1
|
Li H, Bradbury JA, Edin ML, Gruzdev A, Li H, Graves JP, DeGraff LM, Lih FB, Feng C, Wolf ER, Bortner CD, London SJ, Sparks MA, Coffman TM, Zeldin DC. TXA2 attenuates allergic lung inflammation through regulation of Th2, Th9, and Treg differentiation. J Clin Invest 2024; 134:e165689. [PMID: 38483511 PMCID: PMC11060738 DOI: 10.1172/jci165689] [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: 01/03/2023] [Accepted: 03/12/2024] [Indexed: 05/02/2024] Open
Abstract
In lung, thromboxane A2 (TXA2) activates the TP receptor to induce proinflammatory and bronchoconstrictor effects. Thus, TP receptor antagonists and TXA2 synthase inhibitors have been tested as potential asthma therapeutics in humans. Th9 cells play key roles in asthma and regulate the lung immune response to allergens. Herein, we found that TXA2 reduces Th9 cell differentiation during allergic lung inflammation. Th9 cells were decreased approximately 2-fold and airway hyperresponsiveness was attenuated in lungs of allergic mice treated with TXA2. Naive CD4+ T cell differentiation to Th9 cells and IL-9 production were inhibited dose-dependently by TXA2 in vitro. TP receptor-deficient mice had an approximately 2-fold increase in numbers of Th9 cells in lungs in vivo after OVA exposure compared with wild-type mice. Naive CD4+ T cells from TP-deficient mice exhibited increased Th9 cell differentiation and IL-9 production in vitro compared with CD4+ T cells from wild-type mice. TXA2 also suppressed Th2 and enhanced Treg differentiation both in vitro and in vivo. Thus, in contrast to its acute, proinflammatory effects, TXA2 also has longer-lasting immunosuppressive effects that attenuate the Th9 differentiation that drives asthma progression. These findings may explain the paradoxical failure of anti-thromboxane therapies in the treatment of asthma.
Collapse
Affiliation(s)
- Hong Li
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - J. Alyce Bradbury
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Matthew L. Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Artiom Gruzdev
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Huiling Li
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Joan P. Graves
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Laura M. DeGraff
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Fred B. Lih
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Chiguang Feng
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Erin R. Wolf
- Department of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
| | - Carl D. Bortner
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Stephanie J. London
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Matthew A. Sparks
- Department of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
| | - Thomas M. Coffman
- Department of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore
| | - Darryl C. Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| |
Collapse
|
2
|
Battaglia M, Garrett-Sinha LA. Staphylococcus xylosus and Staphylococcus aureus as commensals and pathogens on murine skin. Lab Anim Res 2023; 39:18. [PMID: 37533118 PMCID: PMC10394794 DOI: 10.1186/s42826-023-00169-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023] Open
Abstract
Skin ulcers, skin dermatitis and skin infections are common phenomena in colonies of laboratory mice and are often found at increased prevalence in certain immunocompromised strains. While in many cases these skin conditions are mild, in other cases they can be severe and lead to animal morbidity. Furthermore, the presence of skin infections and ulcerations can complicate the interpretation of experimental protocols, including those examining immune cell activation. Bacterial species in the genus Staphylococcus are the most common pathogens recovered from skin lesions in mice. In particular, Staphylococcus aureus and Staphylococcus xylosus have both been implicated as pathogens on murine skin. Staphylococcus aureus is a well-known pathogen of human skin, but S. xylosus skin infections in humans have not been described, indicating that there is a species-specific difference in the ability of S. xylosus to serve as a skin pathogen. The aim of this review is to summarize studies that link S. aureus and S. xylosus to skin infections of mice and to describe factors involved in their adherence to tissue and their virulence. We discuss potential differences in mouse and human skin that might underlie the ability of S. xylosus to act as a pathogen on murine skin, but not human skin. Finally, we also describe mouse mutants that have shown increased susceptibility to skin infections with staphylococcal bacteria. These mutants point to pathways that are important in the control of commensal staphylococcal bacteria. The information here may be useful to researchers who are working with mouse strains that are prone to skin infections with staphylococcal bacteria.
Collapse
Affiliation(s)
- Michael Battaglia
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Lee Ann Garrett-Sinha
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
3
|
Butrico CE, Klopfenstein N, Green ER, Johnson JR, Peck SH, Ibberson CB, Serezani CH, Cassat JE. Hyperglycemia Increases Severity of Staphylococcus aureus Osteomyelitis and Influences Bacterial Genes Required for Survival in Bone. Infect Immun 2023; 91:e0052922. [PMID: 36877063 PMCID: PMC10112148 DOI: 10.1128/iai.00529-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/13/2023] [Indexed: 03/07/2023] Open
Abstract
Hyperglycemia, or elevated blood glucose, renders individuals more prone to developing severe Staphylococcus aureus infections. S. aureus is the most common etiological agent of musculoskeletal infection, which is a common manifestation of disease in hyperglycemic patients. However, the mechanisms by which S. aureus causes severe musculoskeletal infection during hyperglycemia are incompletely characterized. To examine the influence of hyperglycemia on S. aureus virulence during invasive infection, we used a murine model of osteomyelitis and induced hyperglycemia with streptozotocin. We discovered that hyperglycemic mice exhibited increased bacterial burdens in bone and enhanced dissemination compared to control mice. Furthermore, infected hyperglycemic mice sustained increased bone destruction relative to euglycemic controls, suggesting that hyperglycemia exacerbates infection-associated bone loss. To identify genes contributing to S. aureus pathogenesis during osteomyelitis in hyperglycemic animals relative to euglycemic controls, we used transposon sequencing (TnSeq). We identified 71 genes uniquely essential for S. aureus survival in osteomyelitis in hyperglycemic mice and another 61 mutants with compromised fitness. Among the genes essential for S. aureus survival in hyperglycemic mice was the gene encoding superoxide dismutase A (sodA), one of two S. aureus superoxide dismutases involved in detoxifying reactive oxygen species (ROS). We determined that a sodA mutant exhibits attenuated survival in vitro in high glucose and in vivo during osteomyelitis in hyperglycemic mice. SodA therefore plays an important role during growth in high glucose and promotes S. aureus survival in bone. Collectively, these studies demonstrate that hyperglycemia increases the severity of osteomyelitis and identify genes contributing to S. aureus survival during hyperglycemic infection.
Collapse
Affiliation(s)
- Casey E. Butrico
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nathan Klopfenstein
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Erin R. Green
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Joshua R. Johnson
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sun H. Peck
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Nashville VA Medical Center, Department of Veterans Affairs, Nashville, Tennessee, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Carolyn B. Ibberson
- Department of Microbiology and Plant Biology, The University of Oklahoma, Norman, Oklahoma, USA
| | - C. Henrique Serezani
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James E. Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
4
|
Lin S, Wang Q, Huang X, Feng J, Wang Y, Shao T, Deng X, Cao Y, Chen X, Zhou M, Zhao C. Wounds under diabetic milieu: The role of immune cellar components and signaling pathways. Biomed Pharmacother 2023; 157:114052. [PMID: 36462313 DOI: 10.1016/j.biopha.2022.114052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/19/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
A major challenge in the field of diabetic wound healing is to confirm the body's intrinsic mechanism that could sense the immune system damage promptly and protect the wound from non-healing. Accumulating literature indicates that macrophage, a contributor to prolonged inflammation occurring at the wound site, might play such a role in hindering wound healing. Likewise, other immune cell dysfunctions, such as persistent neutrophils and T cell infection, may also lead to persistent oxidative stress and inflammatory reaction during diabetic wound healing. In this article, we discuss recent advances in the immune cellular components in wounds under the diabetic milieu, and the role of key signaling mechanisms that compromise the function of immune cells leading to persistent wound non-healing.
Collapse
Affiliation(s)
- Siyuan Lin
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qixue Wang
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaoting Huang
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Jiawei Feng
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Yuqing Wang
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Tengteng Shao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Xiaofei Deng
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Yemin Cao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Xinghua Chen
- Jinshan Hospital Affiliated to Fudan University, Shanghai, China.
| | - Mingmei Zhou
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Cheng Zhao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China.
| |
Collapse
|
5
|
Ju M, Joseph T, Hansanant N, Geng M, Williams M, Cothrell A, Buhrow AR, Austin F, Smith L. Evaluation of analogs of mutacin 1140 in systemic and cutaneous methicillin-resistant Staphylococcus aureus infection models in mice. Front Microbiol 2022; 13:1067410. [PMID: 36590413 PMCID: PMC9794991 DOI: 10.3389/fmicb.2022.1067410] [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: 10/11/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
Mutacin 1140 (Mu1140) is a potent antibiotic against Gram-positive bacteria, such as Staphylococcus aureus. The antibiotic is produced by the oral bacterium Streptococcus mutans and is a member of the epidermin family of type AI lantibiotics. The antibiotic exerts its inhibitory activity by binding to the cell wall precursor lipid II, blocking cell wall synthesis, and by disrupting bacterial membranes. In previous studies, the novel K2A and R13A analogs of Mu1140 have been identified to have superior pharmacokinetic properties compared to native Mu1140. In this study, the use of a combined formulation of the Mu1140 K2A and R13A analogs was shown to be more effective at treating MRSA bacteremia than the native Mu1140 or vancomycin. The analogs were also shown to be effective in treating an MRSA skin infection. The use of K2A and R13A analogs may provide a future alternative for treating serious Gram-positive bacterial infections. In a previous study, the Mu1140 analogs were shown to have significantly longer drug clearance times, leading to higher plasma concentrations over time. These properties warranted further testing to determine whether the analogs are effective for the treatment of systemic MRSA and acute skin infections. In this study, Mu1140 analogs were shown to be more effective than currently available treatments for systemic and skin MRSA infections. Further, the study clearly shows that the new analogs are superior to native Mu1140 for the treatment of a systemic MRSA infection. These findings support continued drug product development efforts using the K2A and R13A Mu1140 analogs, and that these analogs may ameliorate the outcome of serious bacterial infections.
Collapse
Affiliation(s)
- Min Ju
- Antimicrobial Division, Sano Chemicals Inc., Bryan, TX, United States
| | - Thushinari Joseph
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Nopakorn Hansanant
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Mengxin Geng
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - McKinley Williams
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Andrew Cothrell
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Andrew Riley Buhrow
- Antimicrobial Division, Sano Chemicals Inc., Bryan, TX, United States,Department of Biology, Texas A&M University, College Station, TX, United States
| | - Frank Austin
- College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, United States
| | - Leif Smith
- Antimicrobial Division, Sano Chemicals Inc., Bryan, TX, United States,Department of Biology, Texas A&M University, College Station, TX, United States,*Correspondence: Leif Smith,
| |
Collapse
|
6
|
Tamburini BAJ. Contributions of PD-L1 reverse signaling to dendritic cell trafficking. FEBS J 2022; 289:6256-6266. [PMID: 34146376 PMCID: PMC8684559 DOI: 10.1111/febs.16084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/18/2021] [Accepted: 06/18/2021] [Indexed: 12/27/2022]
Abstract
Programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) interactions are critical for dampening the immune response to both self and foreign antigens. The signaling of PD-L1 via its cytoplasmic domain, rather than through its interactions with PD-1 via the extracellular domain, has been termed PD-L1 reverse signaling. While this signaling is beneficial for cancer progression, little is understood about the consequences of PD-L1 reverse signaling in immune cells that express PD-L1 at steady state or in response to infection. Loss of PD-L1 during infection leads to unchecked T-cell proliferation and increased autoimmune T-cell responses. While the T-cell intrinsic role of PD-1 for inhibiting T-cell responses has been well explored, little to no effort has been directed at investigating the consequences of PD-L1 reverse signaling on the DCs interacting with PD-1+ T cells. We recently reported a defect in dendritic cell (DC) trafficking from the skin to the draining lymph node (LN) following immunization or infection in the absence of PD-L1. We demonstrated that a region within the cytoplasmic tail was responsible for the defect in DC trafficking. Here, we review the processes involved in DC trafficking and highlight what we know about PD-L1 expression, PD-L1 post-translational modifications, PD-L1 intracellular interactions, and PD-L1 extracellular interactions.
Collapse
Affiliation(s)
- Beth Ann Jirón Tamburini
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine Aurora, CO, USA
| |
Collapse
|
7
|
Lucas ED, Schafer JB, Matsuda J, Kraus M, Burchill MA, Tamburini BAJ. PD-L1 Reverse Signaling in Dermal Dendritic Cells Promotes Dendritic Cell Migration Required for Skin Immunity. Cell Rep 2021; 33:108258. [PMID: 33053342 PMCID: PMC7688291 DOI: 10.1016/j.celrep.2020.108258] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/06/2020] [Accepted: 09/21/2020] [Indexed: 12/26/2022] Open
Abstract
Although the function of the extracellular region of programmed death ligand 1 (PD-L1) through its interactions with PD-1 on T cells is well studied, little is understood regarding the intracellular domain of PD-L1. Here, we outline a major role for PD-L1 intracellular signaling in the control of dendritic cell (DC) migration from the skin to the draining lymph node (dLN). Using a mutant mouse model, we identify a TSS signaling motif within the intracellular domain of PD-L1. The TSS motif proves critical for chemokine-mediated DC migration to the dLN during inflammation. This loss of DC migration, in the PD-L1 TSS mutant, leads to a significant decline in T cell priming when DC trafficking is required for antigen delivery to the dLN. Finally, the TSS motif is required for chemokine receptor signaling downstream of the Gα subunit of the heterotrimeric G protein complex, ERK phosphorylation, and actin polymerization in DCs. Lucas et al. define three residues within the cytoplasmic tail of PD-L1 that are required for proper dendritic cell migration from the skin to the lymph node. These three-amino-acid residues promote chemokine signaling in dendritic cells and productive T cell responses to skin infections.
Collapse
Affiliation(s)
- Erin D Lucas
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Johnathon B Schafer
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA; Molecular Biology Program, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | | | - Madison Kraus
- Gates Summer Research Program, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Matthew A Burchill
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Beth A Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA; Molecular Biology Program, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA.
| |
Collapse
|
8
|
Klopfenstein N, Cassat JE, Monteith A, Miller A, Drury S, Skaar E, Serezani CH. Murine Models for Staphylococcal Infection. Curr Protoc 2021; 1:e52. [PMID: 33656290 PMCID: PMC7935403 DOI: 10.1002/cpz1.52] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Staphylococcus aureus is a Gram-positive bacterium that colonizes almost every organ in humans and mice and is a leading cause of diseases worldwide. S. aureus infections can be challenging to treat due to widespread antibiotic resistance and their ability to cause tissue damage. The primary modes of transmission of S. aureus are via direct contact with a colonized or infected individual or invasive spread from a colonization niche in the same individual. S. aureus can cause a myriad of diseases, including skin and soft tissue infections (SSTIs), osteomyelitis, pneumonia, endocarditis, and sepsis. S. aureus infection is characterized by the formation of purulent lesions known as abscesses, which are rich in live and dead neutrophils, macrophages, and surrounded by a capsule containing fibrin and collagen. Different strains of S. aureus produce varying amounts of toxins that evade and/or elicit immune responses. Therefore, animal models of S. aureus infection provide a unique opportunity to understand the dynamics of organ-specific immune responses and modifications in the pathogen that could favor the establishment of the pathogen. With advances in in vivo imaging of fluorescent transgenic mice, combined with fluorescent/bioluminescent bacteria, we can use mouse models to better understand the immune response to these types of infections. By understanding the host and bacterial dynamics within various organ systems, we can develop therapeutics to eliminate these pathogens. This module describes in vivo mouse models of both local and systemic S. aureus infection. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Murine model of Staphylococcus aureus subcutaneous infection Alternate Protocol: Murine tape stripping skin infection model Basic Protocol 2: Sample collection to determine skin structure, production of inflammatory mediators, and bacterial load Basic Protocol 3: Murine model of post-traumatic Staphylococcus aureus osteomyelitis Basic Protocol 4: Intravenous infection of the retro-orbital sinus Support Protocol: Preparation of the bacterial inoculum.
Collapse
Affiliation(s)
- Nathan Klopfenstein
- Division of Infectious Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - James E Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Andrew Monteith
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anderson Miller
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Sydney Drury
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Eric Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee
| | - C Henrique Serezani
- Division of Infectious Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
9
|
Klopfenstein N, Brandt SL, Castellanos S, Gunzer M, Blackman A, Serezani CH. SOCS-1 inhibition of type I interferon restrains Staphylococcus aureus skin host defense. PLoS Pathog 2021; 17:e1009387. [PMID: 33690673 PMCID: PMC7984627 DOI: 10.1371/journal.ppat.1009387] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/22/2021] [Accepted: 02/16/2021] [Indexed: 12/19/2022] Open
Abstract
The skin innate immune response to methicillin-resistant Staphylococcus aureus (MRSA) culminates in the formation of an abscess to prevent bacterial spread and tissue damage. Pathogen recognition receptors (PRRs) dictate the balance between microbial control and injury. Therefore, intracellular brakes are of fundamental importance to tune the appropriate host defense while inducing resolution. The intracellular inhibitor suppressor of cytokine signaling 1 (SOCS-1), a known JAK/STAT inhibitor, prevents the expression and actions of PRR adaptors and downstream effectors. Whether SOCS-1 is a molecular component of skin host defense remains to be determined. We hypothesized that SOCS-1 decreases type I interferon production and IFNAR-mediated antimicrobial effector functions, limiting the inflammatory response during skin infection. Our data show that MRSA skin infection enhances SOCS-1 expression, and both SOCS-1 inhibitor peptide-treated and myeloid-specific SOCS-1 deficient mice display decreased lesion size, bacterial loads, and increased abscess thickness when compared to wild-type mice treated with the scrambled peptide control. SOCS-1 deletion/inhibition increases phagocytosis and bacterial killing, dependent on nitric oxide release. SOCS-1 inhibition also increases the levels of type I and type II interferon levels in vivo. IFNAR deletion and antibody blockage abolished the beneficial effects of SOCS-1 inhibition in vivo. Notably, we unveiled that hyperglycemia triggers aberrant SOCS-1 expression that correlates with decreased overall IFN signatures in the infected skin. SOCS-1 inhibition restores skin host defense in the highly susceptible hyperglycemic mice. Overall, these data demonstrate a role for SOCS-1-mediated type I interferon actions in host defense and inflammation during MRSA skin infection.
Collapse
Affiliation(s)
- Nathan Klopfenstein
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Stephanie L Brandt
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Sydney Castellanos
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Hufelandstrasse Essen, Germany
- Leibniz-Institut für Analytische Wissenschaften-ISAS -e.V, Dortmund, Germany
| | - Amondrea Blackman
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - C Henrique Serezani
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
10
|
Salina ACG, Brandt SL, Klopfenstein N, Blackman A, Bazzano JMR, Sá-Nunes A, Byers-Glosson N, Brodskyn C, Tavares NM, Da Silva IBS, Medeiros AI, Serezani CH. Leukotriene B 4 licenses inflammasome activation to enhance skin host defense. Proc Natl Acad Sci U S A 2020; 117:30619-30627. [PMID: 33184178 PMCID: PMC7720147 DOI: 10.1073/pnas.2002732117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The initial production of inflammatory mediators dictates host defense as well as tissue injury. Inflammasome activation is a constituent of the inflammatory response by recognizing pathogen and host-derived products and eliciting the production of IL-1β and IL-18 in addition to inducing a type of inflammatory cell death termed "pyroptosis." Leukotriene B4 (LTB4) is a lipid mediator produced quickly (seconds to minutes) by phagocytes and induces chemotaxis, increases cytokine/chemokine production, and enhances antimicrobial effector functions. Whether LTB4 directly activates the inflammasome remains to be determined. Our data show that endogenously produced LTB4 is required for the expression of pro-IL-1β and enhances inflammasome assembly in vivo and in vitro. Furthermore, LTB4-mediated Bruton's tyrosine kinase (BTK) activation is required for inflammasome assembly in vivo as well for IL-1β-enhanced skin host defense. Together, these data unveil a new role for LTB4 in enhancing the expression and assembly of inflammasome components and suggest that while blocking LTB4 actions could be a promising therapeutic strategy to prevent inflammasome-mediated diseases, exogenous LTB4 can be used as an adjuvant to boost inflammasome-dependent host defense.
Collapse
Affiliation(s)
- Ana Carolina Guerta Salina
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Stephanie L Brandt
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-3082
| | - Nathan Klopfenstein
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Amondrea Blackman
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
| | | | - Anderson Sá-Nunes
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Nicole Byers-Glosson
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-3082
| | - Claudia Brodskyn
- Oswaldo Cruz Foundation, Gonçalo Moniz Institute, FIOCRUZ, Salvador 40296-710, Brazil
| | | | | | - Alexandra I Medeiros
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil
| | - C Henrique Serezani
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232;
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
11
|
Huitema L, Phillips T, Alexeev V, Tomic-Canic M, Pastar I, Igoucheva O. Intracellular escape strategies of Staphylococcus aureus in persistent cutaneous infections. Exp Dermatol 2020; 30:1428-1439. [PMID: 33179358 DOI: 10.1111/exd.14235] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/19/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022]
Abstract
Pathogenic invasion of Staphylococcus aureus is a major concern in patients with chronic skin diseases like atopic dermatitis (AD), epidermolysis bullosa (EB), or chronic diabetic foot and venous leg ulcers, and can result in persistent and life-threatening chronic non-healing wounds. Staphylococcus aureus is generally recognized as extracellular pathogens. However, S. aureus can also invade, hide and persist in skin cells to contribute to wound chronicity. The intracellular life cycle of S. aureus is currently incompletely understood, although published studies indicate that its intracellular escape strategies play an important role in persistent cutaneous infections. This review provides current scientific knowledge about the intracellular life cycle of S. aureus in skin cells, which can be classified into professional and non-professional antigen-presenting cells, and its strategies to escape adaptive defense mechanisms. First, we discuss phenotypic switch of S. aureus, which affects intracellular routing and degradation. This review also evaluates potential intracellular escape mechanism of S. aureus to avoid intracellular degradation and antigen presentation, preventing an immune response. Furthermore, we discuss potential drug targets that can interfere with the intracellular life cycle of S. aureus. Taken together, this review aimed to increase scientific understanding about the intracellular life cycle of S. aureus into skin cells and its strategies to evade the host immune response, information that is crucial to reduce pathogenic invasion and life-threatening persistence of S. aureus in chronic cutaneous infections.
Collapse
Affiliation(s)
- Leonie Huitema
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Taylor Phillips
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Vitali Alexeev
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Olga Igoucheva
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
12
|
Davis FM, Tsoi LC, Wasikowski R, denDekker A, Joshi A, Wilke C, Deng H, Wolf S, Obi A, Huang S, Billi AC, Robinson S, Lipinski J, Melvin WJ, Audu CO, Weidinger S, Kunkel SL, Smith A, Gudjonsson JE, Moore BB, Gallagher KA. Epigenetic regulation of the PGE2 pathway modulates macrophage phenotype in normal and pathologic wound repair. JCI Insight 2020; 5:138443. [PMID: 32879137 PMCID: PMC7526451 DOI: 10.1172/jci.insight.138443] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/29/2020] [Indexed: 12/19/2022] Open
Abstract
Macrophages are a primary immune cell involved in inflammation, and their cell plasticity allows for transition from an inflammatory to a reparative phenotype and is critical for normal tissue repair following injury. Evidence suggests that epigenetic alterations play a critical role in establishing macrophage phenotype and function during normal and pathologic wound repair. Here, we find in human and murine wound macrophages that cyclooxygenase 2/prostaglandin E2 (COX-2/PGE2) is elevated in diabetes and regulates downstream macrophage-mediated inflammation and host defense. Using single-cell RNA sequencing of human wound tissue, we identify increased NF-κB-mediated inflammation in diabetic wounds and show increased COX-2/PGE2 in diabetic macrophages. Further, we identify that COX-2/PGE2 production in wound macrophages requires epigenetic regulation of 2 key enzymes in the cytosolic phospholipase A2/COX-2/PGE2 (cPLA2/COX-2/PGE2) pathway. We demonstrate that TGF-β-induced miRNA29b increases COX-2/PGE2 production via inhibition of DNA methyltransferase 3b-mediated hypermethylation of the Cox-2 promoter. Further, we find mixed-lineage leukemia 1 (MLL1) upregulates cPLA2 expression and drives COX-2/PGE2. Inhibition of the COX-2/PGE2 pathway genetically (Cox2fl/fl Lyz2Cre+) or with a macrophage-specific nanotherapy targeting COX-2 in tissue macrophages reverses the inflammatory macrophage phenotype and improves diabetic tissue repair. Our results indicate the epigenetically regulated PGE2 pathway controls wound macrophage function, and cell-targeted manipulation of this pathway is feasible to improve diabetic wound repair.
Collapse
Affiliation(s)
- Frank M Davis
- Section of Vascular Surgery, Department of Surgery.,Department of Microbiology and Immunology
| | | | | | | | - Amrita Joshi
- Section of Vascular Surgery, Department of Surgery
| | - Carol Wilke
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Hongping Deng
- Department of Bioengineering, University of Illinois, Champaign, Illinois, USA
| | - Sonya Wolf
- Section of Vascular Surgery, Department of Surgery
| | - Andrea Obi
- Section of Vascular Surgery, Department of Surgery
| | - Steven Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | - Jay Lipinski
- Section of Vascular Surgery, Department of Surgery
| | | | | | - Stephan Weidinger
- Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Steven L Kunkel
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Andrew Smith
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Bethany B Moore
- Department of Microbiology and Immunology.,Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Katherine A Gallagher
- Section of Vascular Surgery, Department of Surgery.,Department of Microbiology and Immunology
| |
Collapse
|
13
|
Mirzaei R, Ranjbar R, Karampoor S, Goodarzi R, Hasanvand H. The Human Immune System toward Staphylococcus aureus. Open Microbiol J 2020. [DOI: 10.2174/1874285802014010164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The immune system is responsible for protecting the host from pathogens, and it has evolved to deal with these pathogens. On the other hand, the co-evolution of pathogenic bacteria with hosts has led to the rise of an array of virulence genes that enable pathogen bacteria to evade or modulate the immune system. Staphylococcus aureus is a significant pathogen of humans that encodes several virulence factors that can modulate or evade from the innate and adaptive arm of the immune system. Overall, the immune reaction toward S. aureus contributes to stimulate innate and adaptive reactions. A profound understanding of the immune response to S. aureus infections will be critical for the development of vaccines and novel therapies. In this review, we summarized and discussed the novel information about the human immune system against S. aureus.
Collapse
|
14
|
Shivakoti R, Dalli J, Kadam D, Gaikwad S, Barthwal M, Colas RA, Mazzacuva F, Lokhande R, Dharmshale S, Bharadwaj R, Kagal A, Pradhan N, Deshmukh S, Atre S, Sahasrabudhe T, Kakrani A, Kulkarni V, Raskar S, Suryavanshi N, Chon S, Gupte A, Gupta A, Gupte N, Arriaga MB, Fukutani KF, Andrade BB, Golub JE, Mave V. Lipid mediators of inflammation and Resolution in individuals with tuberculosis and tuberculosis-Diabetes. Prostaglandins Other Lipid Mediat 2019; 147:106398. [PMID: 31726221 DOI: 10.1016/j.prostaglandins.2019.106398] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 10/25/2022]
Abstract
Individuals with concurrent tuberculosis (TB) and Type 2 diabetes (DM) have a higher risk of adverse outcomes. To better understand potential immunological differences, we utilized a comprehensive panel to characterize pro-inflammatory and pro-resolving (i.e., mediators involved in the resolution of inflammation) lipid mediators in individuals with TB and TB-DM. A nested cross-sectional study of 40 individuals (20 newly diagnosed DM and 20 without DM) was conducted within a cohort of individuals with active drug-susceptible treatment-naïve pulmonary TB. Lipid mediators were quantified in serum samples through lipid mediator profiling. We conducted correlation-based analysis of these mediators. Overall, the arachidonic acid-derived leukotriene and prostaglandin families were the most abundant pro-inflammatory lipid mediators, while lipoxins and maresins families were the most abundant pro-resolving lipid mediators in individuals with TB and TB-DM. Individuals with TB-DM had increased correlations and connectivity with both pro-inflammatory and pro-resolving lipid mediators compared to those with TB alone. We identified the most abundant lipid mediator metabolomes in circulation among individuals with TB and TB-DM; in addition, our data shows a substantial number of significant correlations between both pro-inflammatory and pro-resolving lipid mediators in individuals with TB-DM, delineating a molecular balance that potentially defines this comorbidity.
Collapse
Affiliation(s)
- Rupak Shivakoti
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Columbia University Mailman School of Public Health, New York, NY, USA.
| | - Jesmond Dalli
- William Harvey Research Institute, Queens Mary University of London, London, UK
| | - Dileep Kadam
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India; Byramjee-Jeejeebhoy Government Medical College, Pune, India
| | - Sanjay Gaikwad
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India; Byramjee-Jeejeebhoy Government Medical College, Pune, India
| | - Madhusudan Barthwal
- Dr. D.Y. Patil Medical College, Hospital & Research Centre, Dr. D.Y. Patil Vidyapeeth, Pune, India
| | - Romain A Colas
- William Harvey Research Institute, Queens Mary University of London, London, UK
| | - Francesca Mazzacuva
- William Harvey Research Institute, Queens Mary University of London, London, UK
| | - Rahul Lokhande
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India; Byramjee-Jeejeebhoy Government Medical College, Pune, India
| | - Sujata Dharmshale
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India; Byramjee-Jeejeebhoy Government Medical College, Pune, India
| | - Renu Bharadwaj
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India; Byramjee-Jeejeebhoy Government Medical College, Pune, India
| | - Anju Kagal
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India; Byramjee-Jeejeebhoy Government Medical College, Pune, India
| | - Neeta Pradhan
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - Sona Deshmukh
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - Sachin Atre
- Dr. D.Y. Patil Medical College, Hospital & Research Centre, Dr. D.Y. Patil Vidyapeeth, Pune, India
| | - Tushar Sahasrabudhe
- Dr. D.Y. Patil Medical College, Hospital & Research Centre, Dr. D.Y. Patil Vidyapeeth, Pune, India
| | - Arjun Kakrani
- Dr. D.Y. Patil Medical College, Hospital & Research Centre, Dr. D.Y. Patil Vidyapeeth, Pune, India
| | - Vandana Kulkarni
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - Swapnil Raskar
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - Nishi Suryavanshi
- Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - Sandy Chon
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Akshay Gupte
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amita Gupta
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - Nikhil Gupte
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| | - María B Arriaga
- Instituto Goncalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil; Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil; Multinational Organization Network Sponsoring Translational and Epidemiological Research, Fundação José Silveira, Salvador, Brazil
| | - Kiyoshi F Fukutani
- Instituto Goncalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil; Multinational Organization Network Sponsoring Translational and Epidemiological Research, Fundação José Silveira, Salvador, Brazil; Curso de Medicina, Faculdade de Tecnologia e Ciências, Salvador, Brazil
| | - Bruno B Andrade
- Instituto Goncalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil; Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil; Multinational Organization Network Sponsoring Translational and Epidemiological Research, Fundação José Silveira, Salvador, Brazil; Curso de Medicina, Faculdade de Tecnologia e Ciências, Salvador, Brazil; Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Brazil; Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil
| | - Jonathan E Golub
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Vidya Mave
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Byramjee-Jeejeebhoy Medical College-Johns Hopkins University Clinical Research Site, Pune, India
| |
Collapse
|
15
|
Chastain CA, Klopfenstein N, Serezani CH, Aronoff DM. A Clinical Review of Diabetic Foot Infections. Clin Podiatr Med Surg 2019; 36:381-395. [PMID: 31079605 DOI: 10.1016/j.cpm.2019.02.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
"Diabetic foot infections (DFIs) are a common cause of morbidity and mortality. This article summarizes current knowledge regarding DFI epidemiology, disease pathogenesis, and the impact of antimicrobial resistance among DFI. An evidence-based approach to clinical assessment, diagnosing osteomyelitis, as well as medical and surgical treatment is discussed, including a review of empiric and directed antibiotic treatment recommendations. The current state and needs of the clinical literature are identified throughout, with a discussion of the supporting role of infectious diseases specialists as well as future directions of the field."
Collapse
Affiliation(s)
- Cody A Chastain
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, A-2200 Medical Center North, Nashville, TN 37232-2582, USA
| | - Nathan Klopfenstein
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, A-2200 Medical Center North, Nashville, TN 37232-2582, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1161 21st Avenue South, A-2200 Medical Center North, Nashville, TN 37232-2582, USA
| | - Carlos H Serezani
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, A-2200 Medical Center North, Nashville, TN 37232-2582, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1161 21st Avenue South, A-2200 Medical Center North, Nashville, TN 37232-2582, USA. https://twitter.com/HSerezani
| | - David M Aronoff
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, A-2200 Medical Center North, Nashville, TN 37232-2582, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1161 21st Avenue South, A-2200 Medical Center North, Nashville, TN 37232-2582, USA.
| |
Collapse
|
16
|
Sonnweber T, Pizzini A, Nairz M, Weiss G, Tancevski I. Arachidonic Acid Metabolites in Cardiovascular and Metabolic Diseases. Int J Mol Sci 2018; 19:ijms19113285. [PMID: 30360467 PMCID: PMC6274989 DOI: 10.3390/ijms19113285] [Citation(s) in RCA: 257] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/20/2018] [Accepted: 10/21/2018] [Indexed: 12/20/2022] Open
Abstract
Lipid and immune pathways are crucial in the pathophysiology of metabolic and cardiovascular disease. Arachidonic acid (AA) and its derivatives link nutrient metabolism to immunity and inflammation, thus holding a key role in the emergence and progression of frequent diseases such as obesity, diabetes, non-alcoholic fatty liver disease, and cardiovascular disease. We herein present a synopsis of AA metabolism in human health, tissue homeostasis, and immunity, and explore the role of the AA metabolome in diverse pathophysiological conditions and diseases.
Collapse
Affiliation(s)
- Thomas Sonnweber
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Alex Pizzini
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Manfred Nairz
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Günter Weiss
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
17
|
Brandt SL, Wang S, Dejani NN, Klopfenstein N, Winfree S, Filgueiras L, McCarthy BP, Territo PR, Serezani CH. Excessive localized leukotriene B4 levels dictate poor skin host defense in diabetic mice. JCI Insight 2018; 3:120220. [PMID: 30185672 DOI: 10.1172/jci.insight.120220] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/26/2018] [Indexed: 11/17/2022] Open
Abstract
Poorly controlled diabetes leads to comorbidities and enhanced susceptibility to infections. While the immune components involved in wound healing in diabetes have been studied, the components involved in susceptibility to skin infections remain unclear. Here, we examined the effects of the inflammatory lipid mediator leukotriene B4 (LTB4) signaling through its receptor B leukotriene receptor 1 (BLT1) in the progression of methicillin-resistant Staphylococcus aureus (MRSA) skin infection in 2 models of diabetes. Diabetic mice produced higher levels of LTB4 in the skin, which correlated with larger nonhealing lesion areas and increased bacterial loads compared with nondiabetic mice. High LTB4 levels were also associated with dysregulated cytokine and chemokine production, excessive neutrophil migration but impaired abscess formation, and uncontrolled collagen deposition. Both genetic deletion and topical pharmacological BLT1 antagonism restored inflammatory response and abscess formation, followed by a reduction in the bacterial load and lesion area in the diabetic mice. Macrophage depletion in diabetic mice limited LTB4 production and improved abscess architecture and skin host defense. These data demonstrate that exaggerated LTB4/BLT1 responses mediate a derailed inflammatory milieu that underlies poor host defense in diabetes. Prevention of LTB4 production/actions could provide a new therapeutic strategy to restore host defense in diabetes.
Collapse
Affiliation(s)
- Stephanie L Brandt
- Department of Medicine, Division of Infectious Diseases.,Indiana University School of Medicine, Department of Microbiology & Immunology, Indiana University, Indianapolis, Indiana, USA
| | - Sue Wang
- Indiana University School of Medicine, Department of Microbiology & Immunology, Indiana University, Indianapolis, Indiana, USA
| | - Naiara N Dejani
- Indiana University School of Medicine, Department of Microbiology & Immunology, Indiana University, Indianapolis, Indiana, USA
| | - Nathan Klopfenstein
- Department of Medicine, Division of Infectious Diseases.,Department of Pathology, Microbiology, and Immunology, and
| | - Seth Winfree
- Indiana Center for Biological Microscopy, Indianapolis, Indiana, USA
| | - Luciano Filgueiras
- Indiana University School of Medicine, Department of Microbiology & Immunology, Indiana University, Indianapolis, Indiana, USA
| | - Brian P McCarthy
- Indiana Institute for Biomedical Imaging Sciences, Department of Radiology, Indianapolis, Indiana, USA
| | - Paul R Territo
- Indiana Institute for Biomedical Imaging Sciences, Department of Radiology, Indianapolis, Indiana, USA
| | - C Henrique Serezani
- Department of Medicine, Division of Infectious Diseases.,Department of Pathology, Microbiology, and Immunology, and.,Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Indiana University School of Medicine, Department of Microbiology & Immunology, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
18
|
Intestinal host defense outcome is dictated by PGE 2 production during efferocytosis of infected cells. Proc Natl Acad Sci U S A 2018; 115:E8469-E8478. [PMID: 30127026 DOI: 10.1073/pnas.1722016115] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Inflammatory responses are terminated by the clearance of dead cells, a process termed efferocytosis. A consequence of efferocytosis is the synthesis of the antiinflammatory mediators TGF-β, PGE2, and IL-10; however, the efferocytosis of infected cells favors Th17 responses by eliciting the synthesis of TGF-β, IL-6, and IL-23. Recently, we showed that the efferocytosis of apoptotic Escherichia coli-infected macrophages by dendritic cells triggers PGE2 production in addition to pro-Th17 cytokine expression. We therefore examined the role of PGE2 during Th17 differentiation and intestinal pathology. The efferocytosis of apoptotic E. coli-infected cells by dendritic cells promoted high levels of PGE2, which impaired IL-1R expression via the EP4-PKA pathway in T cells and consequently inhibited Th17 differentiation. The outcome of murine intestinal Citrobacter rodentium infection was dependent on the EP4 receptor. Infected mice treated with EP4 antagonist showed enhanced intestinal defense against C. rodentium compared with infected mice treated with vehicle control. Those results suggest that EP4 signaling during infectious colitis could be targeted as a way to enhance Th17 immunity and host defense.
Collapse
|
19
|
Brandt SL, Klopfenstein N, Wang S, Winfree S, McCarthy BP, Territo PR, Miller L, Serezani CH. Macrophage-derived LTB4 promotes abscess formation and clearance of Staphylococcus aureus skin infection in mice. PLoS Pathog 2018; 14:e1007244. [PMID: 30102746 PMCID: PMC6107286 DOI: 10.1371/journal.ppat.1007244] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 08/23/2018] [Accepted: 07/26/2018] [Indexed: 01/26/2023] Open
Abstract
The early events that shape the innate immune response to restrain pathogens during skin infections remain elusive. Methicillin-resistant Staphylococcus aureus (MRSA) infection engages phagocyte chemotaxis, abscess formation, and microbial clearance. Upon infection, neutrophils and monocytes find a gradient of chemoattractants that influence both phagocyte direction and microbial clearance. The bioactive lipid leukotriene B4 (LTB4) is quickly (seconds to minutes) produced by 5-lipoxygenase (5-LO) and signals through the G protein-coupled receptors LTB4R1 (BLT1) or BLT2 in phagocytes and structural cells. Although it is known that LTB4 enhances antimicrobial effector functions in vitro, whether prompt LTB4 production is required for bacterial clearance and development of an inflammatory milieu necessary for abscess formation to restrain pathogen dissemination is unknown. We found that LTB4 is produced in areas near the abscess and BLT1 deficient mice are unable to form an abscess, elicit neutrophil chemotaxis, generation of neutrophil and monocyte chemokines, as well as reactive oxygen species-dependent bacterial clearance. We also found that an ointment containing LTB4 synergizes with antibiotics to eliminate MRSA potently. Here, we uncovered a heretofore unknown role of macrophage-derived LTB4 in orchestrating the chemoattractant gradient required for abscess formation, while amplifying antimicrobial effector functions.
Collapse
Affiliation(s)
- Stephanie L. Brandt
- Indiana University School of Medicine, Department of Microbiology & Immunology, Indianapolis, Indiana, United States of America
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Disease, Nashville, Tennessee, United States of America
| | - Nathan Klopfenstein
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Disease, Nashville, Tennessee, United States of America
- Vanderbilt University Medical Center, Department of Pathology, Microbiology and Immunology, Nashville, Tennessee, United States of America
| | - Soujuan Wang
- Indiana University School of Medicine, Department of Microbiology & Immunology, Indianapolis, Indiana, United States of America
| | - Seth Winfree
- Indiana Center for Biological Microscopy, Indianapolis, Indiana, United States of America
| | - Brian P. McCarthy
- Indiana Institute for Biomedical Imaging Sciences, Department of Radiology, Indianapolis, Indiana, United States of America
| | - Paul R. Territo
- Indiana Institute for Biomedical Imaging Sciences, Department of Radiology, Indianapolis, Indiana, United States of America
| | - Lloyd Miller
- Johns Hopkins University School of Medicine, Department of Dermatology, Baltimore, Maryland, United States of America
| | - C. Henrique Serezani
- Indiana University School of Medicine, Department of Microbiology & Immunology, Indianapolis, Indiana, United States of America
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Disease, Nashville, Tennessee, United States of America
- Vanderbilt University Medical Center, Department of Pathology, Microbiology and Immunology, Nashville, Tennessee, United States of America
- Vanderbilt Institute of Infection, Immunology and Inflammation, Nashville, Tennessee, United States of America
| |
Collapse
|
20
|
Brandt SL, Putnam NE, Cassat JE, Serezani CH. Innate Immunity to Staphylococcus aureus: Evolving Paradigms in Soft Tissue and Invasive Infections. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:3871-3880. [PMID: 29866769 PMCID: PMC6028009 DOI: 10.4049/jimmunol.1701574] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/12/2018] [Indexed: 01/18/2023]
Abstract
Staphylococcus aureus causes a wide range of diseases that together embody a significant public health burden. Aided by metabolic flexibility and a large virulence repertoire, S. aureus has the remarkable ability to hematogenously disseminate and infect various tissues, including skin, lung, heart, and bone, among others. The hallmark lesions of invasive staphylococcal infections, abscesses, simultaneously denote the powerful innate immune responses to tissue invasion as well as the ability of staphylococci to persist within these lesions. In this article, we review the innate immune responses to S. aureus during infection of skin and bone, which serve as paradigms for soft tissue and bone disease, respectively.
Collapse
Affiliation(s)
- Stephanie L Brandt
- Division of Infectious Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Nicole E Putnam
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - James E Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232;
- Division of Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232; and
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232
| | - C Henrique Serezani
- Division of Infectious Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232;
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
21
|
Abstract
The body is exposed to foreign pathogens every day, but remarkably, most pathogens are effectively cleared by the innate immune system without the need to invoke the adaptive immune response. Key cellular components of the innate immune system include macrophages and neutrophils and the recruitment and function of these cells are tightly regulated by chemokines and cytokines in the tissue space. Innate immune responses are also known to regulate development of adaptive immune responses often via the secretion of various cytokines. In addition to these protein regulators, numerous lipid mediators can also influence innate and adaptive immune functions. In this review, we cover one particular lipid regulator, prostaglandin E2 (PGE2) and describe its synthesis and signaling and what is known about the ability of this lipid to regulate immunity and host defense against viral, fungal and bacterial pathogens.
Collapse
Affiliation(s)
| | - Bethany B Moore
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
22
|
Diabetes mellitus und Immunantwort bei pyogenen Infektionen. DIABETOLOGE 2018. [DOI: 10.1007/s11428-018-0320-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
23
|
Penteado LDA, Dejani NN, Verdan FF, Orlando AB, Niño VE, Dias FDN, Salina ACG, Medeiros AI. Distinctive role of efferocytosis in dendritic cell maturation and migration in sterile or infectious conditions. Immunology 2017; 151:304-313. [PMID: 28267881 DOI: 10.1111/imm.12731] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 02/07/2017] [Accepted: 02/23/2017] [Indexed: 12/12/2022] Open
Abstract
Efferocytosis, or clearance of apoptotic cells (ACs), by dendritic cells (DCs) leads to immune response suppression and tolerance to self-antigens. However, efferocytosis of infected apoptotic cells (IACs) leads to the production of a mixed pro- and anti-inflammatory cytokine milieu. We examined the DC phenotype and ability to migrate after phagocytosis of ACs or IACs and observed higher levels of CD86 and CCR7 expression in DCs, as well as enhanced migration capacity following efferocytosis of IACs. Interestingly, higher levels of interleukin-1β, interleukin-10 and prostaglandin E2 (PGE2 ) were also produced in this context. Blockage of IAC recognition led to an impaired maturation profile and PGE2 production, which may have contributed to reduced CD86 and CCR7 expression and migration capacity. These data contribute to the understanding of how efferocytosis of sterile or infected cells may regulate the adaptive immune response, although the precise role of PGE2 in this process requires further investigation.
Collapse
Affiliation(s)
- Letícia de Aquino Penteado
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Naiara Naiana Dejani
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil.,Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Felipe Fortino Verdan
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil.,Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Allan Botinhon Orlando
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Victoria Eugenia Niño
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Fernanda De Nuzzi Dias
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Ana Carolina Guerta Salina
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil.,Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Alexandra Ivo Medeiros
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| |
Collapse
|