates increased clustering with tumor invasion. S. aureus supernatant increased clustering of melanoma cells and was abrogated by a Rho-Kinase inhibitor, implicating a role for Rho-GTPases. The melanoma clustering response was specific to S. aureus but not to other staphylococcal species, including S. epidermidis. Our findings suggest that S. aureus promotes melanoma clustering and invasion via lipids generated by the lipase Sal2 (officially known as GehB). Taken together, these findings suggest that specific bacterial products mediate melanoma invasive migration in zebrafish.
Collapse
Affiliation(s)
- Morgan A. Giese
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Gayathri Ramakrishnan
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA
- Cancer Biology Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Laura H. Steenberge
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Jerome X. Dovan
- University of Wisconsin Medical Scientist Training Program (MSTP) Summer Scholars, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53706, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
11
|
Odunitan TT, Apanisile BT, Akinboade MW, Abdulazeez WO, Oyaronbi AO, Ajayi TM, Oyekola SA, Ibrahim NO, Nafiu T, Afolabi HO, Olayiwola DM, David OT, Adeyemo SF, Ayodeji OD, Akinade EM, Saibu OA. Microbial mysteries: Staphylococcus aureus and the enigma of carcinogenesis. Microb Pathog 2024; 194:106831. [PMID: 39089512 DOI: 10.1016/j.micpath.2024.106831] [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: 12/30/2023] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Staphylococcus aureus, a common human pathogen, has long been the focus of scientific investigation due to its association with various infections. However, recent research has unveiled a tantalizing enigma surrounding this bacterium and its potential involvement in carcinogenesis. Chronic S. aureus infections have been linked to an elevated risk of certain cancers, including skin cancer and oral cancer. This review explores the current state of knowledge regarding this connection, examining epidemiological evidence, pathogenic mechanisms, and biological interactions that suggest a correlation. Although initial studies point to a possible link, the precise mechanisms through which S. aureus may contribute to cancer development remain elusive. Emerging evidence suggests that the chronic inflammation induced by persistent S. aureus infections may create a tumor-promoting environment. This inflammation can lead to DNA damage, disrupt cellular signaling pathways, and generate an immunosuppressive microenvironment conducive to cancer progression. Additionally, S. aureus produces a variety of toxins and metabolites that can directly interact with host cells, potentially inducing oncogenic transformations. Despite these insights, significant gaps remain in our understanding of the exact biological processes involved. This review emphasizes the urgent need for more comprehensive research to clarify these microbiological mysteries. Understanding the role of S. aureus in cancer development could lead to novel strategies for cancer prevention and treatment, potentially transforming therapeutic approaches.
Collapse
Affiliation(s)
- Tope T Odunitan
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria; Microbiology Unit, Helix Biogen Institute, Ogbomosho, Oyo State, Nigeria; Ehigie's Biochemistry and Biocomputational Laboratory, Ogbomosho, Oyo State, Nigeria.
| | - Boluwatife T Apanisile
- Department of Nutrition and Dietetics, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Modinat W Akinboade
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Waliu O Abdulazeez
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Adegboye O Oyaronbi
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Temitope M Ajayi
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Samuel A Oyekola
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Najahtulahi O Ibrahim
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Tawakalitu Nafiu
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Hezekiah O Afolabi
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Dolapo M Olayiwola
- Department of Medical Laboratory Science, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Oladunni T David
- Microbiology Unit, Helix Biogen Institute, Ogbomosho, Oyo State, Nigeria
| | - Stephen F Adeyemo
- Department of Biological Sciences, First Technical University, Ibadan, Oyo State, Nigeria; Division of Medical Artificial Intelligence, Helix Biogen Institute, Ogbomosho, Oyo State, Nigeria
| | - Oluwatobi D Ayodeji
- Department of Nursing, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Esther M Akinade
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Oluwatosin A Saibu
- Department of Chemistry and Biochemistry, New Mexico State University, USA
| |
Collapse
|
12
|
Payen S, Roy D, Okura M, Segura M, Gottschalk M. Study of the Role of Lipoprotein Maturation Enzymes in the Pathogenesis of the Infection Caused by the Streptococcus suis Serotype 2 Sequence Type 25 North American Prototype Strain. Pathogens 2023; 12:1325. [PMID: 38003790 PMCID: PMC10675726 DOI: 10.3390/pathogens12111325] [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: 08/29/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Streptococcus suis serotype 2 is an important swine bacterial pathogen causing sudden death, septic shock, and meningitis. However, serotype 2 strains are phenotypically and genotypically heterogeneous and composed of a multitude of sequence types (STs) whose distributions greatly vary worldwide. It has been previously shown that the lipoprotein (LPP) maturation enzymes diacylglyceryl transferase (Lgt) and signal peptidase (Lsp) significantly modulate the inflammatory host response and play a differential role in virulence depending on the genetic background of the strain. Differently from Eurasian ST1/ST7 strains, the capsular polysaccharide of a North American S. suis serotype 2 ST25 representative strain only partially masks sub-capsular domains and bacterial wall components. Thus, our hypothesis is that since LPPs would be more surface exposed in ST25 strains than in their ST1 or ST7 counterparts, the maturation enzymes would play a more important role in the pathogenesis of the infection caused by the North American strain. Using isogenic Δlgt and Δlsp mutants derived from the wild-type ST25 strain, our studies suggest that these enzymes do not seem to play a role in the interaction between S. suis and epithelial and endothelial cells, regardless of the genetics background of the strain used. However, a role in the formation of biofilms (also independently of the STs) has been demonstrated. Moreover, the involvement of LPP dendritic cell activation in vitro seems to be somehow more pronounced with the ST25 strain. Finally, the Lgt enzyme seems to play a more important role in the virulence of the ST25 strain. Although some differences between STs could be observed, our original hypothesis that LPPs would be significantly more important in ST25 strains due to a better bacterial surface exposition could not be confirmed.
Collapse
Affiliation(s)
- Servane Payen
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (M.S.)
| | - David Roy
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Masatoshi Okura
- Division of Transboundary Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Kagoshima 891-0105, Japan;
| | - Mariela Segura
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (M.S.)
| | - Marcelo Gottschalk
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (M.S.)
| |
Collapse
|
13
|
Xu Y, Xie C, Liu Y, Qin X, Liu J. An update on our understanding of Gram-positive bacterial membrane vesicles: discovery, functions, and applications. Front Cell Infect Microbiol 2023; 13:1273813. [PMID: 37860067 PMCID: PMC10582989 DOI: 10.3389/fcimb.2023.1273813] [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: 08/07/2023] [Accepted: 09/19/2023] [Indexed: 10/21/2023] Open
Abstract
Extracellular vesicles (EVs) are nano-sized particles released from cells into the extracellular environment, and are separated from eukaryotic cells, bacteria, and other organisms with cellular structures. EVs alter cell communication by delivering their contents and performing various functions depending on their cargo and release into certain environments or other cells. The cell walls of Gram-positive bacteria have a thick peptidoglycan layer and were previously thought to be unable to produce EVs. However, recent studies have demonstrated that Gram-positive bacterial EVs are crucial for health and disease. In this review, we have summarized the formation, composition, and characteristics of the contents, resistance to external stress, participation in immune regulation, and other functions of Gram-positive bacterial EVs, as well as their application in clinical diagnosis and treatment, to provide a new perspective to further our understanding of Gram-positive bacterial EVs.
Collapse
Affiliation(s)
| | | | | | - Xiaosong Qin
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Jianhua Liu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| |
Collapse
|
14
|
Nguyen MT, Hu Z, Mohammad M, Schöttler H, Niemann S, Schultz M, Barczyk-Kahlert K, Jin T, Hayen H, Herrmann M. Bacterial Lipoproteins Shift Cellular Metabolism to Glycolysis in Macrophages Causing Bone Erosion. Microbiol Spectr 2023; 11:e0429322. [PMID: 37191536 PMCID: PMC10269925 DOI: 10.1128/spectrum.04293-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 04/25/2023] [Indexed: 05/17/2023] Open
Abstract
Belonging to a group of membrane proteins, bacterial lipoproteins (LPPs) are defined by a unique lipid structure at their N-terminus providing the anchor in the bacterial cell membrane. In Gram-positive bacteria, LPPs play a key role in host immune activation triggered through a Toll-like receptor 2 (TLR2)-mediated action resulting in macrophage stimulation and subsequent tissue damage demonstrated in in vivo experimental models. Yet the physiologic links between LPP activation, cytokine release, and any underlying switches in cellular metabolism remain unclear. In this study, we demonstrate that Staphylococcus aureus Lpl1 not only triggers cytokine production but also confers a shift toward fermentative metabolism in bone marrow-derived macrophages (BMDMs). Lpl1 consists of di- and tri-acylated LPP variants; hence, the synthetic P2C and P3C, mimicking di-and tri-acylated LPPs, were employed to reveal their effect on BMDMs. Compared to P3C, P2C was found to shift the metabolism of BMDMs and the human mature monocytic MonoMac 6 (MM6) cells more profoundly toward the fermentative pathway, as indicated by lactate accumulation, glucose consumption, pH reduction, and oxygen consumption. In vivo, P2C caused more severe joint inflammation, bone erosion, and lactate and malate accumulation than P3C. These observed P2C effects were completely abrogated in monocyte/macrophage-depleted mice. Taken together, these findings now solidly confirm the hypothesized link between LPP exposure, a macrophage metabolic shift toward fermentation, and ensuing bone destruction. IMPORTANCE Osteomyelitis caused by S. aureus is a severe infection of the bone, typically associated with severe bone function impairment, therapeutic failure, high morbidity, invalidity, and occasionally even death. The hallmark of staphylococcal osteomyelitis is the destruction of the cortical bone structures, yet the mechanisms contributing to this pathology are hitherto poorly understood. One bacterial membrane constituent found in all bacteria is bacterial lipoproteins (LPPs). Previously, we have shown that injection of purified S. aureus LPPs into wild-type mouse knee joints caused a TLR2-dependent chronic destructive arthritis but failed to elicit such effect in monocyte/macrophage-depleted mice. This observation stirred our interest in investigating the interaction of LPPs and macrophages and analyzing the underlying physiological mechanisms. This ascertainment of LPP-induced changes in the physiology of macrophages provides an important clue in the understanding of the mechanisms of bone disintegration, opening novel avenues to manage the course of S. aureus disease.
Collapse
Affiliation(s)
- Minh-Thu Nguyen
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Zhicheng Hu
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hannah Schöttler
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Silke Niemann
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Michelle Schultz
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Heiko Hayen
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Mathias Herrmann
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| |
Collapse
|
15
|
Zhang K, Jia Y, Qian Y, Jiang X, Zhang S, Liu B, Cao J, Song Y, Mao W. Staphylococcus aureus increases Prostaglandin E 2 secretion in cow neutrophils by activating TLR2, TLR4, and NLRP3 inflammasome signaling pathways. Front Microbiol 2023; 14:1163261. [PMID: 37168122 PMCID: PMC10165004 DOI: 10.3389/fmicb.2023.1163261] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/24/2023] [Indexed: 05/13/2023] Open
Abstract
Introduction In clinical settings, dairy cows are often attacked by pathogenic bacteria after delivery, especially Staphylococcus aureus (S. aureus). Neutrophils have long been regarded as essential for host defense against S. aureus. Prostaglandin E2 (PGE2) can additionally be used as an inflammatory mediator in pathological conditions to promote the repair of inflammatory injuries. However, whether S. aureus can promote the accumulation of PGE2 after the infection of neutrophils in cows and its mechanism remain unclear. Lipoprotein is an important immune bioactive ingredient of S. aureus. Methods In this study, the changes in neutrophils were monitored in dairy cows infected with wild-type S. aureus (SA113) and an S. aureus lipoprotein-deficient strain (Δlgt); meanwhile, we established whether pattern recognition receptors mediate this process and whether S. aureus lipoproteins are necessary for causing the release of PGE2 from cow neutrophils. Results The results showed that Δlgt was less effective than SA113 in inducing the production of IL-1β, IL-6, IL-8, IL-10, and PGE2 within neutrophils; furthermore, TLR2, TLR4, and NLRP3 receptors were found to mediate the inducible effect of lipoprotein on the above inflammation mediators and cytokines, which depended on MAPK and Caspase-1 signaling pathways. In addition, TLR2, TLR4, and NLRP3 inhibitors significantly inhibited PGE2 and cytokine secretion, and PGE2 was involved in the interaction of S. aureus and neutrophils in dairy cows, which could be regulated by TLR2, TLR4, and NLRP3 receptors. We also found that S. aureus was more likely to be killed by neutrophils when it lacked lipoprotein and TLR2, TLR4, and NLRP3 were involved, but PGE2 seemed to have no effect. Discussion Taken together, these results suggest that lipoprotein is a crucial component of S. aureus in inducing cytokine secretion by neutrophils as well as killing within neutrophils, which could be accomplished by the accumulation of PGE2 by activating MAPK and the Caspase-1 signaling pathways through TLR2, TLR4, and NLRP3 receptors. These results will contribute to a better understanding of the interaction between S. aureus and host immune cells in dairy cows.
Collapse
Affiliation(s)
- Kai Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China
- Key Laboratory of Animal Clinical Treatment Technology, Ministry of Agriculture, Huhhot, China
| | - Yan Jia
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China
- Key Laboratory of Animal Clinical Treatment Technology, Ministry of Agriculture, Huhhot, China
| | - Yinghong Qian
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Huhhot, China
| | - Xueying Jiang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China
- Key Laboratory of Animal Clinical Treatment Technology, Ministry of Agriculture, Huhhot, China
| | - Shuangyi Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China
- Key Laboratory of Animal Clinical Treatment Technology, Ministry of Agriculture, Huhhot, China
| | - Bo Liu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China
- Key Laboratory of Animal Clinical Treatment Technology, Ministry of Agriculture, Huhhot, China
| | - Jinshan Cao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China
- Key Laboratory of Animal Clinical Treatment Technology, Ministry of Agriculture, Huhhot, China
| | - Yongli Song
- Stem Cell and Microbiology, Inner Mongolia University, Huhhot, China
| | - Wei Mao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China
- Key Laboratory of Animal Clinical Treatment Technology, Ministry of Agriculture, Huhhot, China
- *Correspondence: Wei Mao
| |
Collapse
|
16
|
Seffer MT, Weinert M, Molinari G, Rohde M, Gröbe L, Kielstein JT, Engelmann S. Staphylococcus aureus binding to Seraph® 100 Microbind® Affinity Filter: Effects of surface protein expression and treatment duration. PLoS One 2023; 18:e0283304. [PMID: 36930680 PMCID: PMC10022791 DOI: 10.1371/journal.pone.0283304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/05/2023] [Indexed: 03/18/2023] Open
Abstract
INTRODUCTION Extracorporeal blood purification systems represent a promising alternative for treatment of blood stream infections with multiresistant bacteria. OBJECTIVES The aim of this study was to analyse the binding activity of S. aureus to Seraph affinity filters based on heparin coated beads and to identify effectors influencing this binding activity. RESULTS To test the binding activity, we used gfp-expressing S. aureus Newman strains inoculated either in 0.9% NaCl or in blood plasma and determined the number of unbound bacteria by FACS analyses after passing through Seraph affinity filters. The binding activity of S. aureus was clearly impaired in human plasma: while a percent removal of 42% was observed in 0.9% NaCl (p-value 0.0472) using Seraph mini columns, a percent removal of only 10% was achieved in human plasma (p-value 0.0934). The different composition of surface proteins in S. aureus caused by the loss of SarA, SigB, Lgt, and SaeS had no significant influence on its binding activity. In a clinically relevant approach using the Seraph® 100 Microbind® Affinity Filter and 1000 ml of human blood plasma from four different donors, the duration of treatment was shown to have a critical effect on the rate of bacterial reduction. Within the first four hours, the number of bacteria decreased continuously and the reduction in bacteria reached statistical significance after two hours of treatment (percentage reduction 64%, p-value 0.01165). The final reduction after four hours of treatment was close to 90% and is dependent on donor. The capacity of Seraph® 100 for S. aureus in human plasma was approximately 5 x 108 cells. CONCLUSIONS The Seraph affinity filter, based on heparin-coated beads, is a highly efficient method for reducing S. aureus in human blood plasma, with efficiency dependent on blood plasma composition and treatment duration.
Collapse
Affiliation(s)
- Malin-Theres Seffer
- Helmholtz Centre for Infection Research, Microbial Proteomics, Braunschweig, Germany
- Medical Clinic V, Nephrology, Rheumatology, Blood Purification, Academic Teaching Hospital Braunschweig, Braunschweig, Germany
| | - Martin Weinert
- Helmholtz Centre for Infection Research, Microbial Proteomics, Braunschweig, Germany
- Technische Universität Braunschweig, Institute for Microbiology, Braunschweig, Germany
| | - Gabriella Molinari
- Helmholtz Centre for Infection Research, Central Facility of Microscopy, Braunschweig Germany
| | - Manfred Rohde
- Helmholtz Centre for Infection Research, Central Facility of Microscopy, Braunschweig Germany
| | - Lothar Gröbe
- Helmholtz Centre for Infection Research, Experimental Immunology, Braunschweig, Germany
| | - Jan T. Kielstein
- Medical Clinic V, Nephrology, Rheumatology, Blood Purification, Academic Teaching Hospital Braunschweig, Braunschweig, Germany
| | - Susanne Engelmann
- Helmholtz Centre for Infection Research, Microbial Proteomics, Braunschweig, Germany
- Technische Universität Braunschweig, Institute for Microbiology, Braunschweig, Germany
- * E-mail:
| |
Collapse
|
17
|
Kuijk MM, Wu Y, van Hensbergen VP, Shanlitourk G, Payré C, Lambeau G, Man-Bovenkerk S, Herrmann J, Müller R, van Strijp JAG, Pannekoek Y, Touqui L, van Sorge NM. Interference with Lipoprotein Maturation Sensitizes Methicillin-Resistant Staphylococcus aureus to Human Group IIA-Secreted Phospholipase A2 and Daptomycin. J Innate Immun 2022; 15:333-350. [PMID: 36473432 PMCID: PMC10643906 DOI: 10.1159/000527549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) has been classified as a high priority pathogen by the World Health Organization underlining the high demand for new therapeutics to treat infections. Human group IIA-secreted phospholipase A2 (hGIIA) is among the most potent bactericidal proteins against Gram-positive bacteria, including S. aureus. To determine hGIIA-resistance mechanisms of MRSA, we screened the Nebraska Transposon Mutant Library using a sublethal concentration of recombinant hGIIA. We identified and confirmed the role of lspA, encoding the lipoprotein signal peptidase LspA, as a new hGIIA resistance gene in both in vitro assays and an infection model in hGIIA-transgenic mice. Increased susceptibility of the lspA mutant was associated with enhanced activity of hGIIA on the cell membrane. Moreover, lspA deletion increased susceptibility to daptomycin, a last-resort antibiotic to treat MRSA infections. MRSA wild type could be sensitized to hGIIA and daptomycin killing through exposure to LspA-specific inhibitors globomycin and myxovirescin A1. Analysis of >26,000 S. aureus genomes showed that LspA is highly sequence-conserved, suggesting universal application of LspA inhibition. The role of LspA in hGIIA resistance was not restricted to MRSA since Streptococcus mutans and Enterococcus faecalis were also more hGIIA-susceptible after lspA deletion or LspA inhibition, respectively. Overall, our data suggest that pharmacological interference with LspA may disarm Gram-positive pathogens, including MRSA, to enhance clearance by innate host defense molecules and clinically applied antibiotics.
Collapse
Affiliation(s)
- Marieke M Kuijk
- Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location University of Amsterdam, Amsterdam, The Netherlands,
| | - Yongzheng Wu
- Unité de Biologie Cellulaire de l'Infection Microbionne, CNRS UMR3691, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Vincent P van Hensbergen
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Gizem Shanlitourk
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Christine Payré
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne Sophia Antipolis, France
| | - Gérard Lambeau
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne Sophia Antipolis, France
| | - Sandra Man-Bovenkerk
- Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location University of Amsterdam, Amsterdam, The Netherlands
| | - Jennifer Herrmann
- Department of Pharmacy at Saarland University, Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarbrücken, Germany
| | - Rolf Müller
- Department of Pharmacy at Saarland University, Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarbrücken, Germany
| | - Jos A G van Strijp
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Yvonne Pannekoek
- Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location University of Amsterdam, Amsterdam, The Netherlands
| | - Lhousseine Touqui
- Mucoviscidose et Bronchopathies Chroniques, Institut Pasteur, Université de Paris Cité, Paris, France
- Sorbonne Université, INSERM UMR S 938, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - Nina M van Sorge
- Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location University of Amsterdam, Amsterdam, The Netherlands
- Netherlands Reference Laboratory for Bacterial Meningitis, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Mohammad M, Ali A, Nguyen MT, Götz F, Pullerits R, Jin T. Staphylococcus aureus lipoproteins in infectious diseases. Front Microbiol 2022; 13:1006765. [PMID: 36262324 PMCID: PMC9574248 DOI: 10.3389/fmicb.2022.1006765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Infections with the Gram-positive bacterial pathogen Staphylococcus aureus remain a major challenge for the healthcare system and demand new treatment options. The increasing antibiotic resistance of S. aureus poses additional challenges, consequently inflicting a huge strain in the society due to enormous healthcare costs. S. aureus expresses multiple molecules, including bacterial lipoproteins (Lpps), which play a role not only in immune response but also in disease pathogenesis. S. aureus Lpps, the predominant ligands of TLR2, are important for bacterial survival as they maintain the metabolic activity of the bacteria. Moreover, Lpps possess many diverse properties that are of vital importance for the bacteria. They also contribute to host cell invasion but so far their role in different staphylococcal infections has not been fully defined. In this review, we summarize the current knowledge about S. aureus Lpps and their distinct roles in various infectious disease animal models, such as septic arthritis, sepsis, and skin and soft tissue infections. The molecular and cellular response of the host to S. aureus Lpp exposure is also a primary focus.
Collapse
Affiliation(s)
- Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Abukar Ali
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Minh-Thu Nguyen
- Section of Medical and Geographical Infectiology, Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Friedrich Götz
- Department of Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
| | - Rille Pullerits
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
19
|
TLR2, TLR4, and NLRP3 mediated the balance between host immune-driven resistance and tolerance in Staphylococcus aureus-infected mice. Microb Pathog 2022; 169:105671. [PMID: 35811022 DOI: 10.1016/j.micpath.2022.105671] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/15/2022] [Accepted: 07/05/2022] [Indexed: 01/02/2023]
Abstract
Staphylococcus aureus (S. aureus) is a gram-positive pathogen that can cause infectious diseases in mammals. S. aureus-induced host innate immune responses have a relationship with Toll-like receptor 2 (TLR2), TLR4, and Nod-like receptor pyrin domain-containing protein 3 (NLRP3). However, the detailed roles of TLR2, TLR4, and NLRP3 in regulating the host inflammatory response to S. aureus infection remain unclear. Our data indicated that the S. aureus-induced mortality was aggravated by deficiency of TLR2, TLR4, and NLRP3 in mice. In the subsequent experiment, we found that during S. aureus infection, the roles of TLR2, TLR4, and NLRP3 seemed to be different at multiple timepoints. The deficiency of TLR2, TLR4, or NLRP3 attenuated the expression of High-mobility group box protein 1 (HMGB1) and Hyaluronic acid-binding protein 2 (HABP2), which is accompanied by decreased proinflammatory cytokine (TNF-α), chemokine (RANTES), and anti-inflammatory cytokine (IL-10) production in lungs and serum at 3 h and 6 h post-infection. However, with S. aureus infection prolonged (24 h post-infection), the trend was diametrically opposite. The results showed that deficiency of TLR2, TLR4, or NLRP3 aggravated HABP2 and HMGB1 expression, which is accompanied by enhanced proinflammatory cytokine (TNF-α), chemokine (RANTES), and anti-inflammatory cytokine (IL-10) production in lungs and serum. These results were consistent with the data observed in S. aureus-infected bone marrow-derived macrophages (BMDMs). All these results suggested that during S. aureus infection, TLR2, TLR4, and NLRP3 has time-dependent effect in regulating the balance between immune-driven resistance and tolerance.
Collapse
|
20
|
Exploring the Role of Staphylococcus aureus in Inflammatory Diseases. Toxins (Basel) 2022; 14:toxins14070464. [PMID: 35878202 PMCID: PMC9318596 DOI: 10.3390/toxins14070464] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 02/04/2023] Open
Abstract
Staphylococcus aureus is a very common Gram-positive bacterium, and S. aureus infections play an extremely important role in a variety of diseases. This paper describes the types of virulence factors involved, the inflammatory cells activated, the process of host cell death, and the associated diseases caused by S. aureus. S. aureus can secrete a variety of enterotoxins and other toxins to trigger inflammatory responses and activate inflammatory cells, such as keratinocytes, helper T cells, innate lymphoid cells, macrophages, dendritic cells, mast cells, neutrophils, eosinophils, and basophils. Activated inflammatory cells can express various cytokines and induce an inflammatory response. S. aureus can also induce host cell death through pyroptosis, apoptosis, necroptosis, autophagy, etc. This article discusses S. aureus and MRSA (methicillin-resistant S. aureus) in atopic dermatitis, psoriasis, pulmonary cystic fibrosis, allergic asthma, food poisoning, sarcoidosis, multiple sclerosis, and osteomyelitis. Summarizing the pathogenic mechanism of Staphylococcus aureus provides a basis for the targeted treatment of Staphylococcus aureus infection.
Collapse
|
21
|
von Köckritz-Blickwede M, Winstel V. Molecular Prerequisites for Neutrophil Extracellular Trap Formation and Evasion Mechanisms of Staphylococcus aureus. Front Immunol 2022; 13:836278. [PMID: 35237275 PMCID: PMC8884242 DOI: 10.3389/fimmu.2022.836278] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/19/2022] [Indexed: 12/15/2022] Open
Abstract
NETosis is a multi-facetted cellular process that promotes the formation of neutrophil extracellular traps (NETs). NETs as web-like structures consist of DNA fibers armed with granular proteins, histones, and microbicidal peptides, thereby exhibiting pathogen-immobilizing and antimicrobial attributes that maximize innate immune defenses against invading microbes. However, clinically relevant pathogens often tolerate entrapment and even take advantage of the remnants of NETs to cause persistent infections in mammalian hosts. Here, we briefly summarize how Staphylococcus aureus, a high-priority pathogen and causative agent of fatal diseases in humans as well as animals, catalyzes and concurrently exploits NETs during pathogenesis and recurrent infections. Specifically, we focus on toxigenic and immunomodulatory effector molecules produced by staphylococci that prime NET formation, and further highlight the molecular and underlying principles of suicidal NETosis compared to vital NET-formation by viable neutrophils in response to these stimuli. We also discuss the inflammatory potential of NET-controlled microenvironments, as excessive expulsion of NETs from activated neutrophils provokes local tissue injury and may therefore amplify staphylococcal disease severity in hospitalized or chronically ill patients. Combined with an overview of adaptation and counteracting strategies evolved by S. aureus to impede NET-mediated killing, these insights may stimulate biomedical research activities to uncover novel aspects of NET biology at the host-microbe interface.
Collapse
Affiliation(s)
- Maren von Köckritz-Blickwede
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Volker Winstel
- Research Group Pathogenesis of Bacterial Infections, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
- *Correspondence: Volker Winstel,
| |
Collapse
|
22
|
Krueger A, Zaugg J, Chisholm S, Linedale R, Lachner N, Teoh SM, Tuong ZK, Lukowski SW, Morrison M, Soyer HP, Hugenholtz P, Hill MM, Frazer IH. Secreted Toxins From Staphylococcus aureus Strains Isolated From Keratinocyte Skin Cancers Mediate Pro-tumorigenic Inflammatory Responses in the Skin. Front Microbiol 2022; 12:789042. [PMID: 35145494 PMCID: PMC8822148 DOI: 10.3389/fmicb.2021.789042] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/08/2021] [Indexed: 12/27/2022] Open
Abstract
Squamous cell carcinoma (SCC) is a common type of skin cancer that typically arises from premalignant precursor lesions named actinic keratoses (AK). Chronic inflammation is a well-known promoter of skin cancer progression. AK and SCC have been associated with an overabundance of the bacterium Staphylococcus aureus (S. aureus). Certain secreted products from S. aureus are known to promote cutaneous pro-inflammatory responses; however, not all S. aureus strains produce these. As inflammation plays a key role in SCC development, we investigated the pro-inflammatory potential and toxin secretion profiles of skin-cancer associated S. aureus. Sterile culture supernatants (“secretomes”) of S. aureus clinical strains isolated from AK and SCC were applied to human keratinocytes in vitro. Some S. aureus secretomes induced keratinocytes to overexpress inflammatory mediators that have been linked to skin carcinogenesis, including IL-6, IL-8, and TNFα. A large phenotypic variation between the tested clinical strains was observed. Strains that are highly pro-inflammatory in vitro also caused more pronounced skin inflammation in mice. Proteomic characterization of S. aureus secretomes using mass spectrometry established that specific S. aureus enzymes and cytolytic toxins, including hemolysins, phenol-soluble modulins, and serine proteases, as well as currently uncharacterized proteins, correlate with the pro-inflammatory S. aureus phenotype. This study is the first to describe the toxin secretion profiles of AK and SCC-associated S. aureus, and their potential to induce a pro-inflammatory environment in the skin. Further studies are needed to establish whether these S. aureus products promote SCC development by mediating chronic inflammation.
Collapse
Affiliation(s)
- Annika Krueger
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Julian Zaugg
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Sarah Chisholm
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Richard Linedale
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Nancy Lachner
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Siok Min Teoh
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Zewen K. Tuong
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Samuel W. Lukowski
- The Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Mark Morrison
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - H. Peter Soyer
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
- Dermatology Department, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Philip Hugenholtz
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Michelle M. Hill
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- The University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Ian H. Frazer
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
- *Correspondence: Ian H. Frazer,
| |
Collapse
|
23
|
Liu B, Li Q, Gong Z, Zhao J, Gu B, Feng S. Staphylococcus aureus lipoproteins play crucial roles in inducing inflammatory responses and bacterial internalization into bovine mammary epithelial cells. Microb Pathog 2021; 162:105364. [PMID: 34921958 DOI: 10.1016/j.micpath.2021.105364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/11/2021] [Accepted: 12/14/2021] [Indexed: 01/10/2023]
Abstract
Bovine mastitis is caused by bacterial infection and characterized by inflammatory and infectious processes. Staphylococcus aureus frequently causes subclinical mastitis in dairy cows. In this study, we aimed to investigate the roles of S. aureus lipoproteins in inducing inflammatory responses and in mediating bacterial internalization into bovine mammary epithelial cells (bMECs). The results showed that TLR2 expression in bMECs infected with S. aureus isogenic mutant deficient in lipoprotein maturation was decreased compared to that in bMECs infected with wild-type S. aureus. Lipoproteins from S. aureus and the engagement of TLR2 were essential for inducing the activation of MAPK and NF-κB signaling, and stimulating the secretion of the inflammatory mediators tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and C-X-C motif chemokine ligand 8 (CXCL8). The production of prostaglandin E2 (PGE2) and the expression of PTGS2 in S. aureus-infected bMECs were dependent on the presence of bacterial lipoproteins. Furthermore, bacterial lipoproteins contributed to S. aureus internalization into bMECs. These findings suggest the S. aureus lipoproteins are key immunobiologically active compounds that trigger inflammatory responses in bMECs and play an important role in S. aureus internalization into bMECs.
Collapse
Affiliation(s)
- Bo Liu
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 306, Zhaowuda Road, Saihan District, 010018, Hohhot, China
| | - Qianru Li
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 306, Zhaowuda Road, Saihan District, 010018, Hohhot, China
| | - Zhiguo Gong
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 306, Zhaowuda Road, Saihan District, 010018, Hohhot, China
| | - Jiamin Zhao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 306, Zhaowuda Road, Saihan District, 010018, Hohhot, China
| | - Baichen Gu
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 306, Zhaowuda Road, Saihan District, 010018, Hohhot, China
| | - Shuang Feng
- Laboratory of Veterinary Public Health, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 306, Zhaowuda Road, Saihan District, 010018, Hohhot, China.
| |
Collapse
|
24
|
Smithers L, Olatunji S, Caffrey M. Bacterial Lipoprotein Posttranslational Modifications. New Insights and Opportunities for Antibiotic and Vaccine Development. Front Microbiol 2021; 12:788445. [PMID: 34950121 PMCID: PMC8689077 DOI: 10.3389/fmicb.2021.788445] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/16/2021] [Indexed: 12/17/2022] Open
Abstract
Lipoproteins are some of the most abundant proteins in bacteria. With a lipid anchor to the cell membrane, they function as enzymes, inhibitors, transporters, structural proteins, and as virulence factors. Lipoproteins activate the innate immune system and have biotechnological applications. The first lipoprotein was described by Braun and Rehn in 1969. Up until recently, however, work on lipoproteins has been sluggish, in part due to the challenges of handling proteins that are anchored to membranes by covalently linked lipids or are membrane integral. Activity in the area has quickened of late. In the past 5 years, high-resolution structures of the membrane enzymes of the canonical lipoprotein synthesis pathway have been determined, new lipoprotein types have been discovered and the enzymes responsible for their synthesis have been characterized biochemically. This has led to a flurry of activity aimed at developing novel antibiotics targeting these enzymes. In addition, surface exposed bacterial lipoproteins have been utilized as candidate vaccine antigens, and their potential to act as self-adjuvanting antigens is increasingly recognized. A summary of the latest developments in lipoproteins and their synthesis, as well as how this information is being exploited for therapeutic purposes is presented here.
Collapse
Affiliation(s)
- Luke Smithers
- School of Medicine, Trinity College Dublin, Dublin, Ireland
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Samir Olatunji
- School of Medicine, Trinity College Dublin, Dublin, Ireland
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Martin Caffrey
- School of Medicine, Trinity College Dublin, Dublin, Ireland
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
25
|
Payen S, Roy D, Boa A, Okura M, Auger JP, Segura M, Gottschalk M. Role of Maturation of Lipoproteins in the Pathogenesis of the Infection Caused by Streptococcus suis Serotype 2. Microorganisms 2021; 9:microorganisms9112386. [PMID: 34835511 PMCID: PMC8621357 DOI: 10.3390/microorganisms9112386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/03/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022] Open
Abstract
Streptococcus suis serotype 2 is an important porcine bacterial pathogen associated with multiple pathologies in piglets. Bacterial lipoproteins (LPPs) have been described as playing important roles in the pathogenesis of the infection of other Gram-positive bacteria as adhesins, pro-inflammatory cell activators and/or virulence factors. In the current study, we aimed to evaluate the role of the prolipoprotein diacylglyceryl transferase (Lgt) and lipoprotein signal peptidase (Lsp) enzymes, which are responsible for LPP maturation, on the pathogenesis of the infection caused by two different sequence types (STs) of S. suis serotype 2 strains (virulent ST1 and highly virulent ST7). Through the use of isogenic Δlgt, Δlsp and double Δlgt/Δlsp mutants, it was shown that lack of these enzymes did not influence S. suis adhesion/invasion to porcine respiratory epithelial cells. However, in the absence of the Lsp and/or Lgt, a significant reduction in the capacity of S. suis to activate phagocytic cells and induce pro-inflammatory mediators (in vitro and in vivo) was observed. In general, results obtained with the double mutant did not differ in comparison to single mutants, indicating lack of an additive effect. Finally, our data suggest that these enzymes play a differential role in virulence, depending on the genetic background of the strain and being more important for the highly virulent ST7 strain.
Collapse
Affiliation(s)
- Servane Payen
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (D.R.); (A.B.); (J.-P.A.); (M.S.)
| | - David Roy
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (D.R.); (A.B.); (J.-P.A.); (M.S.)
| | - Anaïs Boa
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (D.R.); (A.B.); (J.-P.A.); (M.S.)
| | - Masatoshi Okura
- Division of Bacterial and Parasitic Disease, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba 305-0856, Japan;
| | - Jean-Philippe Auger
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (D.R.); (A.B.); (J.-P.A.); (M.S.)
| | - Mariela Segura
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (D.R.); (A.B.); (J.-P.A.); (M.S.)
| | - Marcelo Gottschalk
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (D.R.); (A.B.); (J.-P.A.); (M.S.)
- Correspondence:
| |
Collapse
|
26
|
Chen H, Gao H, Xie HT, Liu ST, Huang YK, Zhang MC. Hyperkeratinization and Proinflammatory Cytokine Expression in Meibomian Glands Induced by Staphylococcus aureus. Invest Ophthalmol Vis Sci 2021; 62:11. [PMID: 34643663 PMCID: PMC8525831 DOI: 10.1167/iovs.62.13.11] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
Purpose This exploratory study aimed to investigate the morphological and pathological alterations of the meibomian gland (MG) with the Staphylococcus aureus crude extracts (SACEs) treatment. Methods Mouse MG explants were cultured and differentiated with or without SACEs for 48 hours. Explant's viability and cell death were determined by thiazolyl blue tetrazolium bromide (MTT) assay and TUNEL assay. MG morphology was observed by Hematoxylin and Eosin staining. Lipid droplet production was detected by Nile Red staining and LipidTox immunostaining. The pro-inflammatory cytokines were detected by ELISA. The relative gene and protein expression in MG explants was determined via quantitative RT-PCR, immunostaining, and immunoblotting. The components of the SACEs were analyzed by immunoblotting and silver staining. Results Our findings demonstrated that the SACEs treatment induced overexpression of keratin 1 (Krt1) in the ducts and acini of MG explants, accompanied by a decrease in viability and an increase in cell death in explants. Furthermore, the SACEs treatment dose-dependently increased the levels of TNF-α, IL-1β, and IL-6 in MG explants. The SACEs treatment induced activation of the nuclear factor kappa B (NF-κB) and AIM2 (absent in melanoma 2)/ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain) inflammasome signaling pathway in explants. Further investigation showed expression of the key adipogenesis-related molecule peroxisome proliferator-activated receptor γ was decreased after SACEs treatment. However, no change was found in the lipid synthesis of MG explants after treatment with the SACEs. Staphylococcal enterotoxins B (SEB) was detected in the SACEs. SEB induced the overexpression of Krt1 and IL-1β in ducts and acini of MG explants. Conclusions Our findings confirm that Staphylococcus aureus induced hyperkeratinization and pro-inflammatory cytokines expression in MG explants ducts and acini. These effects might be mediated by SEB. Activation of the NF-κB and AIM2/ASC signaling pathway is involved in this process.
Collapse
Affiliation(s)
- Hua Chen
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Gao
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua-Tao Xie
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu-Ting Liu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Kan Huang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming-Chang Zhang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Venkataranganayaka Abhilasha K, Kedihithlu Marathe G. Bacterial lipoproteins in sepsis. Immunobiology 2021; 226:152128. [PMID: 34488139 DOI: 10.1016/j.imbio.2021.152128] [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: 01/08/2021] [Revised: 07/09/2021] [Accepted: 08/10/2021] [Indexed: 01/05/2023]
Abstract
Bacterial lipoproteins are membrane proteins derived from both gram-negative and gram-positive bacteria. They seem to have diverse functions not only on bacterial growth, but also play an important role in host's virulence. Bacterial lipoproteins exert their action on host immune cells via TLR2/1 or TLR2/6. Therefore, bacterial lipoproteins also need to be considered while addressing bacterial pathogenicity besides classical bacterial endotoxin like LPS and other microbial associated molecular patterns such as LTA, and peptidoglycans. In this mini-review, we provide an overview of general bacterial lipoprotein biosynthesis and the need to understand the lipoprotein-mediated pathogenicity in diseases like sepsis.
Collapse
Affiliation(s)
- Kandahalli Venkataranganayaka Abhilasha
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru 570006, Karnataka, India; Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Gopal Kedihithlu Marathe
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru 570006, Karnataka, India; Department of Studies in Molecular Biology, University of Mysore, Manasagangothri, Mysuru 570006, Karnataka, India.
| |
Collapse
|
28
|
Chen X, Teoh WP, Stock MR, Resko ZJ, Alonzo F. Branched chain fatty acid synthesis drives tissue-specific innate immune response and infection dynamics of Staphylococcus aureus. PLoS Pathog 2021; 17:e1009930. [PMID: 34496007 PMCID: PMC8452012 DOI: 10.1371/journal.ppat.1009930] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/20/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022] Open
Abstract
Fatty acid-derived acyl chains of phospholipids and lipoproteins are central to bacterial membrane fluidity and lipoprotein function. Though it can incorporate exogenous unsaturated fatty acids (UFA), Staphylococcus aureus synthesizes branched chain fatty acids (BCFA), not UFA, to modulate or increase membrane fluidity. However, both endogenous BCFA and exogenous UFA can be attached to bacterial lipoproteins. Furthermore, S. aureus membrane lipid content varies based upon the amount of exogenous lipid in the environment. Thus far, the relevance of acyl chain diversity within the S. aureus cell envelope is limited to the observation that attachment of UFA to lipoproteins enhances cytokine secretion by cell lines in a TLR2-dependent manner. Here, we leveraged a BCFA auxotroph of S. aureus and determined that driving UFA incorporation disrupted infection dynamics and increased cytokine production in the liver during systemic infection of mice. In contrast, infection of TLR2-deficient mice restored inflammatory cytokines and bacterial burden to wildtype levels, linking the shift in acyl chain composition toward UFA to detrimental immune activation in vivo. In in vitro studies, bacterial lipoproteins isolated from UFA-supplemented cultures were resistant to lipase-mediated ester hydrolysis and exhibited heightened TLR2-dependent innate cell activation, whereas lipoproteins with BCFA esters were completely inactivated after lipase treatment. These results suggest that de novo synthesis of BCFA reduces lipoprotein-mediated TLR2 activation and improves lipase-mediated hydrolysis making it an important determinant of innate immunity. Overall, this study highlights the potential relevance of cell envelope acyl chain repertoire in infection dynamics of bacterial pathogens.
Collapse
Affiliation(s)
- Xi Chen
- Department of Microbiology and Immunology, Loyola University Chicago–Stritch School of Medicine, Maywood, Illinois, United States of America
| | - Wei Ping Teoh
- Department of Microbiology and Immunology, Loyola University Chicago–Stritch School of Medicine, Maywood, Illinois, United States of America
| | - Madison R. Stock
- Department of Microbiology and Immunology, Loyola University Chicago–Stritch School of Medicine, Maywood, Illinois, United States of America
| | - Zachary J. Resko
- Department of Microbiology and Immunology, Loyola University Chicago–Stritch School of Medicine, Maywood, Illinois, United States of America
| | - Francis Alonzo
- Department of Microbiology and Immunology, Loyola University Chicago–Stritch School of Medicine, Maywood, Illinois, United States of America
| |
Collapse
|
29
|
Hook JS, Patel PA, O'Malley A, Xie L, Kavanaugh JS, Horswill AR, Moreland JG. Lipoproteins from Staphylococcus aureus Drive Neutrophil Extracellular Trap Formation in a TLR2/1- and PAD-Dependent Manner. THE JOURNAL OF IMMUNOLOGY 2021; 207:966-973. [PMID: 34290104 DOI: 10.4049/jimmunol.2100283] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/25/2021] [Indexed: 11/19/2022]
Abstract
Neutrophils, polymorphonuclear leukocytes (PMN), play a critical role in the innate immune response to Staphylococcus aureus, a pathogen that continues to be associated with significant morbidity and mortality. Neutrophil extracellular trap (NET) formation is involved in ensnaring and killing of S. aureus, but this host-pathogen interaction also leads to host tissue damage. Importantly, NET components including neutrophil proteases are under consideration as therapeutic targets in a variety of disease processes. Although S. aureus lipoproteins are recognized to activate cells via TLRs, specific mechanisms of interaction with neutrophils are poorly delineated. We hypothesized that a lipoprotein-containing cell membrane preparation from methicillin-resistant S. aureus (MRSA-CMP) would elicit PMN activation, including NET formation. We investigated MRSA-CMP-elicited NET formation, regulated elastase release, and IL-8 production in human neutrophils. We studied PMN from healthy donors with or without a common single-nucleotide polymorphism in TLR1, previously demonstrated to impact TLR2/1 signaling, and used cell membrane preparation from both wild-type methicillin-resistant S. aureus and a mutant lacking palmitoylated lipoproteins (lgt). MRSA-CMP elicited NET formation, elastase release, and IL-8 production in a lipoprotein-dependent manner. TLR2/1 signaling was involved in NET formation and IL-8 production, but not elastase release, suggesting that MRSA-CMP-elicited elastase release is not mediated by triacylated lipoproteins. MRSA-CMP also primed neutrophils for enhanced NET formation in response to a subsequent stimulus. MRSA-CMP-elicited NET formation did not require Nox2-derived reactive oxygen species and was partially dependent on the activity of peptidyl arginine deiminase (PAD). In conclusion, lipoproteins from S. aureus mediate NET formation via TLR2/1 with clear implications for patients with sepsis.
Collapse
Affiliation(s)
- Jessica S Hook
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Parth A Patel
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Aidan O'Malley
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Lihua Xie
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jeffrey S Kavanaugh
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO.,Department of Veterans Affairs, Eastern Colorado Healthcare System, Aurora, CO; and
| | - Jessica G Moreland
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX; .,Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
30
|
Lipoproteins Are Responsible for the Pro-Inflammatory Property of Staphylococcus aureus Extracellular Vesicles. Int J Mol Sci 2021; 22:ijms22137099. [PMID: 34281154 PMCID: PMC8268867 DOI: 10.3390/ijms22137099] [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: 06/15/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 01/08/2023] Open
Abstract
Staphylococcal aureus
(S. aureus), a Gram-positive bacteria, is known to cause various infections. Extracellular vesicles (EVs) are a heterogeneous array of membranous structures secreted by cells from all three domains of life, i.e., eukaryotes, bacteria, and archaea. Bacterial EVs are implied to be involved in both bacteria–bacteria and bacteria–host interactions during infections. It is still unclear how S. aureus EVs interact with host cells and induce inflammatory responses. In this study, EVs were isolated from S. aureus and mutant strains deficient in either prelipoprotein lipidation (Δlgt) or major surface proteins (ΔsrtAB). Their immunostimulatory capacities were assessed both in vitro and in vivo. We found that S. aureus EVs induced pro-inflammatory responses both in vitro and in vivo. However, this activity was dependent on lipidated lipoproteins (Lpp), since EVs isolated from the Δlgt showed no stimulation. On the other hand, EVs isolated from the ΔsrtAB mutant showed full immune stimulation, indicating the cell wall anchoring of surface proteins did not play a role in immune stimulation. The immune stimulation of S. aureus EVs was mediated mainly by monocytes/macrophages and was TLR2 dependent. In this study, we demonstrated that not only free Lpp but also EV-imbedded Lpp had high pro-inflammatory activity.
Collapse
|
31
|
Inhibition of Escherichia coli Lipoprotein Diacylglyceryl Transferase Is Insensitive to Resistance Caused by Deletion of Braun's Lipoprotein. J Bacteriol 2021; 203:e0014921. [PMID: 33875545 PMCID: PMC8316002 DOI: 10.1128/jb.00149-21] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Lipoprotein diacylglyceryl transferase (Lgt) catalyzes the first step in the biogenesis of Gram-negative bacterial lipoproteins which play crucial roles in bacterial growth and pathogenesis. We demonstrate that Lgt depletion in a clinical uropathogenic Escherichia coli strain leads to permeabilization of the outer membrane and increased sensitivity to serum killing and antibiotics. Importantly, we identify G2824 as the first-described Lgt inhibitor that potently inhibits Lgt biochemical activity in vitro and is bactericidal against wild-type Acinetobacter baumannii and E. coli strains. While deletion of a gene encoding a major outer membrane lipoprotein, lpp, leads to rescue of bacterial growth after genetic depletion or pharmacologic inhibition of the downstream type II signal peptidase, LspA, no such rescue of growth is detected after Lgt depletion or treatment with G2824. Inhibition of Lgt does not lead to significant accumulation of peptidoglycan-linked Lpp in the inner membrane. Our data validate Lgt as a novel antibacterial target and suggest that, unlike downstream steps in lipoprotein biosynthesis and transport, inhibition of Lgt may not be sensitive to one of the most common resistance mechanisms that invalidate inhibitors of bacterial lipoprotein biosynthesis and transport. IMPORTANCE As the emerging threat of multidrug-resistant (MDR) bacteria continues to increase, no new classes of antibiotics have been discovered in the last 50 years. While previous attempts to inhibit the lipoprotein biosynthetic (LspA) or transport (LolCDE) pathways have been made, most efforts have been hindered by the emergence of a common mechanism leading to resistance, namely, the deletion of the gene encoding a major Gram-negative outer membrane lipoprotein lpp. Our unexpected finding that inhibition of Lgt is not susceptible to lpp deletion-mediated resistance uncovers the complexity of bacterial lipoprotein biogenesis and the corresponding enzymes involved in this essential outer membrane biogenesis pathway and potentially points to new antibacterial targets in this pathway.
Collapse
|
32
|
Liu K, Mao W, Liu B, Li T, Wang X, Pei L, Cao J, Wang F. Prostaglandin E2 promotes Staphylococcus aureus infection via EP4 receptor in bovine endometrium. Microb Pathog 2021; 158:105019. [PMID: 34107344 DOI: 10.1016/j.micpath.2021.105019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 10/21/2022]
Abstract
Prostaglandin E2 (PGE2) enhances Staphylococcus aureus infection but its mechanism is not well understood. Here, we examined the effect of PGE2 on Staphylococcal Protein A (SPA) expression in bovine endometrium and determined the role of select PGE2 receptors (i.e., EP2 and EP4) in adhesion and internalization of S. aureus. S. aureus isolate SA113 was used for in vitro infection of bovine endometrial tissues and epithelial cells, with treatment conditions consisting of untreated control, SA113 treatment, SA113 + PGE2, SA113 + PGE2 + EP2 receptor antagonist (AH-6809), and SA113 + PGE2 + EP4 receptor antagonist (AH-23848). Immunofluorescence assay revealed that PGE2 could promote SPA expression in S. aureus-infected bovine endometrial tissues. PGE2 also enhanced the adhesion and internalization of S. aureus in bovine endometrial cells. The addition of EP4 antagonist, but not the EP2 antagonist, abrogated the ability of PGE2 to promote S. aureus SPA expression, adhesion, and internalization in endometrial cells. Our findings suggest that S. aureus infection in the endometrium is enhanced by PGE2 through the EP4 receptor. This result is essential for the development of new approach to treating S. aureus infection, such as the application of EP4 antagonist as an adjunct drug treatment.
Collapse
Affiliation(s)
- Kun Liu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Anima Disease, Ministry of Agriculture, Hohhot, China; Laboratory of Veterinary Pathology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China.
| | - Wei Mao
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Anima Disease, Ministry of Agriculture, Hohhot, China.
| | - Bo Liu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Anima Disease, Ministry of Agriculture, Hohhot, China.
| | - Tingting Li
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Anima Disease, Ministry of Agriculture, Hohhot, China.
| | - Xinfei Wang
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Anima Disease, Ministry of Agriculture, Hohhot, China.
| | - Le Pei
- Veterinary Research Institute, Inner Mongolia Academy of Agriculture and Animal Husbandry Sciences, Hohhot, China.
| | - Jinshan Cao
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Anima Disease, Ministry of Agriculture, Hohhot, China.
| | - Fenglong Wang
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China.
| |
Collapse
|
33
|
Lipoteichoic Acid from Staphylococcus aureus Activates the Complement System via C3 Induction and CD55 Inhibition. Microorganisms 2021; 9:microorganisms9061135. [PMID: 34074052 PMCID: PMC8225101 DOI: 10.3390/microorganisms9061135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 11/23/2022] Open
Abstract
Staphylococcus aureus inhibits complement activity by secreting a variety of toxins. However, the underlying mechanism of complement component regulation by lipoteichoic acid (LTA), a cell wall component of S. aureus, has not been elucidated. In this study, we observed that aLTA (LTA of S. aureus) increased C3 expression in THP-1 cells. The mechanism of aLTA-mediated C3 induction includes an aLTA-toll-like receptor (TLR) 2 interaction, interleukin 1 receptor associated kinase (IRAK) 2 recruitment, and nuclear factor kappa B (NF-kB) activation. In HepG2 cells, C3 protein production begins to increase from 3 h and increases steadily until 48 h. On the other hand, CD55 levels increased up to 6 h after aLTA treatment and started to decrease after 24 h and levels were decreased at 48 h by more than 50% compared to untreated cells. The expression of CD55 in HepG2 cells was shown to be regulated by IRAK-M induced by aLTA. Serum C3 levels increased in mice injected with aLTA, which resulted in an increase in the amount and activity of the membrane attack complex (MAC). We also observed that CD55 mRNA was increased in the liver 24 h after aLTA injection, but was decreased 48 h after injection. These results suggest that aLTA increases complement levels via induction of C3 and inhibition of CD55, which may cause associated MAC-mediated liver damage.
Collapse
|
34
|
Mohammad M, Na M, Hu Z, Nguyen MT, Kopparapu PK, Jarneborn A, Karlsson A, Ali A, Pullerits R, Götz F, Jin T. Staphylococcus aureus lipoproteins promote abscess formation in mice, shielding bacteria from immune killing. Commun Biol 2021; 4:432. [PMID: 33785850 PMCID: PMC8010101 DOI: 10.1038/s42003-021-01947-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 03/02/2021] [Indexed: 12/27/2022] Open
Abstract
Despite being a major bacterial factor in alerting the human immune system, the role of Staphylococcus aureus (S. aureus) lipoproteins (Lpp) in skin infections remains largely unknown. Here, we demonstrated that subcutaneous injection of S. aureus Lpp led to infiltration of neutrophils and monocytes/macrophages and induced skin lesions in mice. Lipid-moiety of S. aureus Lpp and host TLR2 was responsible for such effect. Lpp-deficient S. aureus strains exhibited smaller lesion size and reduced bacterial loads than their parental strains; the altered phenotype in bacterial loads was TLR2-independent. Lpp expression in skin infections contributed to imbalanced local hemostasis toward hypercoagulable state. Depletion of leukocytes or fibrinogen abrogated the effects induced by Lpp in terms of skin lesions and bacterial burden. Our data suggest that S. aureus Lpp induce skin inflammation and promote abscess formation that protects bacteria from innate immune killing. This suggests an intriguing bacterial immune evasion mechanism.
Collapse
Affiliation(s)
- Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Manli Na
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Zhicheng Hu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Microbiology and Immunology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Minh-Thu Nguyen
- Department of Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
- Section of Medical and Geographical Infectiology, Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Pradeep Kumar Kopparapu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Jarneborn
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anna Karlsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Abukar Ali
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rille Pullerits
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Friedrich Götz
- Department of Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
35
|
Soe YM, Bedoui S, Stinear TP, Hachani A. Intracellular Staphylococcus aureus and host cell death pathways. Cell Microbiol 2021; 23:e13317. [PMID: 33550697 DOI: 10.1111/cmi.13317] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 12/13/2022]
Abstract
Staphylococcus aureus is a major opportunistic human pathogen that is globally prevalent. Although S. aureus and humans may have co-evolved to the point of commensalism, the bacterium is equipped with virulence factors causing devastating infections. The adoption of an intracellular lifestyle by S. aureus is an important facet of its pathogenesis. Occupying a privileged intracellular compartment permits evasion from the bactericidal actions of host immunity and antibiotics. However, this localization exposes S. aureus to cell-intrinsic processes comprising autophagy, metabolic challenges and clearance mechanisms orchestrated by host programmed cell death pathways (PCDs), including apoptosis, pyroptosis and necroptosis. Mounting evidence suggests that S. aureus deploys pathoadaptive mechanisms that modulate the expression of its virulence factors to prevent elimination through PCD pathways. In this review, we critically analyse the current literature on the interplay between S. aureus virulence factors with the key, intertwined nodes of PCD. We discuss how S. aureus adaptation to the human host plays an essential role in the evasion of PCD, and we consider future directions to study S. aureus-PCD interactions.
Collapse
Affiliation(s)
- Ye Mon Soe
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Abderrahman Hachani
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
36
|
Forrest S, Welch M. Arming the troops: Post-translational modification of extracellular bacterial proteins. Sci Prog 2020; 103:36850420964317. [PMID: 33148128 PMCID: PMC10450907 DOI: 10.1177/0036850420964317] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Protein secretion is almost universally employed by bacteria. Some proteins are retained on the cell surface, whereas others are released into the extracellular milieu, often playing a key role in virulence. In this review, we discuss the diverse types and potential functions of post-translational modifications (PTMs) occurring to extracellular bacterial proteins.
Collapse
Affiliation(s)
- Suzanne Forrest
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Martin Welch
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
37
|
Nguyen MT, Matsuo M, Niemann S, Herrmann M, Götz F. Lipoproteins in Gram-Positive Bacteria: Abundance, Function, Fitness. Front Microbiol 2020; 11:582582. [PMID: 33042100 PMCID: PMC7530257 DOI: 10.3389/fmicb.2020.582582] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022] Open
Abstract
When one thinks of the Gram+ cell wall, the peptidoglycan (PG) scaffold in particular comes to mind. However, the cell wall also consists of many other components, for example those that are covalently linked to the PG: the wall teichoic acid and the cell wall proteins tethered by the sortase. In addition, there are completely different molecules that are anchored in the cytoplasmic membrane and span the cell wall. These are lipoteichoic acids and bacterial lipoproteins (Lpp). The latter are in the focus of this review. Lpp are present in almost all bacteria. They fulfill a wealth of different tasks. They represent the window to the outside world by recognizing nutrients and incorporating them into the bacterial cell via special transport systems. Furthermore, they perform very diverse and special tasks such as acting as chaperonin, as cyclomodulin, contributing to invasion of host cells or uptake of plasmids via conjugation. All these functions are taken over by the protein part. Nevertheless, the lipid part of the Lpp plays an as important role as the protein part. It is the released lipoproteins and derived lipopeptides that massively modulate our immune system and ultimately play an important role in immune tolerance or non-tolerance. All these varied activities of the Lpp are considered in this review article.
Collapse
Affiliation(s)
- Minh-Thu Nguyen
- Section of Medical and Geographical Infectiology, Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Miki Matsuo
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Microbial Genetics, University of Tübingen, Tübingen, Germany
| | - Silke Niemann
- Section of Medical and Geographical Infectiology, Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Mathias Herrmann
- Section of Medical and Geographical Infectiology, Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Friedrich Götz
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Microbial Genetics, University of Tübingen, Tübingen, Germany
| |
Collapse
|
38
|
NisI Maturation and Its Influence on Nisin Resistance in Lactococcus lactis. Appl Environ Microbiol 2020; 86:AEM.01306-20. [PMID: 32709730 DOI: 10.1128/aem.01306-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/22/2020] [Indexed: 02/02/2023] Open
Abstract
NisI confers immunity against nisin, with high substrate specificity to prevent a suicidal effect in nisin-producing Lactococcus lactis strains. However, the NisI maturation process as well as its influence on nisin resistance has not been characterized. Here, we report the roles of lipoprotein signal peptidase II (Lsp) and prolipoprotein diacylglyceryl transferase (Lgt) in NisI maturation and nisin resistance of L. lactis F44. We found that the resistance of nisin of an Lsp-deficient mutant remarkably decreased, while no significant differences in growth were observed. We demonstrated that Lsp could cleave signal peptide of NisI precursor in vitro Moreover, diacylglyceryl modification of NisI catalyzed by Lgt played a decisive role in attachment of NisI on the cell envelope, while it exhibited no effects on cleavage of the signal peptides of NisI precursor. The dissociation constant (KD ) for the interaction between nisin and NisI exhibited a 2.8-fold increase compared with that between nisin and pre-NisI with signal peptide by surface plasmon resonance (SPR) analysis, providing evidence that Lsp-catalyzed signal peptide cleavage was critical for the immune activity of NisI. Our study revealed the process of NisI maturation in L. lactis and presented a potential strategy to enhance industrial nisin production.IMPORTANCE Nisin, a safe and natural antimicrobial peptide, has a long and impressive history as a food preservative and is also considered a novel candidate to alleviate the increasingly serious threat of antibiotic resistance. Nisin is produced by certain L. lactis strains. The nisin immunity protein NisI, a membrane-bound lipoprotein, is expressed by nisin producers to avoid suicidal action. Here, we report the roles of Lsp and Lgt in NisI maturation and nisin resistance of L. lactis F44. The results verified the importance of Lsp to NisI-conferred immunity and Lgt to localization. Our study revealed the process of NisI maturation in L. lactis and presented a potential strategy to enhance industrial nisin production.
Collapse
|
39
|
Garland K, Pantua H, Braun MG, Burdick DJ, Castanedo GM, Chen YC, Cheng YX, Cheong J, Daniels B, Deshmukh G, Fu Y, Gibbons P, Gloor SL, Hua R, Labadie S, Liu X, Pastor R, Stivala C, Xu M, Xu Y, Zheng H, Kapadia SB, Hanan EJ. Optimization of globomycin analogs as novel gram-negative antibiotics. Bioorg Med Chem Lett 2020; 30:127419. [PMID: 32768648 DOI: 10.1016/j.bmcl.2020.127419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 11/29/2022]
Abstract
Discovery of novel classes of Gram-negative antibiotics with activity against multi-drug resistant infections is a critical unmet need. As an essential member of the lipoprotein biosynthetic pathway, lipoprotein signal peptidase II (LspA) is an attractive target for antibacterial drug discovery, with the natural product inhibitor globomycin offering a modestly-active starting point. Informed by structure-based design, the globomycin depsipeptide was optimized to improve activity against E. coli. Backbone modifications, together with adjustment of physicochemical properties, afforded potent compounds with good in vivo pharmacokinetic profiles. Optimized compounds such as 51 (E. coli MIC 3.1 μM) and 61 (E. coli MIC 0.78 μM) demonstrate broad spectrum activity against gram-negative pathogens and may provide opportunities for future antibiotic discovery.
Collapse
Affiliation(s)
- Keira Garland
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Homer Pantua
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | | - Yi-Chen Chen
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yun-Xing Cheng
- Pharmaron Beijing Co., Ltd, No. 6 Taihe Road, BDA, Beijing 100176, PR China
| | - Jonathan Cheong
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Blake Daniels
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Gauri Deshmukh
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yuhong Fu
- Pharmaron Beijing Co., Ltd, No. 6 Taihe Road, BDA, Beijing 100176, PR China
| | - Paul Gibbons
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Susan L Gloor
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Rongbao Hua
- Pharmaron Beijing Co., Ltd, No. 6 Taihe Road, BDA, Beijing 100176, PR China
| | - Sharada Labadie
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Xiongcai Liu
- Pharmaron Beijing Co., Ltd, No. 6 Taihe Road, BDA, Beijing 100176, PR China
| | - Richard Pastor
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Craig Stivala
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Min Xu
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yiming Xu
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hao Zheng
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Emily J Hanan
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
40
|
Lee IC, van Swam II, Boeren S, Vervoort J, Meijerink M, Taverne N, Starrenburg M, Bron PA, Kleerebezem M. Lipoproteins Contribute to the Anti-inflammatory Capacity of Lactobacillus plantarum WCFS1. Front Microbiol 2020; 11:1822. [PMID: 32849426 PMCID: PMC7403179 DOI: 10.3389/fmicb.2020.01822] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022] Open
Abstract
Bacterial lipoproteins are well-recognized microorganism-associated molecular patterns, which interact with Toll-like receptor (TLR) 2, an important pattern recognition receptor of the host innate immune system. Lipoproteins are conjugated with two- or three-acyl chains (di- or tri-acyl), which is essential for appropriate anchoring in the cell membrane as well as for the interaction with TLR2. Lipoproteins have mostly been studied in pathogens and have established roles in various biological processes, such as nutrient import, cell wall cross-linking and remodeling, and host-cell interaction. By contrast, information on the role of lipoproteins in the physiology and host interaction of probiotic bacteria is scarce. By deletion of lgt, encoding prolipoprotein diacylglyceryl transferase, responsible for lipidation of lipoprotein precursors, we investigated the roles of the collective group of lipoproteins in the physiology of the probiotic model strain Lactobacillus plantarum WCFS1 using proteomic analysis of secreted proteins. To investigate the consequences of the lgt mutation in host-cell interaction, the capacity of mutant and wild-type bacteria to stimulate TLR2 signaling and inflammatory responses was compared using (reporter-) cell-based models. These experiments exemplified the critical contribution of the acyl chains of lipoproteins in immunomodulation. To the best of our knowledge, this is the first study that investigated collective lipoprotein functions in a model strain for probiotic lactobacilli, and we show that the lipoproteins in L. plantarum WCFS1 are critical drivers of anti-inflammatory host responses toward this strain.
Collapse
Affiliation(s)
- I-Chiao Lee
- Host-Microbe Interactomics Group, Wageningen University & Research, Wageningen, Netherlands.,TiFN Food & Nutrition, Wageningen, Netherlands.,NIZO Food Research, Ede, Netherlands
| | - Iris I van Swam
- TiFN Food & Nutrition, Wageningen, Netherlands.,NIZO Food Research, Ede, Netherlands
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University & Research, Wageningen, Netherlands
| | - Jacques Vervoort
- Laboratory of Biochemistry, Wageningen University & Research, Wageningen, Netherlands
| | - Marjolein Meijerink
- Host-Microbe Interactomics Group, Wageningen University & Research, Wageningen, Netherlands
| | - Nico Taverne
- Host-Microbe Interactomics Group, Wageningen University & Research, Wageningen, Netherlands.,TiFN Food & Nutrition, Wageningen, Netherlands
| | | | - Peter A Bron
- TiFN Food & Nutrition, Wageningen, Netherlands.,NIZO Food Research, Ede, Netherlands
| | - Michiel Kleerebezem
- Host-Microbe Interactomics Group, Wageningen University & Research, Wageningen, Netherlands.,TiFN Food & Nutrition, Wageningen, Netherlands
| |
Collapse
|
41
|
Lipoprotein N-Acylation in Staphylococcus aureus Is Catalyzed by a Two-Component Acyl Transferase System. mBio 2020; 11:mBio.01619-20. [PMID: 32723923 PMCID: PMC7387801 DOI: 10.1128/mbio.01619-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although it has long been known that S. aureus forms triacylated Lpps, a lack of homologs to known N-acylation genes found in Gram-negative bacteria has until now precluded identification of the genes responsible for this Lpp modification. Here, we demonstrate N-terminal Lpp acylation and chemotype conversion to the tri-acylated state is directed by a unique acyl transferase system encoded by two noncontiguous staphylococci genes (lnsAB). Since triacylated Lpps stimulate TLR2 more weakly than their diacylated counterparts, Lpp N-acylation is an important TLR2 immunoevasion factor for determining tolerance or nontolerance in niches such as in the skin microbiota. The discovery of the LnsAB system expands the known diversity of Lpp biosynthesis pathways and acyl transfer biochemistry in bacteria, advances our understanding of Lpp structural heterogeneity, and helps differentiate commensal and noncommensal microbiota. Bacterial lipoproteins (Lpps) are a class of membrane-associated proteins universally distributed among all bacteria. A characteristic N-terminal cysteine residue that is variably acylated anchors C-terminal globular domains to the extracellular surface, where they serve numerous roles, including in the capture and transport of essential nutrients. Lpps are also ligands for the Toll-like receptor 2 (TLR2) family, a key component of the innate immune system tasked with bacterial recognition. While Lpp function is conserved in all prokaryotes, structural heterogeneity in the N-terminal acylation state is widespread among Firmicutes and can differ between otherwise closely related species. In this study, we identify a novel two-gene system that directs the synthesis of N-acylated Lpps in the commensal and opportunistic pathogen subset of staphylococci. The two genes, which we have named the lipoprotein N-acylation transferase system (Lns), bear no resemblance to previously characterized N-terminal Lpp tailoring enzymes. LnsA (SAOUHSC_00822) is an NlpC/P60 superfamily enzyme, whereas LnsB (SAOHSC_02761) has remote homology to the CAAX protease and bacteriocin-processing enzyme (CPBP) family. Both LnsA and LnsB are together necessary and alone sufficient for N-acylation in Staphylococcus aureus and convert the Lpp chemotype from diacyl to triacyl when heterologously expressed in Listeria monocytogenes. Acquisition of lnsAB decreases TLR2-mediated detection of S. aureus by nearly 10-fold and shifts the activated TLR2 complex from TLR2/6 to TLR2/1. LnsAB thus has a dual role in attenuating TLR2 signaling in addition to a broader role in bacterial cell envelope physiology.
Collapse
|
42
|
Belikova D, Jochim A, Power J, Holden MTG, Heilbronner S. "Gene accordions" cause genotypic and phenotypic heterogeneity in clonal populations of Staphylococcus aureus. Nat Commun 2020; 11:3526. [PMID: 32665571 PMCID: PMC7360770 DOI: 10.1038/s41467-020-17277-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 06/15/2020] [Indexed: 12/18/2022] Open
Abstract
Gene tandem amplifications are thought to drive bacterial evolution, but they are transient in the absence of selection, making their investigation challenging. Here, we analyze genomic sequences of Staphylococcus aureus USA300 isolates from the same geographical area to identify variations in gene copy number, which we confirm by long-read sequencing. We find several hotspots of variation, including the csa1 cluster encoding lipoproteins known to be immunogenic. We also show that the csa1 locus expands and contracts during bacterial growth in vitro and during systemic infection of mice, and recombination creates rapid heterogeneity in initially clonal cultures. Furthermore, csa1 copy number variants differ in their immunostimulatory capacity, revealing a mechanism by which gene copy number variation can modulate the host immune response. Gene tandem amplifications can drive bacterial evolution. Here, Belikova et al. identify copy number variations of lipoprotein-encoding genes in Staphylococcus aureus clinical isolates, and show that the loci expand and contract during bacterial growth in vitro and in mice, leading to changes in immunostimulatory capacity.
Collapse
Affiliation(s)
- Darya Belikova
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
| | - Angelika Jochim
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
| | - Jeffrey Power
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
| | | | - Simon Heilbronner
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany. .,German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany. .,(DFG) Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, Tübingen, Germany.
| |
Collapse
|
43
|
Bleuzé M, Auger JP, Lavagna A, Gisch N, Gottschalk M, Segura M. In vitro characterization of granulocyte-colony stimulating factor (G-CSF) production by dendritic cells and macrophages during Streptococcus suis infection. Immunobiology 2020; 225:151979. [PMID: 32747024 DOI: 10.1016/j.imbio.2020.151979] [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: 05/07/2020] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 12/26/2022]
Abstract
Streptococcus suis serotype 2 is an important porcine bacterial pathogen and emerging zoonotic agent. Infections induce an exacerbated inflammation that can result in sudden death (septic shock) and meningitis. Though neutrophilic leukocytosis characterizes S. suis infection, the mediators involved are poorly understood. Among them, granulocyte-colony stimulating factor (G-CSF), a pro-inflammatory cytokine, triggers proliferation of neutrophil progenitors and neutrophil mobilization. However, the systemic production of G-CSF induced during S. suis infection, the cell types involved, and the underlying mechanisms remain unknown. In a S. suis serotype 2 mouse model of systemic infection, plasma levels of G-CSF rapidly increased after infection. S. suis activation of DCs and macrophages resulted in high (> 1000 pg/mL) and comparable production levels of G-CSF, as measured by ELISA. By using mutant strains deficient in capsular polysaccharide (CPS) or lipoprotein maturation in combination with purified lipoteichoic acid (LTA) from the latter mutant strain, it was showed that G-CSF production is mainly mediated by S. suis lipoproteins. The Toll-like receptor (TLR) pathway via myeloid differentiation primary response 88 (MyD88) is required for G-CSF production by DCs and macrophages following S. suis activation, with a partial involvement of TLR2. On the other hand, TLR2-independant G-CSF production induced by S. suis requires internalization and bacterial DNA might play a role in this pathway. Finally, these signals activated nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways leading to G-CSF production. In conclusion, this study demonstrated for the first time that S. suis induces G-CSF production in vivo and DCs and macrophages are key cellular sources of this cytokine mediator, mainly via the binding of lipoproteins to TLR2. The CPS significantly reduced this activation, confirming the powerful role of this component in S. suis virulence. As such, this study contributes to better understand how DCs and macrophages produce G-CSF in response to S. suis, and potentially to other streptococci.
Collapse
Affiliation(s)
- Marêva Bleuzé
- Research Group on Infectious Diseases in Production Animals (GREMIP) & Swine and Poultry Infectious Diseases Research Center (CRIPA), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, J2S 2M2, QC, Canada
| | - Jean-Philippe Auger
- Research Group on Infectious Diseases in Production Animals (GREMIP) & Swine and Poultry Infectious Diseases Research Center (CRIPA), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, J2S 2M2, QC, Canada
| | - Agustina Lavagna
- Research Group on Infectious Diseases in Production Animals (GREMIP) & Swine and Poultry Infectious Diseases Research Center (CRIPA), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, J2S 2M2, QC, Canada
| | - Nicolas Gisch
- Division of Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Marcelo Gottschalk
- Research Group on Infectious Diseases in Production Animals (GREMIP) & Swine and Poultry Infectious Diseases Research Center (CRIPA), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, J2S 2M2, QC, Canada
| | - Mariela Segura
- Research Group on Infectious Diseases in Production Animals (GREMIP) & Swine and Poultry Infectious Diseases Research Center (CRIPA), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, J2S 2M2, QC, Canada.
| |
Collapse
|
44
|
Mohammad M, Hu Z, Ali A, Kopparapu PK, Na M, Jarneborn A, Stroparo MDN, Nguyen MT, Karlsson A, Götz F, Pullerits R, Jin T. The role of Staphylococcus aureus lipoproteins in hematogenous septic arthritis. Sci Rep 2020; 10:7936. [PMID: 32404866 PMCID: PMC7221087 DOI: 10.1038/s41598-020-64879-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/23/2020] [Indexed: 01/31/2023] Open
Abstract
Permanent joint dysfunction is a devastating complication in patients with septic arthritis. Staphylococcus aureus (S. aureus) lipoproteins (Lpp), the predominant ligands for TLR2, are known to be arthritogenic and induce bone destruction when introduced directly into the joint. Here, we aim to investigate the importance of S. aureus Lpp and TLR2 in a hematogenous septic arthritis model, which is the most common route of infection in humans. C57BL/6 wild-type and TLR2 deficient mice were intravenously inoculated with S. aureus Newman parental strain or its lipoprotein-deficient Δlgt mutant strain. The clinical course of septic arthritis, radiological changes, and serum levels of cytokines and chemokines, were assessed. Newman strain induced more severe and frequent clinical septic polyarthritis compared to its Δlgt mutant in TLR2 deficient mice, but not in wild-type controls. Bone destruction, however, did not differ between groups. Lpp expression was associated with higher mortality, weight loss as well as impaired bacterial clearance in mouse kidneys independent of TLR2. Furthermore, Lpp expression induced increased systemic pro-inflammatory cytokine and neutrophil chemokine release. Staphylococcal Lpp are potent virulence factors in S. aureus systemic infection independent of host TLR2 signalling. However, they have a limited impact on bone erosion in hematogenous staphylococcal septic arthritis.
Collapse
Affiliation(s)
- Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Zhicheng Hu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Microbiology and Immunology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Abukar Ali
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pradeep Kumar Kopparapu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Manli Na
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Jarneborn
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mariana do Nascimento Stroparo
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Minh-Thu Nguyen
- Department of Microbial Genetics, University of Tübingen, Tübingen, Germany
| | - Anna Karlsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Friedrich Götz
- Department of Microbial Genetics, University of Tübingen, Tübingen, Germany
| | - Rille Pullerits
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
45
|
Bitschar K, Staudenmaier L, Klink L, Focken J, Sauer B, Fehrenbacher B, Herster F, Bittner Z, Bleul L, Schaller M, Wolz C, Weber AN, Peschel A, Schittek B. Staphylococcus aureus Skin Colonization Is Enhanced by the Interaction of Neutrophil Extracellular Traps with Keratinocytes. J Invest Dermatol 2020; 140:1054-1065.e4. [DOI: 10.1016/j.jid.2019.10.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 11/30/2022]
|
46
|
Mlynek KD, Bulock LL, Stone CJ, Curran LJ, Sadykov MR, Bayles KW, Brinsmade SR. Genetic and Biochemical Analysis of CodY-Mediated Cell Aggregation in Staphylococcus aureus Reveals an Interaction between Extracellular DNA and Polysaccharide in the Extracellular Matrix. J Bacteriol 2020; 202:e00593-19. [PMID: 32015143 PMCID: PMC7099133 DOI: 10.1128/jb.00593-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 01/24/2020] [Indexed: 12/14/2022] Open
Abstract
The global regulator CodY links nutrient availability to the regulation of virulence factor gene expression in Staphylococcus aureus, including many genes whose products affect biofilm formation. Antithetical phenotypes of both biofilm deficiency and accumulation have been reported for codY-null mutants; thus, the role of CodY in biofilm development remains unclear. codY mutant cells of a strain producing a robust biofilm elaborate proaggregation surface-associated features not present on codY mutant cells that do not produce a robust biofilm. Biochemical analysis of the clinical isolate SA564, which aggregates when deficient for CodY, revealed that these features are sensitive to nuclease treatment and are resistant to protease exposure. Genetic analyses revealed that disrupting lgt (the diacylglycerol transferase gene) in codY mutant cells severely weakened aggregation, indicating a role for lipoproteins in the attachment of the biofilm matrix to the cell surface. An additional and critical role of IcaB in producing functional poly-N-acetylglucosamine (PIA) polysaccharide in extracellular DNA (eDNA)-dependent biofilm formation was shown. Moreover, overproducing PIA is sufficient to promote aggregation in a DNA-dependent manner regardless of source of nucleic acids. Taken together, our results point to PIA synthesis as the primary determinant of biofilm formation when CodY activity is reduced and suggest a modified electrostatic net model for matrix attachment whereby PIA associates with eDNA, which interacts with the cell surface via covalently attached membrane lipoproteins. This work counters the prevailing view that polysaccharide- and eDNA/protein-based biofilms are mutually exclusive. Rather, we demonstrate that eDNA and PIA can work synergistically to form a biofilm.IMPORTANCEStaphylococcus aureus remains a global health concern and exemplifies the ability of an opportunistic pathogen to adapt and persist within multiple environments, including host tissue. Not only does biofilm contribute to persistence and immune evasion in the host environment, it also may aid in the transition to invasive disease. Thus, understanding how biofilms form is critical for developing strategies for dispersing biofilms and improving biofilm disease-related outcomes. Using biochemical, genetic, and cell biology approaches, we reveal a synergistic interaction between PIA and eDNA that promotes cell aggregation and biofilm formation in a CodY-dependent manner in S. aureus We also reveal that envelope-associated lipoproteins mediate attachment of the biofilm matrix to the cell surface.
Collapse
Affiliation(s)
- Kevin D Mlynek
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Logan L Bulock
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Carl J Stone
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Luke J Curran
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Marat R Sadykov
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Kenneth W Bayles
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | |
Collapse
|
47
|
Recognition of Lipoproteins by Toll-like Receptor 2 and DNA by the AIM2 Inflammasome Is Responsible for Production of Interleukin-1β by Virulent Suilysin-negative Streptococcus suis Serotype 2. Pathogens 2020; 9:pathogens9020147. [PMID: 32098284 PMCID: PMC7168628 DOI: 10.3390/pathogens9020147] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/20/2022] Open
Abstract
Streptococcus suis serotype 2 is an important porcine bacterial pathogen and zoonotic agent causing sudden death, septic shock and meningitis. These pathologies are the consequence of an exacerbated inflammatory response composed of various mediators including interleukin (IL)-1β. Elevated levels of the toxin suilysin (SLY) were demonstrated to play a key role in S. suis-induced IL-1β production. However, 95% of serotype 2 strains isolated from diseased pigs in North America, many of which are virulent, do not produce SLY. In this study, we demonstrated that SLY-negative S. suis induces elevated levels of IL-1β in systemic organs, with dendritic cells contributing to this production. SLY-negative S. suis-induced IL-1β production requires MyD88 and TLR2 following recognition of lipoproteins. However, the higher internalization rate of the SLY-negative strain results in intracellularly located DNA being recognized by the AIM2 inflammasome, which promotes IL-1β production. Finally, the role of IL-1 in host survival during the S. suis systemic infection is beneficial and conserved, regardless of SLY production, via modulation of the inflammation required to control bacterial burden. In conclusion, this study demonstrates that SLY is not required for S. suis-induced IL-1β production.
Collapse
|
48
|
Wang X, Eagen WJ, Lee JC. Orchestration of human macrophage NLRP3 inflammasome activation by Staphylococcus aureus extracellular vesicles. Proc Natl Acad Sci U S A 2020; 117:3174-3184. [PMID: 31988111 PMCID: PMC7022218 DOI: 10.1073/pnas.1915829117] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Release of extracellular vesicles (EVs) is a common feature among eukaryotes, archaea, and bacteria. However, the biogenesis and downstream biological effects of EVs released from gram-positive bacteria remain poorly characterized. Here, we report that EVs purified from a community-associated methicillin-resistant Staphylococcus aureus strain were internalized into human macrophages in vitro and that this process was blocked by inhibition of the dynamin-dependent endocytic pathway. Human macrophages responded to S. aureus EVs by TLR2 signaling and activation of NLRP3 inflammasomes through K+ efflux, leading to the recruitment of ASC and activation of caspase-1. Cleavage of pro-interleukin (IL)-1β, pro-IL-18, and gasdermin-D by activated caspase-1 resulted in the cellular release of the mature cytokines IL-1β and IL-18 and induction of pyroptosis. Consistent with this result, a dose-dependent cytokine response was detected in the extracellular fluids of mice challenged intraperitoneally with S. aureus EVs. Pore-forming toxins associated with S. aureus EVs were critical for NLRP3-dependent caspase-1 activation of human macrophages, but not for TLR2 signaling. In contrast, EV-associated lipoproteins not only mediated TLR2 signaling to initiate the priming step of NLRP3 activation but also modulated EV biogenesis and the toxin content of EVs, resulting in alterations in IL-1β, IL-18, and caspase-1 activity. Collectively, our study describes mechanisms by which S. aureus EVs induce inflammasome activation and reveals an unexpected role of staphylococcal lipoproteins in EV biogenesis. EVs may serve as a novel secretory pathway for S. aureus to transport protected cargo in a concentrated form to host cells during infections to modulate cellular functions.
Collapse
Affiliation(s)
- Xiaogang Wang
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - William J Eagen
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Jean C Lee
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
49
|
Yue Y, Zhou T, Xu X, Sun Q, Wang C, Zhu J, Zheng F. Influence of transcription regulator SAUSA300_1968 on the virulence protein secretion and immune evasion by Staphylococcus aureus. Microb Pathog 2019; 136:103690. [DOI: 10.1016/j.micpath.2019.103690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/21/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022]
|
50
|
Musilova J, Mulcahy ME, Kuijk MM, McLoughlin RM, Bowie AG. Toll-like receptor 2-dependent endosomal signaling by Staphylococcus aureus in monocytes induces type I interferon and promotes intracellular survival. J Biol Chem 2019; 294:17031-17042. [PMID: 31558608 PMCID: PMC6851302 DOI: 10.1074/jbc.ra119.009302] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/24/2019] [Indexed: 12/11/2022] Open
Abstract
Pathogen activation of innate immune pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs) stimulates cellular signaling pathways. This often leads to outcomes that contribute to pathogen clearance. Alternatively, activation of specific PRR pathways can aid pathogen survival. The human pathogen Staphylococcus aureus is a case in point, employing strategies to escape innate immune recognition and killing by the host. As for other bacteria, PRR-stimulated type I interferon (IFN-I) induction has been proposed as one such immune escape pathway that may favor S. aureus. Cell wall components of S. aureus elicit TLR2-dependent cellular responses, but the exact signaling pathways activated by S. aureus–TLR2 engagement and the consequences of their activation for the host and bacterium are not fully known. We previously showed that TLR2 activates both a cytoplasmic and an endosome-dependent signaling pathway, the latter leading to IFN-I production. Here, we demonstrate that S. aureus infection of human monocytes activates a TLR2-dependent endosomal signaling pathway, leading to IFN-I induction. We mapped the signaling components of this pathway and identified roles in IFN-I stimulation for the Toll-interleukin-1 receptor (TIR) adaptor Myd88 adaptor-like (Mal), TNF receptor-associated factor 6 (TRAF6), and IκB kinase (IKK)-related kinases, but not for TRIF-related adaptor molecule (TRAM) and TRAF3. Importantly, monocyte TLR2-dependent endosomal signaling enabled immune escape for S. aureus, because this pathway, but not IFN-I per se, contributed to intracellular bacterial survival. These results reveal a TLR2-dependent mechanism in human monocytes whereby S. aureus manipulates innate immune signaling for its survival in cells.
Collapse
Affiliation(s)
- Jana Musilova
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Michelle E Mulcahy
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Marieke M Kuijk
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Rachel M McLoughlin
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|