1
|
Akbari MS, Joyce LR, Spencer BL, Brady A, McIver KS, Doran KS. Identification of glyoxalase A in group B Streptococcus and its contribution to methylglyoxal tolerance and virulence. Infect Immun 2025; 93:e0054024. [PMID: 40008888 PMCID: PMC11977320 DOI: 10.1128/iai.00540-24] [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: 12/18/2024] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Group B Streptococcus (GBS) is a Gram-positive pathobiont that commonly colonizes the gastrointestinal and lower female genital tracts but can cause sepsis and pneumonia in newborns and is a leading cause of neonatal meningitis. Despite the resulting disease severity, the pathogenesis of GBS is not completely understood, especially during the early phases of infection. To investigate GBS factors necessary for bloodstream survival, we performed a transposon (Tn) mutant screen in our bacteremia infection model using a GBS mariner transposon mutant library previously developed by our group. We identified significantly underrepresented mutations in 623 genes that contribute to survival in the blood, including those encoding known virulence factors such as capsule, the β-hemolysin, and inorganic metal ion transport systems. Most of the underrepresented genes have not been previously characterized or studied in GBS, including gloA and gloB, which are homologs for genes involved in methylglyoxal (MG) detoxification. MG is a byproduct of glycolysis and a highly reactive toxic aldehyde that is elevated in immune cells during infection. Here, we observed MG sensitivity across multiple GBS isolates and confirmed that gloA contributes to MG tolerance and invasive GBS infection. We show specifically that gloA contributes to GBS survival in the presence of neutrophils and depleting neutrophils in mice abrogates the decreased survival and infection of the gloA mutant. The requirement of the glyoxalase pathway during GBS infection suggests that MG detoxification is important for bacterial survival during host-pathogen interactions.IMPORTANCEA transposon-mutant screen of group B Streptococcus (GBS) in a bacteremia mouse model of infection revealed virulence factors known to be important for GBS survival such as the capsule, β-hemolysin/cytolysin, and genes involved in metal homeostasis. Many uncharacterized factors were also identified including genes that are part of the metabolic pathway that breaks down methylglyoxal (MG). The glyoxalase pathway is the most ubiquitous metabolic pathway for MG breakdown and is only a two-step process using glyoxalase A (gloA) and B (gloB) enzymes. MG is a highly reactive byproduct of glycolysis and is made by most cells. Here, we show that in GBS, the first enzyme in the glyoxalase pathway, encoded by gloA, contributes to MG resistance and blood survival. We further demonstrate that GloA contributes to GBS survival against neutrophils in vitro and in vivo and, therefore, is an important virulence factor required for invasive infection.
Collapse
Affiliation(s)
- Madeline S. Akbari
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Luke R. Joyce
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Brady L. Spencer
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Amanda Brady
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kevin S. McIver
- Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Kelly S. Doran
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
2
|
Manuel G, Twentyman J, Noble K, Eastman AJ, Aronoff DM, Seepersaud R, Rajagopal L, Adams Waldorf KM. Group B streptococcal infections in pregnancy and early life. Clin Microbiol Rev 2025; 38:e0015422. [PMID: 39584819 PMCID: PMC11905376 DOI: 10.1128/cmr.00154-22] [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: 11/26/2024] Open
Abstract
SUMMARYBacterial infections with Group B Streptococcus (GBS) are an important cause of adverse outcomes in pregnant individuals, neonates, and infants. GBS is a common commensal in the genitourinary and gastrointestinal tracts and can be detected in the vagina of approximately 20% of women globally. GBS can infect the fetus either during pregnancy or vaginal delivery resulting in preterm birth, stillbirth, or early-onset neonatal disease (EOD) in the first week of life. The mother can also become infected with GBS leading to postpartum endometritis, and rarely, maternal sepsis. An invasive GBS infection of the neonate may present after the first week of life (late-onset disease, LOD) through transmission from caregivers, breast milk, and other sources. Invasive GBS infections in neonates can result in sepsis, pneumonia, meningitis, neurodevelopmental impairment, death, and lifelong disability. A policy of routine screening for GBS rectovaginal colonization in well-resourced countries can trigger the administration of intrapartum antibiotic prophylaxis (IAP) when prenatal testing is positive, which drastically reduces rates of EOD. However, many countries do not routinely screen pregnant women for GBS colonization but may administer IAP in cases with a high risk of EOD. IAP does not reduce rates of LOD. A global vaccination campaign is needed to reduce the significant burden of invasive GBS disease that remains among infants and pregnant individuals. In this narrative review, we provide a comprehensive overview of the global impact of GBS colonization and infection, virulence factors and pathogenesis, and current and future prophylactics and therapeutics.
Collapse
Affiliation(s)
- Gygeria Manuel
- Department of Obstetrics & Gynecology, University of Washington, Seattle, Washington, USA
| | - Joy Twentyman
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Kristen Noble
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alison J. Eastman
- Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - David M. Aronoff
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ravin Seepersaud
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Lakshmi Rajagopal
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
- Global Health, University of Washington, Seattle, Washington, USA
| | - Kristina M. Adams Waldorf
- Department of Obstetrics & Gynecology, University of Washington, Seattle, Washington, USA
- Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
3
|
Spencer BL, Nguyen DT, Marroquin SM, Gapin L, O’Brien RL, Doran KS. Characterization of the Cellular Immune Response to Group B Streptococcal Vaginal Colonization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635275. [PMID: 39975125 PMCID: PMC11838357 DOI: 10.1101/2025.01.29.635275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Introduction Group B Streptococcus (GBS) asymptomatic colonizes the female genital tract (FGT) but can contribute to adverse pregnancy outcomes including pre-term birth, chorioamnionitis, and neonatal infection. We previously observed that GBS elicits FGT cytokine responses, including IL-17, during murine vaginal colonization; yet the anti-GBS cellular immune response during colonization remained unknown. We hypothesized that GBS may induce cellular immunity, resulting in FGT clearance. Methods Herein, we utilize depleting antibodies and knockout mice and performed flow cytometry to investigate cellular immunes responses during GBS colonization. Results We found that neutrophils (effectors of the IL-17 response) are important for GBS mucosal control as neutrophil depletion promoted increased GBS burdens in FGT tissues. Flow cytometric analysis of immune populations in the vagina, cervix, and uterus revealed, however, that GBS colonization did not induce a marked increase in FGT CD45+ immune cells. We also found that that Vγ6+ γδ T cells comprise a primary source of FGT IL-17. Finally, using knockout mice, we observed that IL-17-producing γδ T cells are important for the control of GBS in the FGT during murine colonization. Conclusions Taken together, this work characterizes FGT cellular immunity and suggests that GBS colonization does not elicit a significant immune response, which may be a bacterial directed adaptive outcome. However, certain FGT immune cells, such as neutrophils and ɣδ T cells, contribute to host defense and control of GBS colonization.
Collapse
Affiliation(s)
- Brady L. Spencer
- University of Colorado-Anschutz, Department of Immunology & Microbiology, Aurora, CO, USA
| | - Dustin T. Nguyen
- University of Colorado-Anschutz, Department of Immunology & Microbiology, Aurora, CO, USA
| | - Stephanie M. Marroquin
- University of Colorado-Anschutz, Department of Immunology & Microbiology, Aurora, CO, USA
| | - Laurent Gapin
- University of Colorado-Anschutz, Department of Immunology & Microbiology, Aurora, CO, USA
| | - Rebecca L. O’Brien
- National Jewish Health, Department of Biomedical Research, Denver, CO, USA
| | - Kelly S. Doran
- University of Colorado-Anschutz, Department of Immunology & Microbiology, Aurora, CO, USA
| |
Collapse
|
4
|
Wang Y, Yang Q, Dong Y, Wang L, Zhang Z, Niu R, Wang Y, Bi Y, Liu G. Piezo1-directed neutrophil extracellular traps regulate macrophage differentiation during influenza virus infection. Cell Death Dis 2025; 16:60. [PMID: 39890818 PMCID: PMC11785962 DOI: 10.1038/s41419-025-07395-5] [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: 08/16/2024] [Revised: 12/20/2024] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Neutrophils and macrophages are critical for antiviral immunity, but their reciprocal regulatory roles and mechanisms in the response to viral infection remain unclear. Herein, we found that the ion channel Piezo1 directs neutrophil extracellular trap (NET) formation and regulates macrophage functional differentiation in anti-influenza virus immunity. Genetic deletion of Piezo1 in neutrophils inhibited the generation of NETs and M1 macrophage differentiation while driving the development of M2 macrophages during viral infection. Piezo1-directed neutrophil NET DNA directly regulates macrophage differentiation in vitro and in vivo. Mechanistically, neutrophil Piezo1 deficiency inhibited NET DNA production, leading to decreased TLR9 and cGAS-STING signalling activity while inducing reciprocal differentiation from M1 to M2 macrophages. In addition, Piezo1 integrates magnesium signalling and the SIRT2-hypoxia-inducible factor-1 alpha (HIF1α)-dependent pathway to orchestrate reciprocal M1 and M2 macrophage lineage commitment through neutrophil-derived NET DNA. Our studies provide critical insight into the role of neutrophil-based mechanical regulation of immunopathology in directing macrophage lineage commitment during the response to influenza virus infection.
Collapse
Affiliation(s)
- Yuexin Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Qiuli Yang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Yingjie Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Likun Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, 100080, Beijing, China
| | - Zhiyuan Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Ruiying Niu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Yufei Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, 100080, Beijing, China.
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China.
| |
Collapse
|
5
|
Dong Y, Yang Q, Wang L, Niu R, Xia J, Bi Y, Liu G. Protocol for building an in vivo model for observation and evaluation of neutrophil extracellular trapping nets in influenza virus-infected mice. STAR Protoc 2024; 5:103436. [PMID: 39499615 PMCID: PMC11568775 DOI: 10.1016/j.xpro.2024.103436] [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: 08/21/2024] [Revised: 09/20/2024] [Accepted: 10/14/2024] [Indexed: 11/07/2024] Open
Abstract
Neutrophils sense microbes and selectively release neutrophil extracellular traps (NETs) in response to pathogens. Here, we present a protocol for qualitative and quantitative analysis of NETs in PR8-infected mouse models. We describe steps for obtaining single-cell suspensions of infiltrated neutrophils from mouse bronchoalveolar lavage fluid (BALF). We then detail procedures to determine NET marker proteins using flow cytometry, confocal microscopes, and western blotting. This protocol may be applied to qualitative and quantitative analysis of NETs in influenza virus-infected mice. For complete details on the use and execution of this protocol, please refer to Zhang et al.1.
Collapse
Affiliation(s)
- Yingjie Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| | - Qiuli Yang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Likun Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing 100071, China
| | - Ruiying Niu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Jingxuan Xia
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing 100071, China.
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
6
|
Akbari MS, Joyce LR, Spencer BL, Brady A, McIver KS, Doran KS. Identification of Glyoxalase A in Group B Streptococcus and its contribution to methylglyoxal tolerance and virulence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605887. [PMID: 39131367 PMCID: PMC11312555 DOI: 10.1101/2024.07.30.605887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Group B Streptococcus (GBS) is a Gram-positive pathobiont that commonly colonizes the gastrointestinal and lower female genital tracts but can cause sepsis and pneumonia in newborns and is a leading cause of neonatal meningitis. Despite the resulting disease severity, the pathogenesis of GBS is not completely understood, especially during the early phases of infection. To investigate GBS factors necessary for blood stream survival, we performed a transposon (Tn) mutant screen in our bacteremia infection model using a GBS mariner transposon mutant library previously developed by our group. We identified significantly underrepresented mutations in 623 genes that contribute to survival in the blood, including those encoding known virulence factors such as capsule, the β-hemolysin, and inorganic metal ion transport systems. Most of the underrepresented genes have not been previously characterized or studied in GBS, including gloA and gloB, which are homologs for genes involved in methylglyoxal (MG) detoxification. MG is a byproduct of glycolysis and a highly reactive toxic aldehyde that is elevated in immune cells during infection. Here, we observed MG sensitivity across multiple GBS isolates and confirm that gloA contributes to MG tolerance and invasive GBS infection. We show specifically that gloA contributes to GBS survival in the presence of neutrophils and depleting neutrophils in mice abrogates the decreased survival and infection of the gloA mutant. The requirement of the glyoxalase pathway during GBS infection suggests that MG detoxification is important for bacterial survival during host-pathogen interactions.
Collapse
Affiliation(s)
- Madeline S. Akbari
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Luke R. Joyce
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Brady L. Spencer
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Amanda Brady
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Kevin S. McIver
- Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Kelly S. Doran
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| |
Collapse
|
7
|
Pongchaikul P, Romero R, Wongsurawat T, Jenjaroenpun P, Kruasuwan W, Mongkolsuk P, Vivithanaporn P, Thaipisuttikul I, Singsaneh A, Khamphakul J, Santanirand P, Kotchompoo K, Bhuwapathanapun M, Warintaksa P, Chaemsaithong P. Molecular evidence that GBS early neonatal sepsis results from ascending infection: comparative hybrid genomics analyses show that microorganisms in the vaginal ecosystem, amniotic fluid, chorioamniotic membranes, and neonatal blood are the same. J Perinat Med 2024; 52:977-990. [PMID: 39405032 PMCID: PMC11560570 DOI: 10.1515/jpm-2024-0310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/20/2024] [Indexed: 11/12/2024]
Abstract
OBJECTIVES Streptococcus agalactiae, or Group B Streptococcus (GBS), is a leading cause of neonatal sepsis. Materno-fetal transmission of the microorganisms present in the lower genital tract/perineum is considered to be the most frequent mode for acquisition of infection. It has also been proposed that, in a subset of cases, GBS causes acute chorioamnionitis, intraamniotic infection, and fetal/neonatal sepsis. However, the evidence to support this ascending pathway is derived from microbiologic studies that rely on cultivation methods, which do not have the resolution to determine if the microorganisms causing neonatal sepsis are the same as those found in the amniotic fluid and the vaginal ecosystem. METHODS We used whole genome sequencing of the microorganisms isolated from the vagina, amniotic fluid, chorioamniotic membranes, and neonatal blood (four isolates) in a case of early neonatal sepsis. Using hybrid genome assembly, we characterized the genomic features including virulence factors and antimicrobial resistance in four isolates from the same mother, placenta, and newborn. RESULTS Whole genome sequencing revealed that the microorganisms in the four clinical isolates corresponded to S. agalactiae sequence type 1, clonal complexes 1, and serotype Ib. Comparative genomic analysis illustrated similar DNA sequences of the four genomes. CONCLUSIONS This study presents the first evidence of the genomic similarity of microorganisms in the vaginal ecosystem, the space between the chorioamniotic membranes of the placenta, amniotic fluid, and neonatal blood.
Collapse
Affiliation(s)
- Pisut Pongchaikul
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakan 10540, Thailand
- Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom 73170, Thailand
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L7 3EA, United Kingdom
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland 20892, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan 48824, USA
| | - Thidathip Wongsurawat
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Piroon Jenjaroenpun
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Worarat Kruasuwan
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Paninee Mongkolsuk
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakan 10540, Thailand
| | - Pornpun Vivithanaporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakan 10540, Thailand
| | - Iyarit Thaipisuttikul
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Arunee Singsaneh
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Jakkrit Khamphakul
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Pitak Santanirand
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Kanyaphat Kotchompoo
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Maolee Bhuwapathanapun
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Puntabut Warintaksa
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Piya Chaemsaithong
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
8
|
Brummerhop AS, Lee CT, Weltman R, Tribble GD, van der Hoeven R, Chiu Y, Hong J, Wang BY. Synergistic effects of antimicrobial components of the human-derived composite amnion-chorion membrane on bacterial growth. Front Cell Infect Microbiol 2024; 14:1472737. [PMID: 39435187 PMCID: PMC11491435 DOI: 10.3389/fcimb.2024.1472737] [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: 07/29/2024] [Accepted: 09/17/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction The human-derived amnion-chorion membrane (ACM) has endogenous antimicrobial properties, which are important for preventing the colonization and survival of oral bacteria on exposed membranes. This project aimed to decipher the underlying mechanism by identifying the components of ACM that confer antibacterial properties. In addition, the antimicrobial efficacy of these identified components on oral bacteria was assessed. Methods Four antimicrobial proteins, histone H2A/H2B, cathelicidin LL-37, lactoferrin, and lysozyme, were identified via mass spectrometry in ACM. These proteins were then assessed for their efficacy in killing Streptococcus gordonii Challis. Log-phased bacterial cells were cultured with the commercially available proteins that were identified in ACM, either individually or in combination, at different concentrations. After incubation for 8 or 24 hours, the bacteria were stained with a live/dead viability kit and analyzed via confocal microscopy. Results The combination of these proteins effectively killed S. gordonii in a dose-dependent fashion after 8 or 24 hours of incubation. When each protein was tested individually, it killed S. gordonii at a much lower efficacy relative to the combinations. The synergistic effects of the antimicrobial protein combinations were also observed in both the viable cell count recovery and minimum inhibitory concentration assays. Discussion By shedding light on the mechanisms in the ACM's antimicrobial property, this study may raise more awareness of the potential benefit of utilization of a membrane with endogenous antimicrobial properties in regeneration surgeries.
Collapse
Affiliation(s)
- Alexandra Su Brummerhop
- Department of Periodontics and Dental Hygiene, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
| | - Chun-Teh Lee
- Department of Periodontics and Dental Hygiene, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
| | - Robin Weltman
- Department of Periodontics and Dental Hygiene, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
- Department of Clinical Sciences, University of Nevada School of Dental Medicine, Las Vegas, NV, United States
| | - Gena D. Tribble
- Department of Periodontics and Dental Hygiene, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
| | - Ransome van der Hoeven
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry, Iowa City, IA, United States
- Department of Periodontics, University of Iowa College of Dentistry, Iowa City, IA, United States
| | - Yulun Chiu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jianming Hong
- Department of Periodontics and Dental Hygiene, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
| | - Bing-Yan Wang
- Department of Periodontics and Dental Hygiene, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
| |
Collapse
|
9
|
Ling J, Hryckowian AJ. Re-framing the importance of Group B Streptococcus as a gut-resident pathobiont. Infect Immun 2024; 92:e0047823. [PMID: 38436256 PMCID: PMC11392526 DOI: 10.1128/iai.00478-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
Streptococcus agalactiae (Group B Streptococcus, GBS) is a Gram-positive bacterial species that causes disease in humans across the lifespan. While antibiotics are used to mitigate GBS infections, it is evident that antibiotics disrupt human microbiomes (which can predispose people to other diseases later in life), and antibiotic resistance in GBS is on the rise. Taken together, these unintended negative impacts of antibiotics highlight the need for precision approaches for minimizing GBS disease. One possible approach involves selectively depleting GBS in its commensal niches before it can cause disease at other body sites or be transmitted to at-risk individuals. One understudied commensal niche of GBS is the adult gastrointestinal (GI) tract, which may predispose colonization at other body sites in individuals at risk for GBS disease. However, a better understanding of the host-, microbiome-, and GBS-determined variables that dictate GBS GI carriage is needed before precise GI decolonization approaches can be developed. In this review, we synthesize current knowledge of the diverse body sites occupied by GBS as a pathogen and as a commensal. We summarize key molecular factors GBS utilizes to colonize different host-associated niches to inform future efforts to study GBS in the GI tract. We also discuss other GI commensals that are pathogenic in other body sites to emphasize the broader utility of precise de-colonization approaches for mitigating infections by GBS and other bacterial pathogens. Finally, we highlight how GBS treatments could be improved with a more holistic understanding of GBS enabled by continued GI-focused study.
Collapse
Affiliation(s)
- Joie Ling
- Department of
Medicine, Division of Gastroenterology and Hepatology, University of
Wisconsin School of Medicine and Public
Health, Madison,
Wisconsin, USA
- Department of Medical
Microbiology and Immunology, University of Wisconsin School of Medicine
and Public Healthon,
Madison, Wisconsin, USA
- Microbiology Doctoral
Training Program, University of
Wisconsin-Madison, Madison,
Wisconsin, USA
| | - Andrew J. Hryckowian
- Department of
Medicine, Division of Gastroenterology and Hepatology, University of
Wisconsin School of Medicine and Public
Health, Madison,
Wisconsin, USA
- Department of Medical
Microbiology and Immunology, University of Wisconsin School of Medicine
and Public Healthon,
Madison, Wisconsin, USA
| |
Collapse
|
10
|
Georges HM, Cassin C, Tong M, Abrahams VM. TLR8-activating miR-146a-3p is an intermediate signal contributing to fetal membrane inflammation in response to bacterial LPS. Immunology 2024; 172:577-587. [PMID: 38631842 PMCID: PMC11223956 DOI: 10.1111/imm.13794] [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: 12/10/2023] [Accepted: 04/05/2024] [Indexed: 04/19/2024] Open
Abstract
Preterm birth is the largest contributor to neonatal morbidity and is often associated with chorioamnionitis, defined as inflammation/infection of the fetal membranes (FMs). Chorioamnionitis is characterised by neutrophil infiltration of the FMs and is associated with elevated levels of the neutrophil chemoattractant, interleukin (IL)-8 and the proinflammatory cytokine, IL-1β. While FMs can respond to infections through innate immune sensors, such as toll-like receptors (TLRs), the downstream mechanisms by which chorioamnionitis arises are not fully understood. A novel group of non-classical microRNAs (miR-21a, miR-29a, miR-146a-3p, Let-7b) function as endogenous danger signals by activating the ssRNA viral sensors TLR7 and TLR8. In this study, the pro-inflammatory roles of TLR7/TLR8-activating miRs were examined as mediators of FM inflammation in response to bacterial lipopolysaccharide (LPS) using an in vitro human FM explant system, an in vivo mouse model of pregnancy, and human clinical samples. Following LPS exposure, miR-146a-3p was significantly increased in both human FM explants and wild-type mouse FMs. Expression of miR-146a-3p was also significantly elevated in FMs from women with preterm birth and chorioamnionitis. FM IL-8 and inflammasome-mediated IL-1β production in response to LPS was dependent on miR-146a-3p and TLR8 downstream of TLR4 activation. In wild-type mice, LPS exposure increased FM IL-8 and IL-1β production and induced preterm birth. In TLR7-/-/TLR8-/- mice, LPS exposure was able to initiate but not sustain preterm birth, and FM inflammation was reduced. Together, we demonstrate a novel signalling mechanism at the maternal-fetal interface in which TLR8-activating miR-146a-3p acts as an intermediate danger signal to drive FM inflammasome-dependent and -independent mechanisms of inflammation and, thus, may play a role in chorioamnionitis and subsequent preterm birth.
Collapse
Affiliation(s)
- Hanah M. Georges
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Caterina Cassin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Mancy Tong
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Vikki M. Abrahams
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
11
|
Zhang Z, Yang Q, Dong Y, Wang L, Niu R, Xia J, Bi Y, Liu G. Sirtuin 2 regulates neutrophil functions through NAD + synthesis pathway in virus infection. iScience 2024; 27:110184. [PMID: 38974970 PMCID: PMC11226967 DOI: 10.1016/j.isci.2024.110184] [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: 01/11/2024] [Revised: 04/12/2024] [Accepted: 06/01/2024] [Indexed: 07/09/2024] Open
Abstract
Neutrophils play an important role in antiviral immunity, but the underlying mechanisms remain unclear. Here, we found that SIRT2 deficiency inhibited the infiltration of neutrophils, as well as the secretion of inflammatory cytokines and the formation of neutrophil extracellular traps (NETs), ameliorating disease symptoms during acute respiratory virus infection. Mechanistically, SIRT2 deficiency upregulates quinolinic acid (QA)-producing enzyme 3-hydroxyanthranilate oxygenase (3-HAO) and leads to expression of quinolinate phosphoribosyltransferase (QPRT), which promotes the synthesis of QA for NAD+ and limits viral infection when de novo NAD+ synthesis is blocked. Tryptophan-2,3-oxygenase expressed in epithelial cells metabolizes tryptophan to produce kynurenine and 3-hydroxyaminobenzoic acid, which is a source of intracellular QA in neutrophils. Thus, our findings reveal a previously unrecognized QPRT-mediated switch in NAD+ metabolism by exploiting neutrophil-derived QA as an alternative source of replenishing intracellular NAD+ pools induced by SIRT2 to regulate neutrophil functions during virus infection, with implications for future immunotherapy approaches.
Collapse
Affiliation(s)
- Zhiyuan Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Qiuli Yang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Yingjie Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Likun Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing 100071, China
| | - Ruiying Niu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Jingxuan Xia
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing 100071, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
12
|
Mallada B, Villalobos F, Donoso B, Casares R, Longhi G, Mendieta-Moreno JI, Jiménez-Martín A, Haïdour A, Seepersaud R, Rajagopal L, de la Torre B, Millán A, Cuerva JM. Single-Molecule Identification of the Isomers of a Lipidic Antibody Activator. J Phys Chem Lett 2024; 15:6935-6942. [PMID: 38935930 PMCID: PMC11247479 DOI: 10.1021/acs.jpclett.4c00164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/29/2024] [Accepted: 05/03/2024] [Indexed: 06/29/2024]
Abstract
Molecular structural elucidation can be accomplished by different techniques, such as nuclear magnetic resonance or X-ray diffraction. However, the former does not give information about the three-dimensional atomic arrangement, and the latter needs crystallizable solid samples. An alternative is direct, real-space visualization of the molecules by cryogenic scanning tunneling microscopy (STM). This technique is usually limited to thermally robust molecules because an annealing step is required for sample deposition. A landmark development has been the coupling of STM with electrospray deposition (ESD), which smooths the process and widens the scope of the visualization technique. In this work, we present the on-surface characterization of air-, light-, and temperature-sensitive rhamnopolyene with relevance in molecular biology. Supported by theoretical calculations, we characterize two isomers of this flexible molecule, confirming the potential of the technique to inspect labile, non-crystallizable compounds.
Collapse
Affiliation(s)
- Benjamin Mallada
- Institute
of Physics, Czech Academy of Sciences, 16200 Prague, Czech Republic
- Regional
Centre of Advanced Technologies and Materials, Czech Advanced Technology
and Research Institute (CATRIN), Palacký
University Olomouc, 78371 Olomouc, Czech Republic
| | - Federico Villalobos
- Departamento
de Química Orgánica, Unidad de Excelencia de Química
Aplicada a la Biomedicina y Medioambiente, C. U. Fuentenueva, Universidad de Granada, 18071 Granada, Spain
| | - Beatriz Donoso
- Departamento
de Química Orgánica, Unidad de Excelencia de Química
Aplicada a la Biomedicina y Medioambiente, C. U. Fuentenueva, Universidad de Granada, 18071 Granada, Spain
| | - Raquel Casares
- Departamento
de Química Orgánica, Unidad de Excelencia de Química
Aplicada a la Biomedicina y Medioambiente, C. U. Fuentenueva, Universidad de Granada, 18071 Granada, Spain
| | - Giovanna Longhi
- Dipartimento
di Medicina Molecolare e Traslazionale, Universitá di Brescia, Viale Europa 11, 25121 Brescia, Italy
| | - Jesús I. Mendieta-Moreno
- Instituto
de Ciencia de Materiales de Madrid (ICMM), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain
| | - Alejandro Jiménez-Martín
- Institute
of Physics, Czech Academy of Sciences, 16200 Prague, Czech Republic
- Regional
Centre of Advanced Technologies and Materials, Czech Advanced Technology
and Research Institute (CATRIN), Palacký
University Olomouc, 78371 Olomouc, Czech Republic
- Faculty
of Nuclear Sciences and Physical Engineering, Czech Technical University, 11519 Prague, Czech
Republic
| | - Ali Haïdour
- Unidad
de Resonancia Magnética Nuclear, Centro de Instrumentación
Científica, Universidad de Granada, Paseo Juan Osorio s/n, 18071 Granada, Spain
| | - Ravin Seepersaud
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| | - Lakshmi Rajagopal
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
- Department
of Global Health, University of Washington, Seattle, Washington 98105, United States
- Department
of Pediatrics, University of Washington, Seattle, Washington 98105, United States
| | - Bruno de la Torre
- Regional
Centre of Advanced Technologies and Materials, Czech Advanced Technology
and Research Institute (CATRIN), Palacký
University Olomouc, 78371 Olomouc, Czech Republic
| | - Alba Millán
- Departamento
de Química Orgánica, Unidad de Excelencia de Química
Aplicada a la Biomedicina y Medioambiente, C. U. Fuentenueva, Universidad de Granada, 18071 Granada, Spain
| | - Juan M. Cuerva
- Departamento
de Química Orgánica, Unidad de Excelencia de Química
Aplicada a la Biomedicina y Medioambiente, C. U. Fuentenueva, Universidad de Granada, 18071 Granada, Spain
| |
Collapse
|
13
|
Aslanian-Kalkhoran L, Mehdizadeh A, Aghebati-Maleki L, Danaii S, Shahmohammadi-Farid S, Yousefi M. The role of neutrophils and neutrophil extracellular traps (NETs) in stages, outcomes and pregnancy complications. J Reprod Immunol 2024; 163:104237. [PMID: 38503075 DOI: 10.1016/j.jri.2024.104237] [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: 08/28/2023] [Revised: 01/23/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024]
Abstract
Neutrophils are the main components of innate immunity to eliminate infectious pathogens. Neutrophils play a role in several stages of the reproductive cycle, and their presence in the female reproductive system is highly regulated, so their function may change during pregnancy. Emerging evidence suggests that neutrophils are important at all stages of pregnancy, from implantation, placentation, and connective tissue regeneration to birth, as well as birth itself. Neutrophil extracellular traps (NETs) are defined as extracellular strands of unfolded DNA together with histone complexes and neutrophil granule proteins. NET formation is a new mechanism of these cells for their defense function. These strands containing DNA and antimicrobial peptides were initially recognized as one of the defense mechanisms of neutrophils, but later it was explained that they are involved in a variety of non-infectious diseases. Since the source of inflammation and tissue damage is the irregular activity of neutrophils, it is not surprising that NETosis are associated with a number of inflammatory conditions and diseases. The overexpression of NET components or non-principled NET clearance is associated with the risk of production and activation of autoantibodies, which results in participation in autoinflammatory and autoimmune disorders (SLE, RA), fibrosis, sepsis and other disorders such as vascular diseases, for example, thrombosis and atherosclerosis. Recent published articles have shown the role of neutrophils and extracellular traps (NETs) in pregnancy, childbirth and pregnancy-related diseases. The aim of this study was to identify and investigate the role of neutrophils and neutrophil extracellular traps (NETs) in the stages of pregnancy, as well as the complications caused by these cells.
Collapse
Affiliation(s)
- Lida Aslanian-Kalkhoran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Shahla Danaii
- Gynecology Department, Eastern Azerbaijan ACECR ART Centre, Eastern Azerbaijan Branch of ACECR, Tabriz, Iran
| | | | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
14
|
Goh KGK, Desai D, Thapa R, Prince D, Acharya D, Sullivan MJ, Ulett GC. An opportunistic pathogen under stress: how Group B Streptococcus responds to cytotoxic reactive species and conditions of metal ion imbalance to survive. FEMS Microbiol Rev 2024; 48:fuae009. [PMID: 38678005 PMCID: PMC11098048 DOI: 10.1093/femsre/fuae009] [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: 08/31/2023] [Revised: 03/26/2024] [Accepted: 04/16/2024] [Indexed: 04/29/2024] Open
Abstract
Group B Streptococcus (GBS; also known as Streptococcus agalactiae) is an opportunistic bacterial pathogen that causes sepsis, meningitis, pneumonia, and skin and soft tissue infections in neonates and healthy or immunocompromised adults. GBS is well-adapted to survive in humans due to a plethora of virulence mechanisms that afford responses to support bacterial survival in dynamic host environments. These mechanisms and responses include counteraction of cell death from exposure to excess metal ions that can cause mismetallation and cytotoxicity, and strategies to combat molecules such as reactive oxygen and nitrogen species that are generated as part of innate host defence. Cytotoxicity from reactive molecules can stem from damage to proteins, DNA, and membrane lipids, potentially leading to bacterial cell death inside phagocytic cells or within extracellular spaces within the host. Deciphering the ways in which GBS responds to the stress of cytotoxic reactive molecules within the host will benefit the development of novel therapeutic and preventative strategies to manage the burden of GBS disease. This review summarizes knowledge of GBS carriage in humans and the mechanisms used by the bacteria to circumvent killing by these important elements of host immune defence: oxidative stress, nitrosative stress, and stress from metal ion intoxication/mismetallation.
Collapse
Affiliation(s)
- Kelvin G K Goh
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Devika Desai
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Ruby Thapa
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Darren Prince
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Dhruba Acharya
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Matthew J Sullivan
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Glen C Ulett
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| |
Collapse
|
15
|
Jung E, Romero R, Suksai M, Gotsch F, Chaemsaithong P, Erez O, Conde-Agudelo A, Gomez-Lopez N, Berry SM, Meyyazhagan A, Yoon BH. Clinical chorioamnionitis at term: definition, pathogenesis, microbiology, diagnosis, and treatment. Am J Obstet Gynecol 2024; 230:S807-S840. [PMID: 38233317 PMCID: PMC11288098 DOI: 10.1016/j.ajog.2023.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 04/05/2023]
Abstract
Clinical chorioamnionitis, the most common infection-related diagnosis in labor and delivery units, is an antecedent of puerperal infection and neonatal sepsis. The condition is suspected when intrapartum fever is associated with two other maternal and fetal signs of local or systemic inflammation (eg, maternal tachycardia, uterine tenderness, maternal leukocytosis, malodorous vaginal discharge or amniotic fluid, and fetal tachycardia). Clinical chorioamnionitis is a syndrome caused by intraamniotic infection, sterile intraamniotic inflammation (inflammation without bacteria), or systemic maternal inflammation induced by epidural analgesia. In cases of uncertainty, a definitive diagnosis can be made by analyzing amniotic fluid with methods to detect bacteria (Gram stain, culture, or microbial nucleic acid) and inflammation (white blood cell count, glucose concentration, interleukin-6, interleukin-8, matrix metalloproteinase-8). The most common microorganisms are Ureaplasma species, and polymicrobial infections occur in 70% of cases. The fetal attack rate is low, and the rate of positive neonatal blood cultures ranges between 0.2% and 4%. Intrapartum antibiotic administration is the standard treatment to reduce neonatal sepsis. Treatment with ampicillin and gentamicin have been recommended by professional societies, although other antibiotic regimens, eg, cephalosporins, have been used. Given the importance of Ureaplasma species as a cause of intraamniotic infection, consideration needs to be given to the administration of antimicrobial agents effective against these microorganisms such as azithromycin or clarithromycin. We have used the combination of ceftriaxone, clarithromycin, and metronidazole, which has been shown to eradicate intraamniotic infection with microbiologic studies. Routine testing of neonates born to affected mothers for genital mycoplasmas could improve the detection of neonatal sepsis. Clinical chorioamnionitis is associated with decreased uterine activity, failure to progress in labor, and postpartum hemorrhage; however, clinical chorioamnionitis by itself is not an indication for cesarean delivery. Oxytocin is often administered for labor augmentation, and it is prudent to have uterotonic agents at hand to manage postpartum hemorrhage. Infants born to mothers with clinical chorioamnionitis near term are at risk for early-onset neonatal sepsis and for long-term disability such as cerebral palsy. A frontier is the noninvasive assessment of amniotic fluid to diagnose intraamniotic inflammation with a transcervical amniotic fluid collector and a rapid bedside test for IL-8 for patients with ruptured membranes. This approach promises to improve diagnostic accuracy and to provide a basis for antimicrobial administration.
Collapse
Affiliation(s)
- Eunjung Jung
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Obstetrics and Gynecology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI.
| | - Manaphat Suksai
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Francesca Gotsch
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Piya Chaemsaithong
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Obstetrics and Gynecology, Mahidol University, Faculty of Medicine, Ramathibodi Hospital, Bangkok, Thailand
| | - Offer Erez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Agustin Conde-Agudelo
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI
| | - Stanley M Berry
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Arun Meyyazhagan
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Centre of Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Bo Hyun Yoon
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea; Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
16
|
Manuel G, Coleman M, Orvis AS, Munson J, Li A, Kapur RP, Li M, Li E, Armistead B, Rajagopal L, Adams Waldorf KM. Spatial profiling of the placental chorioamniotic membranes reveals upregulation of immune checkpoint proteins during Group B Streptococcus infection in a nonhuman primate model. Front Cell Infect Microbiol 2024; 13:1299644. [PMID: 38239507 PMCID: PMC10794649 DOI: 10.3389/fcimb.2023.1299644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/05/2023] [Indexed: 01/22/2024] Open
Abstract
Background Preterm birth is a leading cause of neonatal mortality, which is often complicated by intrauterine infection and inflammation. We have established a nonhuman primate model of Group B Streptococcus (GBS, Streptococcus agalactiae) infection-associated preterm birth. Immune checkpoints are modulators of the immune response by activating or suppressing leukocyte function and are understudied in preterm birth. The objective of this study was to spatially profile changes in immune protein expression at the maternal-fetal interface during a GBS infection with a focus on immune checkpoints. Methods Twelve nonhuman primates (pigtail macaques, Macaca nemestrina) received a choriodecidual inoculation of either: 1) 1-5 X 108 colony forming units (CFU) of hyperhemolytic/hypervirulent GBS (GBSΔcovR, N=4); 2) an isogenic/nonpigmented strain (GBS ΔcovRΔcylE, N=4); or, 3) saline (N=4). A Cesarean section was performed at preterm labor or 3 days after GBS infection or 7 days after saline inoculation. Nanostring GeoMx® Digital Spatial Profiling technology was used to segment protein expression within the amnion, chorion, and maternal decidua at the inoculation site using an immuno-oncology panel targeting 56 immunoproteins enriched in stimulatory and inhibitory immune checkpoint proteins or their protein ligands. Statistical analysis included R studio, Kruskal-Wallis, Pearson and Spearman tests. Results Both inhibitory and stimulatory immune checkpoint proteins were significantly upregulated within the chorioamniotic membranes and decidua (VISTA, LAG3, PD-1, CD40, GITR), as well as their ligands (PD-L1, PD-L2, CD40L; all p<0.05). Immunostaining for VISTA revealed positive (VISTA+) cells, predominantly in the chorion and decidua. There were strong correlations between VISTA and amniotic fluid concentrations of IL-1β, IL-6, IL-8, and TNF-α (all p<0.05), as well as maternal placental histopathology scores (p<0.05). Conclusion Differential regulation of multiple immune checkpoint proteins in the decidua at the site of a GBS infection indicates a major perturbation in immunologic homeostasis that could benefit the host by restricting immune-driven pathologies or the pathogen by limiting immune surveillance. Protein expression of VISTA, an inhibitory immune checkpoint, was upregulated in the chorion and decidua after GBS infection. Investigating the impact of innate immune cell expression of inhibitory immune checkpoints may reveal new insights into placental host-pathogen interactions at the maternal-fetal interface.
Collapse
Affiliation(s)
- Gygeria Manuel
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- Morehouse School of Medicine, Atlanta, GA, United States
| | - Michelle Coleman
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA, United States
| | - Austyn S. Orvis
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA, United States
| | - Jeff Munson
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States
| | - Amanda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- Department of Biology, Case Western Reserve University, Cleveland, OH, United States
| | - Raj P. Kapur
- Department of Laboratory Medicine and Pathology, Seattle Children’s Hospital and University of Washington, Seattle, WA, United States
| | - Miranda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- School of Medicine, University of Washington, Seattle, WA, United States
| | - Edmunda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Blair Armistead
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA, United States
| | - Lakshmi Rajagopal
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Kristina M. Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
17
|
Galaz J, Romero R, Greenberg JM, Theis KR, Arenas-Hernandez M, Xu Y, Farias-Jofre M, Miller D, Kanninen T, Garcia-Flores V, Gomez-Lopez N. Host-microbiome interactions in distinct subsets of preterm labor and birth. iScience 2023; 26:108341. [PMID: 38047079 PMCID: PMC10692673 DOI: 10.1016/j.isci.2023.108341] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/06/2023] [Accepted: 10/23/2023] [Indexed: 12/05/2023] Open
Abstract
Preterm birth, the leading cause of perinatal morbidity, often follows premature labor, a syndrome whose prevention remains a challenge. To better understand the relationship between premature labor and host-microbiome interactions, we conducted a mechanistic investigation using three preterm birth models. We report that intra-amniotic delivery of LPS triggers inflammatory responses in the amniotic cavity and cervico-vaginal microenvironment, causing vaginal microbiome changes and signs of active labor. Intra-amniotic IL-1α delivery causes a moderate inflammatory response in the amniotic cavity but increasing inflammation in the cervico-vaginal space, leading to vaginal microbiome disruption and signs of active labor. Conversely, progesterone action blockade by RU-486 triggers local immune responses accompanying signs of active labor without altering the vaginal microbiome. Preterm labor facilitates ascension of cervico-vaginal bacteria into the amniotic cavity, regardless of stimulus. This study provides compelling mechanistic insights into the dynamic host-microbiome interactions within the cervico-vaginal microenvironment that accompany premature labor and birth.
Collapse
Affiliation(s)
- Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
| | - Jonathan M. Greenberg
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Kevin R. Theis
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Marcia Arenas-Hernandez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yi Xu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Marcelo Farias-Jofre
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tomi Kanninen
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Valeria Garcia-Flores
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
18
|
Kammala AK, Mosebarger A, Radnaa E, Rowlinson E, Vora N, Fortunato SJ, Sharma S, Safarzadeh M, Menon R. Extracellular Vesicles-mediated recombinant IL-10 protects against ascending infection-associated preterm birth by reducing fetal inflammatory response. Front Immunol 2023; 14:1196453. [PMID: 37600782 PMCID: PMC10437065 DOI: 10.3389/fimmu.2023.1196453] [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: 03/29/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Background Fetal inflammatory response mediated by the influx of immune cells and activation of pro-inflammatory transcription factor NF-κB in feto-maternal uterine tissues is the major determinant of infection-associated preterm birth (PTB, live births < 37 weeks of gestation). Objective To reduce the incidence of PTB by minimizing inflammation, extracellular vesicles (EVs) were electroporetically engineered to contain anti-inflammatory cytokine interleukin (IL)-10 (eIL-10), and their efficacy was tested in an ascending model of infection (vaginal administration of E. coli) induced PTB in mouse models. Study design EVs (size: 30-170 nm) derived from HEK293T cells were electroporated with recombinant IL-10 at 500 volts and 125 Ω, and 6 pulses to generate eIL-10. eIL-10 structural characters (electron microscopy, nanoparticle tracking analysis, ExoView [size and cargo content] and functional properties (co-treatment of macrophage cells with LPS and eIL-10) were assessed. To test efficacy, CD1 mice were vaginally inoculated with E. coli (1010CFU) and subsequently treated with either PBS, eIL-10 (500ng) or Gentamicin (10mg/kg) or a combination of eIL-10+gentamicin. Fetal inflammatory response in maternal and fetal tissues after the infection or treatment were conducted by suspension Cytometer Time of Flight (CyTOF) using a transgenic mouse model that express red fluorescent TdTomato (mT+) in fetal cells. Results Engineered EVs were structurally and functionally stable and showed reduced proinflammatory cytokine production from LPS challenged macrophage cells in vitro. Maternal administration of eIL-10 (10 µg/kg body weight) crossed feto-maternal barriers to delay E. coli-induced PTB to deliver live pups at term. Delay in PTB was associated with reduced feto-maternal uterine inflammation (immune cell infiltration and histologic chorioamnionitis, NF-κB activation, and proinflammatory cytokine production). Conclusions eIL-10 administration was safe, stable, specific, delayed PTB by over 72 hrs and delivered live pups. The delivery of drugs using EVs overcomes the limitations of in-utero fetal interventions. Protecting IL-10 in EVs eliminates the need for the amniotic administration of recombinant IL-10 for its efficacy.
Collapse
Affiliation(s)
- Ananth Kumar Kammala
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Angela Mosebarger
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Enkhtuya Radnaa
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Emma Rowlinson
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Natasha Vora
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Stephen J. Fortunato
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Surendra Sharma
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Providence, RI, United States
| | - Melody Safarzadeh
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| |
Collapse
|
19
|
Zhang C, Cao J, Xu M, Wu D, Li W, Chang Y. The role of neutrophils in chorioamnionitis. Front Immunol 2023; 14:1198831. [PMID: 37475854 PMCID: PMC10354368 DOI: 10.3389/fimmu.2023.1198831] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/16/2023] [Indexed: 07/22/2023] Open
Abstract
Chorioamnionitis, commonly referred to as intrauterine infection or inflammation, is pathologically defined by neutrophil infiltration and inflammation at the maternal-fetal interface. Chorioamnionitis is the common complication during late pregnancy, which lead to a series of serious consequences, such as preterm labor, preterm premature rupture of the fetal membranes, and fetal inflammatory response syndrome. During infection, a large number of neutrophils migrate to the chorio-decidua in response to chemokines. Although neutrophils, a crucial part of innate immune cells, have strong anti-inflammatory properties, over-activating them can harm the body while also eliminating pathogens. This review concentrated on the latest studies on chorioamnionitis-related consequences as well as the function and malfunction of neutrophils. The release of neutrophil extracellular traps, production of reactive oxygen species, and degranulation from neutrophils during intrauterine infection, as well as their pathological roles in complications related to chorioamnionitis, were discussed in detail, offering fresh perspectives on the treatment of chorioamnionitis.
Collapse
Affiliation(s)
| | | | | | | | | | - Ying Chang
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin central hospital of Gynecology Obstetrics, Tianjin, China
| |
Collapse
|
20
|
Furuta A, Coleman M, Casares R, Seepersaud R, Orvis A, Brokaw A, Quach P, Nguyen S, Sweeney E, Sharma K, Wallen G, Sanghavi R, Mateos-Gil J, Cuerva JM, Millán A, Rajagopal L. CD1 and iNKT cells mediate immune responses against the GBS hemolytic lipid toxin induced by a non-toxic analog. PLoS Pathog 2023; 19:e1011490. [PMID: 37384812 DOI: 10.1371/journal.ppat.1011490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 06/14/2023] [Indexed: 07/01/2023] Open
Abstract
Although hemolytic lipids have been discovered from many human pathogens including Group B Streptococcus (GBS), strategies that neutralize their function are lacking. GBS is a leading cause of pregnancy-associated neonatal infections, and adult GBS infections are on the rise. The GBS hemolytic lipid toxin or granadaene, is cytotoxic to many immune cells including T and B cells. We previously showed that mice immunized with a synthetic nontoxic analog of granadaene known as R-P4 had reduced bacterial dissemination during systemic infection. However, mechanisms important for R-P4 mediated immune protection was not understood. Here, we show that immune serum from R-P4-immunized mice facilitate GBS opsonophagocytic killing and protect naïve mice from GBS infection. Further, CD4+ T cells isolated from R-P4-immunized mice proliferated in response to R-P4 stimulation in a CD1d- and iNKT cell-dependent manner. Consistent with these observations, R-P4 immunized mice lacking CD1d or CD1d-restricted iNKT cells exhibit elevated bacterial burden. Additionally, adoptive transfer of iNKT cells from R-P4 vaccinated mice significantly reduced GBS dissemination compared to adjuvant controls. Finally, maternal R-P4 vaccination provided protection against ascending GBS infection during pregnancy. These findings are relevant in the development of therapeutic strategies targeting lipid cytotoxins.
Collapse
Affiliation(s)
- Anna Furuta
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Michelle Coleman
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Raquel Casares
- Department of Organic Chemistry, University of Granada, Granada, Spain
| | - Ravin Seepersaud
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Austyn Orvis
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Alyssa Brokaw
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Phoenicia Quach
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Shayla Nguyen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Erin Sweeney
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Kavita Sharma
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Grace Wallen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Rhea Sanghavi
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Jaime Mateos-Gil
- Department of Organic Chemistry, University of Granada, Granada, Spain
| | | | - Alba Millán
- Department of Organic Chemistry, University of Granada, Granada, Spain
| | - Lakshmi Rajagopal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
21
|
Keestra-Gounder AM, Nagao PE. Inflammasome activation by Gram-positive bacteria: Mechanisms of activation and regulation. Front Immunol 2023; 14:1075834. [PMID: 36761775 PMCID: PMC9902775 DOI: 10.3389/fimmu.2023.1075834] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/06/2023] [Indexed: 01/26/2023] Open
Abstract
The inflammasomes are intracellular multimeric protein complexes consisting of an innate immune sensor, the adapter protein ASC and the inflammatory caspases-1 and/or -11 and are important for the host defense against pathogens. Activaton of the receptor leads to formation of the inflammasomes and subsequent processing and activation of caspase-1 that cleaves the proinflammatory cytokines IL-1β and IL-18. Active caspase-1, and in some instances caspase-11, cleaves gasdermin D that translocates to the cell membrane where it forms pores resulting in the cell death program called pyroptosis. Inflammasomes can detect a range of microbial ligands through direct interaction or indirectly through diverse cellular processes including changes in ion fluxes, production of reactive oxygen species and disruption of various host cell functions. In this review, we will focus on the NLRP3, NLRP6, NLRC4 and AIM2 inflammasomes and how they are activated and regulated during infections with Gram-positive bacteria, including Staphylococcus spp., Streptococcus spp. and Listeria monocytogenes.
Collapse
Affiliation(s)
- A. Marijke Keestra-Gounder
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Prescilla Emy Nagao
- Laboratory of Molecular Biology and Physiology of Streptococci, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Group B Streptococcus: Virulence Factors and Pathogenic Mechanism. Microorganisms 2022; 10:microorganisms10122483. [PMID: 36557736 PMCID: PMC9784991 DOI: 10.3390/microorganisms10122483] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Group B Streptococcus (GBS) or Streptococcus agalactiae is a major cause of neonatal mortality. When colonizing the lower genital tract of pregnant women, GBS may cause premature birth and stillbirth. If transmitted to the newborn, it may result in life-threatening illnesses, including sepsis, meningitis, and pneumonia. Moreover, through continuous evolution, GBS can use its original structure and unique factors to greatly improve its survival rate in the human body. This review discusses the key virulence factors that facilitate GBS invasion and colonization and their action mechanisms. A comprehensive understanding of the role of virulence factors in GBS infection is crucial to develop better treatment options and screen potential candidate molecules for the development of the vaccine.
Collapse
|
23
|
de Jesus Gonzalez-Contreras F, Zarate X. Neutrophil extracellular traps: Modulation mechanisms by pathogens. Cell Immunol 2022; 382:104640. [PMID: 36413806 DOI: 10.1016/j.cellimm.2022.104640] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022]
Abstract
Neutrophils, as innate effector cells, play an essential role in the containment and elimination of pathogens. Among the main neutrophil mechanisms use for these processes is the release of neutrophil extracellular traps (NETs), which consist of decondensed DNA decorated with various cytoplasmic proteins. NETs' principal role is the trapping and elimination of infectious agents; therefore, the formation of NETs is regulated by bacteria, fungi, parasites, and viruses through different mechanisms: the presence of virulence factors (adhered or secreted), microbial load, size of the microorganism, and even due to other immune cells activation (mainly platelets). This review summarizes the significant aspects that contribute to NETs modulation by pathogens and their components, and the effect NETs have on these pathogens as a cellular defense mechanism.
Collapse
Affiliation(s)
| | - Xristo Zarate
- Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n, San Nicolas de los Garza 66455, NL, Mexico
| |
Collapse
|
24
|
Huebner EM, Gudjónsdóttir MJ, Dacanay MB, Nguyen S, Brokaw A, Sharma K, Elfvin A, Hentz E, Rivera YR, Burd N, Shivakumar M, Coler B, Li M, Li A, Munson J, Orvis A, Coleman M, Jacobsson B, Rajagopal L, Adams Waldorf KM. Virulence, phenotype and genotype characteristics of invasive group B Streptococcus isolates obtained from Swedish pregnant women and neonates. Ann Clin Microbiol Antimicrob 2022; 21:43. [PMID: 36229877 PMCID: PMC9560721 DOI: 10.1186/s12941-022-00534-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
Group B streptococci (GBS) are bacteria that can cause preterm birth and invasive neonatal disease. Heterogeneous expression of virulence factors enables GBS to exist as both commensal bacteria and to become highly invasive. A molecular epidemiological study comparing GBS bacterial traits, genotype and host characteristics may indicate whether it is possible to predict the risk of perinatal invasive GBS disease and more accurately target intrapartum antibiotic prophylaxis. A total of 229 invasive GBS isolates from Swedish pregnant women or neonates were assessed for virulence and phenotypic traits: hemolysis zone, hemolytic pigment (Granada agar), Streptococcus B Carrot Broth (SBCB) assay, CAMP factor, and hyaluronidase activity. Genes regulating hemolytic pigment synthesis (covR/covS, abx1, stk1, stp1) were sequenced. Of the virulence factors and phenotypes assessed, a Granada pigment or SBCB score ≥ 2 captured more than 90% of EOD isolates with excellent inter-rater reliability. High enzyme activity of hyaluronidase was observed in 16% (36/229) of the invasive GBS isolates and notably, in one case of stillbirth. Hyaluronidase activity was also significantly higher in GBS isolates obtained from pregnant/postpartum individuals versus the stillbirth or neonatal invasive isolates (p < 0.001). Sequencing analysis found that abx1 (g.T106I), stk1 (g.T211N), stp1 (g.K469R) and covS (g.V343M) variants were present significantly more often in the higher (Granada pigment score ≥ 2) versus lower pigmented isolates (p < 0.001, each variant). Among the 203 higher Granada pigment scoring isolates, 22 (10.8%) isolates had 3 of the four sequence variants and 10 (4.9%) had 2 of the four sequence variants. Although heterogeneity in GBS virulence factor expression was observed, the vast majority were more highly pigmented and contained several common sequence variants in genes regulating pigment synthesis. High activity of hyaluronidase may increase risk for stillbirth and invasive disease in pregnant or postpartum individuals. Our findings suggest that testing for GBS pigmentation and hyaluronidase may, albeit imperfectly, identify pregnant people at risk for invasive disease and represent a step towards a personalized medical approach for the administration of intrapartum antibiotic prophylaxis.
Collapse
Affiliation(s)
- Emily M. Huebner
- grid.34477.330000000122986657School of Medicine, University of Washington, Seattle, WA USA ,grid.34477.330000000122986657Department of Obstetrics and Gynecology, University of Washington, Seattle, WA USA
| | - Margrét Johansson Gudjónsdóttir
- grid.8761.80000 0000 9919 9582Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden ,grid.1649.a000000009445082XDepartment of Pediatrics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Matthew B. Dacanay
- grid.34477.330000000122986657Department of Obstetrics and Gynecology, University of Washington, Seattle, WA USA
| | - Shayla Nguyen
- grid.240741.40000 0000 9026 4165Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA USA
| | - Alyssa Brokaw
- grid.240741.40000 0000 9026 4165Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA USA
| | - Kavita Sharma
- grid.240741.40000 0000 9026 4165Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA USA
| | - Anders Elfvin
- grid.8761.80000 0000 9919 9582Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden ,grid.1649.a000000009445082XDepartment of Pediatrics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Elisabet Hentz
- grid.8761.80000 0000 9919 9582Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden ,grid.1649.a000000009445082XDepartment of Pediatrics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ysabella Raceli Rivera
- grid.34477.330000000122986657Department of Obstetrics and Gynecology, University of Washington, Seattle, WA USA
| | - Nicole Burd
- grid.34477.330000000122986657Department of Obstetrics and Gynecology, University of Washington, Seattle, WA USA
| | - Megana Shivakumar
- grid.34477.330000000122986657Department of Obstetrics and Gynecology, University of Washington, Seattle, WA USA
| | - Brahm Coler
- grid.34477.330000000122986657Department of Obstetrics and Gynecology, University of Washington, Seattle, WA USA ,grid.30064.310000 0001 2157 6568Elson S. Floyd College of Medicine, Washington State University, Spokane, WA USA
| | - Miranda Li
- grid.34477.330000000122986657Department of Obstetrics and Gynecology, University of Washington, Seattle, WA USA ,grid.21729.3f0000000419368729Columbia University, New York, NY USA
| | - Amanda Li
- grid.34477.330000000122986657Department of Obstetrics and Gynecology, University of Washington, Seattle, WA USA
| | - Jeff Munson
- grid.34477.330000000122986657Department of Psychiatry and Behavior Sciences, University of Washington, Seattle, WA USA
| | - Austyn Orvis
- grid.240741.40000 0000 9026 4165Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA USA
| | - Michelle Coleman
- grid.240741.40000 0000 9026 4165Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA USA
| | - Bo Jacobsson
- grid.1649.a000000009445082XDepartment of Pediatrics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden ,grid.8761.80000 0000 9919 9582Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lakshmi Rajagopal
- grid.240741.40000 0000 9026 4165Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA USA ,grid.34477.330000000122986657Department of Pediatrics, University of Washington, Seattle, WA USA
| | - Kristina M. Adams Waldorf
- grid.34477.330000000122986657Department of Obstetrics and Gynecology, University of Washington, Seattle, WA USA ,grid.8761.80000 0000 9919 9582Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
25
|
Gomez-Lopez N, Galaz J, Miller D, Farias-Jofre M, Liu Z, Arenas-Hernandez M, Garcia-Flores V, Shaffer Z, Greenberg J, Theis KR, Romero R. The immunobiology of preterm labor and birth: intra-amniotic inflammation or breakdown of maternal-fetal homeostasis. Reproduction 2022; 164:R11-R45. [PMID: 35559791 PMCID: PMC9233101 DOI: 10.1530/rep-22-0046] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/13/2022] [Indexed: 11/08/2022]
Abstract
In brief The syndrome of preterm labor comprises multiple established and novel etiologies. This review summarizes the distinct immune mechanisms implicated in preterm labor and birth and highlights potential strategies for its prevention. Abstract Preterm birth, the leading cause of neonatal morbidity and mortality worldwide, results from preterm labor, a syndrome that includes multiple etiologies. In this review, we have summarized the immune mechanisms implicated in intra-amniotic inflammation, the best-characterized cause of preterm labor and birth, as well as novel etiologies non-associated with intra-amniotic inflammation (i.e. formally known as idiopathic). While the intra-amniotic inflammatory responses driven by microbes (infection) or alarmins (sterile) have some overlap in the participating cellular and molecular processes, the distinct natures of these two conditions necessitate the implementation of specific approaches to prevent adverse pregnancy and neonatal outcomes. Intra-amniotic infection can be treated with the correct antibiotics, whereas sterile intra-amniotic inflammation could potentially be treated by administering a combination of anti-inflammatory drugs (e.g. betamethasone, inflammasome inhibitors, etc.). Recent evidence also supports the role of fetal T-cell activation as a newly described trigger for preterm labor and birth in a subset of cases diagnosed as idiopathic. Moreover, herein we also provide evidence of two maternally-driven immune mechanisms responsible for preterm births formerly considered to be idiopathic. First, the impairment of maternal Tregs can lead to preterm birth, likely due to the loss of immunosuppressive activity resulting in unleashed effector T-cell responses. Secondly, homeostatic macrophages were shown to be essential for maintaining pregnancy and promoting fetal development, and the adoptive transfer of homeostatic M2-polarized macrophages shows great promise for preventing inflammation-induced preterm birth. Collectively, in this review, we discuss the established and novel immune mechanisms responsible for preterm birth and highlight the potential targets for novel strategies aimed at preventing the multi-etiological syndrome of preterm labor leading to preterm birth.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, Maryland, 20892 and Detroit, Michigan, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Jose Galaz
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, Maryland, 20892 and Detroit, Michigan, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, Maryland, 20892 and Detroit, Michigan, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Marcelo Farias-Jofre
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, Maryland, 20892 and Detroit, Michigan, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Zhenjie Liu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, Maryland, 20892 and Detroit, Michigan, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Marcia Arenas-Hernandez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, Maryland, 20892 and Detroit, Michigan, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, Maryland, 20892 and Detroit, Michigan, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Zachary Shaffer
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, Maryland, 20892 and Detroit, Michigan, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | - Jonathan Greenberg
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, Maryland, 20892 and Detroit, Michigan, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Kevin R. Theis
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, Maryland, 20892 and Detroit, Michigan, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, Maryland, 20892 and Detroit, Michigan, 48201, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, 48824, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, 48201, USA
- Detroit Medical Center, Detroit, Michigan, 48201, USA
| |
Collapse
|
26
|
Jahn K, Shumba P, Quach P, Müsken M, Wesche J, Greinacher A, Rajagopal L, Hammerschmidt S, Siemens N. Group B Streptococcal Hemolytic Pigment Impairs Platelet Function in a Two-Step Process. Cells 2022; 11:cells11101637. [PMID: 35626674 PMCID: PMC9139542 DOI: 10.3390/cells11101637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 11/22/2022] Open
Abstract
Group B streptococci (GBS) cause a range of invasive maternal-fetal diseases during pregnancy and post-partum. However, invasive infections in non-pregnant adults are constantly increasing. These include sepsis and streptococcal toxic shock syndrome, which are often complicated by systemic coagulation and thrombocytopenia. GBS express a hyper-hemolytic ornithine rhamnolipid pigment toxin with cytolytic and coagulatory activity. Here, we investigated the effects of GBS pigment on human platelets. Infections of platelets with pigmented GBS resulted initially in platelet activation, followed by necrotic cell death. Thus, this study shows that GBS pigment kills human platelets.
Collapse
Affiliation(s)
- Kristin Jahn
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, 17489 Greifswald, Germany; (K.J.); (P.S.)
| | - Patience Shumba
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, 17489 Greifswald, Germany; (K.J.); (P.S.)
| | - Phoenicia Quach
- Department of Global Health, University of Washington, Seattle, WA 98105, USA; (P.Q.); (L.R.)
| | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;
| | - Jan Wesche
- Department of Transfusion Medicine, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, 17475 Greifswald, Germany; (J.W.); (A.G.)
| | - Andreas Greinacher
- Department of Transfusion Medicine, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, 17475 Greifswald, Germany; (J.W.); (A.G.)
| | - Lakshmi Rajagopal
- Department of Global Health, University of Washington, Seattle, WA 98105, USA; (P.Q.); (L.R.)
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98019, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98105, USA
| | - Sven Hammerschmidt
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, 17489 Greifswald, Germany; (K.J.); (P.S.)
- Correspondence: (S.H.); (N.S.); Tel.: +49-3-834-420-5701 (S.H.); +49-3-834-420-5711 (N.S.)
| | - Nikolai Siemens
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, 17489 Greifswald, Germany; (K.J.); (P.S.)
- Correspondence: (S.H.); (N.S.); Tel.: +49-3-834-420-5701 (S.H.); +49-3-834-420-5711 (N.S.)
| |
Collapse
|
27
|
Ma F, Yang S, Zhou M, Lu Y, Deng B, Zhang J, Fan H, Wang G. NADPH oxidase-derived reactive oxygen species production activates the ERK1/2 pathway in neutrophil extracellular traps formation by Streptococcus agalactiae isolated from clinical mastitis bovine. Vet Microbiol 2022; 268:109427. [PMID: 35405476 DOI: 10.1016/j.vetmic.2022.109427] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 03/08/2022] [Accepted: 04/01/2022] [Indexed: 11/29/2022]
Abstract
Streptococcus agalactiae (S. agalactiae) continues to be challenging for milk quality in some countries and leads to huge economic losses. A large number of neutrophils are recruited into inflammatory foci when S. agalactiae infection occurs, and most studies have focused on the interaction between neutrophil extracellular traps (NETs) and this bacterium in the context of human pathogenicity. However, there is little information on the NET formation mechanism induced by S. agalactiae in the context of bovine mastitis. Here, neutrophils isolated from BALB/c mice were infected with S. agalactiae SAG-FX17, and NET formation was evaluated. SAG-FX17 could induce NADPH oxidase-derived reactive oxygen species (NOX-ROS)-dependent NET formation, and 21.8% of bacteria could be eliminated by NETs via NET DNA and associated proteins. SAG-FX17 could induce the phosphorylation of p38 MAPK, ERK1/2 MAPK, and JNK/SAPK in neutrophils. However, only ERK1/2 MAPK was shown to play an important role in SAG-FX17-induced NET formation. Importantly, NOX-ROS production occurs upstream of ERK1/2 MAPK activation and then induces NET release. ERK1/2 MAPK phosphorylation can, in turn, enhance NOX-ROS generation, which further contributes to NET release and bacterial elimination. This study provides evidence of the molecular mechanism underlying serotype Ia S. agalactiae SAG-FX17-induced NET formation and the interaction between bacteria and NETs, and these findings will increase our knowledge about bacterial mastitis in dairy cattle and contribute to the prevention and clinical treatment of bovine mastitis.
Collapse
Affiliation(s)
- Fang Ma
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing 210014, China
| | - Shifang Yang
- College of Veterinary Medicine, Nanjing Agriculture University, Nanjing, China
| | - Mingxu Zhou
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Yu Lu
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Bihua Deng
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Jinqiu Zhang
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Hongjie Fan
- College of Veterinary Medicine, Nanjing Agriculture University, Nanjing, China
| | - Guangyu Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
28
|
Furuta A, Brokaw A, Manuel G, Dacanay M, Marcell L, Seepersaud R, Rajagopal L, Adams Waldorf K. Bacterial and Host Determinants of Group B Streptococcal Infection of the Neonate and Infant. Front Microbiol 2022; 13:820365. [PMID: 35265059 PMCID: PMC8899651 DOI: 10.3389/fmicb.2022.820365] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/27/2022] [Indexed: 12/15/2022] Open
Abstract
Group B streptococci (GBS) are Gram-positive β-hemolytic bacteria that can cause serious and life-threatening infections in neonates manifesting as sepsis, pneumonia, meningitis, osteomyelitis, and/or septic arthritis. Invasive GBS infections in neonates in the first week of life are referred to as early-onset disease (EOD) and thought to be acquired by the fetus through exposure to GBS in utero or to vaginal fluids during birth. Late-onset disease (LOD) refers to invasive GBS infections between 7 and 89 days of life. LOD transmission routes are incompletely understood, but may include breast milk, household contacts, nosocomial, or community sources. Invasive GBS infections and particularly meningitis may result in significant neurodevelopmental injury and long-term disability that persists into childhood and adulthood. Globally, EOD and LOD occur in more than 300,000 neonates and infants annually, resulting in 90,000 infant deaths and leaving more than 10,000 infants with a lifelong disability. In this review, we discuss the clinical impact of invasive GBS neonatal infections and then summarize virulence and host factors that allow the bacteria to exploit the developing neonatal immune system and target organs. Specifically, we consider the mechanisms known to enable GBS invasion into the neonatal lung, blood vessels and brain. Understanding mechanisms of GBS invasion and pathogenesis relevant to infections in the neonate and infant may inform the development of therapeutics to prevent or mitigate injury, as well as improve risk stratification.
Collapse
Affiliation(s)
- Anna Furuta
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Alyssa Brokaw
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Gygeria Manuel
- Morehouse School of Medicine, Atlanta, GA, United States
| | - Matthew Dacanay
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
| | - Lauren Marcell
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
| | - Ravin Seepersaud
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Lakshmi Rajagopal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Kristina Adams Waldorf
- Department of Global Health, University of Washington, Seattle, WA, United States.,Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States.,Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
29
|
Shahi I, Llaneras CN, Perelman SS, Torres VJ, Ratner AJ. Genome-Wide CRISPR-Cas9 Screen Does Not Identify Host Factors Modulating Streptococcus agalactiae β-Hemolysin/Cytolysin-Induced Cell Death. Microbiol Spectr 2022; 10:e0218621. [PMID: 35196804 PMCID: PMC8865549 DOI: 10.1128/spectrum.02186-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/07/2022] [Indexed: 11/20/2022] Open
Abstract
Pore-forming toxins (PFTs) are commonly produced by pathogenic bacteria, and understanding them is key to the development of virulence-targeted therapies. Streptococcus agalactiae, or group B Streptococcus (GBS), produces several factors that enhance its pathogenicity, including the PFT β-hemolysin/cytolysin (βhc). Little is understood about the cellular factors involved in βhc pore formation. We conducted a whole-genome CRISPR-Cas9 forward genetic screen to identify host genes that might contribute to βhc pore formation and cell death. While the screen identified the established receptor, CD59, in control experiments using the toxin intermedilysin (ILY), no clear candidate genes were identified that were required for βhc-mediated lethality. Of the top targets from the screen, two genes involved in membrane remodeling and repair represented candidates that might modulate the kinetics of βhc-induced cell death. Upon attempted validation of the results using monoclonal cell lines with targeted disruption of these genes, no effect on βhc-mediated cell lysis was observed. The CRISPR-Cas9 screen results are consistent with the hypothesis that βhc does not require a single nonessential host factor to mediate target cell death. IMPORTANCE CRISPR-Cas9 forward genetic screens have been used to identify host cell targets required by bacterial toxins. They have been used successfully to both verify known targets and elucidate novel host factors required by toxins. Here, we show that this approach fails to identify host factors required for cell death due to βhc, a toxin required for GBS virulence. These data suggest that βhc may not require a host cell receptor for toxin function or may require a host receptor that is an essential gene and would not be identified using this screening strategy.
Collapse
Affiliation(s)
- Ifrah Shahi
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Cristina N. Llaneras
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Sofya S. Perelman
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
| | - Adam J. Ratner
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Department of Pediatrics, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
30
|
Ma X, Wu M, Wang C, Li H, Fan A, Wang Y, Han C, Xue F. The pathogenesis of prevalent aerobic bacteria in aerobic vaginitis and adverse pregnancy outcomes: a narrative review. Reprod Health 2022; 19:21. [PMID: 35090514 PMCID: PMC8796570 DOI: 10.1186/s12978-021-01292-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/15/2021] [Indexed: 01/08/2023] Open
Abstract
Background Aerobic vaginitis is a common cause of vaginal discharge in reproductive-age women, increasing the risk of negative pregnancy outcomes such as premature delivery, abortion, premature rupture of membranes and stillbirth. However, the aetiology and pathogenesis of aerobic vaginitis causing negative pregnancy outcomes are still unclear, and there is no unified and standardized treatment method for aerobic vaginitis in the pregnancy period. Methods We conducted a literature search of published studies in the English language focusing on aerobic vaginitis and its association with adverse pregnancy outcomes utilizing PubMed and Web of Science from January 1973 through June 2021. The common pathogenic bacteria of aerobic vaginitis during pregnancy, such as group B Streptococcus, Escherichia coli, Staphylococcus aureus, Enterococcus faecalis and Klebsiella pneumoniae, as well as the related adverse pregnancy outcomes and existing treatments were reviewed. Results A total of 4534 articles were identified, and 97 studies that had inclusion criteria were subjected to careful review. The pathogenic bacteria of aerobic vaginitis can produce different toxins or affect the local immunity of patients and then lead to the occurrence of infection. Fresh wet mount microscopy is the preferred diagnostic method for aerobic vaginitis. Clindamycin is a common antibiotic used for aerobic vaginitis in pregnant women. The use of products combining probiotics has achieved excellent treatment success. Conclusions Future research in this field can provide insights regarding the mechanism of aerobic vaginitis-induced adverse pregnancy outcomes in humans and ways to prevent their occurrence. Aerobic vaginitis is an infection of the vagina that increases the risk of negative pregnancy outcomes. The aetiology and pathogenesis of aerobic vaginitis causing negative pregnancy outcomes are still unclear. This paper reviews the common pathogenic bacteria of aerobic vaginitis during pregnancy, and the related adverse pregnancy outcomes. We also review the existing treatment. Currently, it is believed that the microflora in aerobic vaginitis is composed of commensal aerobic microorganisms of intestinal origin, and the most frequently encountered bacteria are group B Streptococcus, Escherichia coli, Staphylococcus aureus, Enterococcus faecalis and Klebsiella pneumoniae. The pathogenic bacteria of aerobic vaginitis can produce different toxins or affect the local immunity of patients and then lead to the occurrence of infection. Fresh wet mount microscopy is the preferred diagnostic method for aerobic vaginitis. Clindamycin is a common antibiotic used for aerobic vaginitis in pregnant women. The use of products combining probiotics has achieved excellent treatment success. This study provides a reference for future research and early diagnosis and treatment during pregnancy. Future research in this field can provide insights regarding the mechanisms of aerobic vaginitis-induced adverse pregnancy outcomes in humans and ways to prevent their occurrence.
Collapse
Affiliation(s)
- Xiaotong Ma
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, China
| | - Ming Wu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, China
| | - Chen Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, China
| | - Huiyang Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, China
| | - Aiping Fan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, China
| | - Cha Han
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China. .,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, China.
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China. .,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, China.
| |
Collapse
|
31
|
Brokaw A, Furuta A, Dacanay M, Rajagopal L, Adams Waldorf KM. Bacterial and Host Determinants of Group B Streptococcal Vaginal Colonization and Ascending Infection in Pregnancy. Front Cell Infect Microbiol 2021; 11:720789. [PMID: 34540718 PMCID: PMC8446444 DOI: 10.3389/fcimb.2021.720789] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022] Open
Abstract
Group B streptococcus (GBS) is a gram-positive bacteria that asymptomatically colonizes the vaginal tract. However, during pregnancy maternal GBS colonization greatly predisposes the mother and baby to a wide range of adverse outcomes, including preterm birth (PTB), stillbirth, and neonatal infection. Although many mechanisms involved in GBS pathogenesis are partially elucidated, there is currently no approved GBS vaccine. The development of a safe and effective vaccine that can be administered during or prior to pregnancy remains a principal objective in the field, because current antibiotic-based therapeutic strategies do not eliminate all cases of invasive GBS infections. Herein, we review our understanding of GBS disease pathogenesis at the maternal-fetal interface with a focus on the bacterial virulence factors and host defenses that modulate the outcome of infection. We follow GBS along its path from an asymptomatic colonizer of the vagina to an invasive pathogen at the maternal-fetal interface, noting factors critical for vaginal colonization, ascending infection, and vertical transmission to the fetus. Finally, at each stage of infection we emphasize important host-pathogen interactions, which, if targeted therapeutically, may help to reduce the global burden of GBS.
Collapse
Affiliation(s)
- Alyssa Brokaw
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Anna Furuta
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Matthew Dacanay
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
| | - Lakshmi Rajagopal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Kristina M Adams Waldorf
- Department of Global Health, University of Washington, Seattle, WA, United States.,Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States.,Department of Obstetrics and Gynecology, University of Washington and Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
32
|
Cappelletti M, Presicce P, Feiyang M, Senthamaraikannan P, Miller LA, Pellegrini M, Sim MS, Jobe AH, Divanovic S, Way SS, Chougnet CA, Kallapur SG. The induction of preterm labor in rhesus macaques is determined by the strength of immune response to intrauterine infection. PLoS Biol 2021; 19:e3001385. [PMID: 34495952 PMCID: PMC8452070 DOI: 10.1371/journal.pbio.3001385] [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: 01/14/2021] [Revised: 09/20/2021] [Accepted: 08/04/2021] [Indexed: 12/18/2022] Open
Abstract
Intrauterine infection/inflammation (IUI) is a major contributor to preterm labor (PTL). However, IUI does not invariably cause PTL. We hypothesized that quantitative and qualitative differences in immune response exist in subjects with or without PTL. To define the triggers for PTL, we developed rhesus macaque models of IUI driven by lipopolysaccharide (LPS) or live Escherichia coli. PTL did not occur in LPS challenged rhesus macaques, while E. coli–infected animals frequently delivered preterm. Although LPS and live E. coli both caused immune cell infiltration, E. coli–infected animals showed higher levels of inflammatory mediators, particularly interleukin 6 (IL-6) and prostaglandins, in the chorioamnion-decidua and amniotic fluid (AF). Neutrophil infiltration in the chorio-decidua was a common feature to both LPS and E. coli. However, neutrophilic infiltration and IL6 and PTGS2 expression in the amnion was specifically induced by live E. coli. RNA sequencing (RNA-seq) analysis of fetal membranes revealed that specific pathways involved in augmentation of inflammation including type I interferon (IFN) response, chemotaxis, sumoylation, and iron homeostasis were up-regulated in the E. coli group compared to the LPS group. Our data suggest that the intensity of the host immune response to IUI may determine susceptibility to PTL.
Collapse
Affiliation(s)
- Monica Cappelletti
- Divisions of Neonatology and Developmental Biology, UCLA Mattel Children’s Hospital, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, UCLA Mattel Children’s Hospital, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Ma Feiyang
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- Institute for Quantitative and Computational Biosciences–Collaboratory, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Paranthaman Senthamaraikannan
- Division of Neonatology and Pulmonary Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Lisa A. Miller
- California National Primate Research Center, University of California Davis, Davis, California, United States of America
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- Institute for Quantitative and Computational Biosciences–Collaboratory, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Myung S. Sim
- Department of Medicine Statistics Core, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Alan H. Jobe
- Division of Neonatology and Pulmonary Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Senad Divanovic
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Sing Sing Way
- Infectious Diseases, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Claire A. Chougnet
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Suhas G. Kallapur
- Divisions of Neonatology and Developmental Biology, UCLA Mattel Children’s Hospital, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
33
|
Li M, Brokaw A, Furuta AM, Coler B, Obregon-Perko V, Chahroudi A, Wang HY, Permar SR, Hotchkiss CE, Golos TG, Rajagopal L, Adams Waldorf KM. Non-human Primate Models to Investigate Mechanisms of Infection-Associated Fetal and Pediatric Injury, Teratogenesis and Stillbirth. Front Genet 2021; 12:680342. [PMID: 34290739 PMCID: PMC8287178 DOI: 10.3389/fgene.2021.680342] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/25/2021] [Indexed: 12/25/2022] Open
Abstract
A wide array of pathogens has the potential to injure the fetus and induce teratogenesis, the process by which mutations in fetal somatic cells lead to congenital malformations. Rubella virus was the first infectious disease to be linked to congenital malformations due to an infection in pregnancy, which can include congenital cataracts, microcephaly, hearing impairment and congenital heart disease. Currently, human cytomegalovirus (HCMV) is the leading infectious cause of congenital malformations globally, affecting 1 in every 200 infants. However, our knowledge of teratogenic viruses and pathogens is far from complete. New emerging infectious diseases may induce teratogenesis, similar to Zika virus (ZIKV) that caused a global pandemic in 2016-2017; thousands of neonates were born with congenital microcephaly due to ZIKV exposure in utero, which also included a spectrum of injuries to the brain, eyes and spinal cord. In addition to congenital anomalies, permanent injury to fetal and neonatal organs, preterm birth, stillbirth and spontaneous abortion are known consequences of a broader group of infectious diseases including group B streptococcus (GBS), Listeria monocytogenes, Influenza A virus (IAV), and Human Immunodeficiency Virus (HIV). Animal models are crucial for determining the mechanism of how these various infectious diseases induce teratogenesis or organ injury, as well as testing novel therapeutics for fetal or neonatal protection. Other mammalian models differ in many respects from human pregnancy including placentation, labor physiology, reproductive tract anatomy, timeline of fetal development and reproductive toxicology. In contrast, non-human primates (NHP) most closely resemble human pregnancy and exhibit key similarities that make them ideal for research to discover the mechanisms of injury and for testing vaccines and therapeutics to prevent teratogenesis, fetal and neonatal injury and adverse pregnancy outcomes (e.g., stillbirth or spontaneous abortion). In this review, we emphasize key contributions of the NHP model pre-clinical research for ZIKV, HCMV, HIV, IAV, L. monocytogenes, Ureaplasma species, and GBS. This work represents the foundation for development and testing of preventative and therapeutic strategies to inhibit infectious injury of human fetuses and neonates.
Collapse
Affiliation(s)
- Miranda Li
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
- Department of Biological Sciences, Columbia University, New York, NY, United States
| | - Alyssa Brokaw
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Anna M. Furuta
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Brahm Coler
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Veronica Obregon-Perko
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, GA, United States
| | - Hsuan-Yuan Wang
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Charlotte E. Hotchkiss
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Thaddeus G. Golos
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Lakshmi Rajagopal
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Kristina M. Adams Waldorf
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
34
|
McCartney SA, Kapur R, Liggitt HD, Baldessari A, Coleman M, Orvis A, Ogle J, Katz R, Rajagopal L, Adams Waldorf KM. Amniotic fluid interleukin 6 and interleukin 8 are superior predictors of fetal lung injury compared with maternal or fetal plasma cytokines or placental histopathology in a nonhuman primate model. Am J Obstet Gynecol 2021; 225:89.e1-89.e16. [PMID: 33412130 DOI: 10.1016/j.ajog.2020.12.1214] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/20/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Intra-amniotic infection or inflammation is common in early preterm birth and associated with substantial neonatal lung morbidity owing to fetal exposure to proinflammatory cytokines and infectious organisms. Amniotic fluid interleukin 8, a proinflammatory cytokine, was previously correlated with the development of neonatal bronchopulmonary dysplasia, but whether amniotic fluid cytokines or placental pathology more accurately predicts neonatal lung pathology and morbidity is unknown. We have used a pregnant nonhuman primate model of group B Streptococcus infection to study the pathogenesis of intra-amniotic infection, bacterial invasion of the amniotic cavity and fetus, and microbial-host interactions. In this nonhuman primate model, we have studied the pathogenesis of group B Streptococcus strains with differing potential for virulence, which has resulted in a spectrum of intra-amniotic infection and fetal lung injury that affords the opportunity to study the inflammatory predictors of fetal lung pathology and injury. OBJECTIVE This study aimed to determine whether fetal lung injury is best predicted by placental histopathology or the cytokine response in amniotic fluid or maternal plasma. STUDY DESIGN Chronically catheterized pregnant monkeys (Macaca nemestrina, pigtail macaque) at 116 to 125 days gestation (term at 172 days) received a choriodecidual inoculation of saline (n=5), weakly hemolytic group B Streptococcus strain (n=5, low virulence), or hyperhemolytic group B Streptococcus strain (n=5, high virulence). Adverse pregnancy outcomes were defined as either preterm labor, microbial invasion of the amniotic cavity, or development of the fetal inflammatory response syndrome. Amniotic fluid and maternal and fetal plasma samples were collected after inoculation, and proinflammatory cytokines (tumor necrosis factor alpha, interleukin beta, interleukin 6, interleukin 8) were measured by a multiplex assay. Cesarean delivery was performed at the time of preterm labor or within 1 week of inoculation. Fetal necropsy was performed at the time of delivery. Placental pathology was scored in a blinded fashion by a pediatric pathologist, and fetal lung injury was determined by a semiquantitative score from histopathology evaluating inflammatory infiltrate, necrosis, tissue thickening, or collapse scored by a veterinary pathologist. RESULTS The principal findings in our study are as follows: (1) adverse pregnancy outcomes occurred more frequently in animals receiving hyperhemolytic group B Streptococcus (80% with preterm labor, 80% with fetal inflammatory response syndrome) than in animals receiving weakly hemolytic group B Streptococcus (40% with preterm labor, 20% with fetal inflammatory response syndrome) and in controls (0% preterm labor, 0% fetal inflammatory response syndrome); (2) despite differences in the rate of adverse pregnancy outcomes and fetal inflammatory response syndrome, fetal lung injury scores were similar between animals receiving the weakly hemolytic group B Streptococcus strains and animals receiving the hyperhemolytic group B Streptococcus strains; (3) fetal lung injury score was significantly correlated with peak amniotic fluid cytokines interleukin 6 and interleukin 8 but not tumor necrosis factor alpha or interleukin 1 beta; and (4) fetal lung scores were poorly correlated with maternal and fetal plasma cytokine levels and placental pathology. CONCLUSION Amniotic fluid interleukin 6 and interleukin 8 levels were superior predictors of fetal lung injury than placental histopathology or maternal plasma cytokines. This evidence supports a role for amniocentesis in the prediction of neonatal lung morbidity owing to intra-amniotic infection, which cannot be provided by cytokine analysis of maternal plasma or placental histopathology.
Collapse
Affiliation(s)
- Stephen A McCartney
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA
| | - Raj Kapur
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle Children's Hospital, Seattle, WA
| | - H Denny Liggitt
- Department of Comparative Medicine, University of Washington, Seattle, WA
| | - Audrey Baldessari
- Washington National Primate Research Center, University of Washington, Seattle, WA
| | - Michelle Coleman
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Austyn Orvis
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Jason Ogle
- Washington National Primate Research Center, University of Washington, Seattle, WA
| | - Ronit Katz
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA
| | - Lakshmi Rajagopal
- Department of Pediatrics, Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Kristina M Adams Waldorf
- Department of Obstetrics and Gynecology and Global Health, University of Washington, Seattle, WA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA; Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA; Sahlgrenska Academy, University of Gothenburg, Sweden.
| |
Collapse
|
35
|
Kumherová M, Veselá K, Kosová M, Mašata J, Horáčková Š, Šmidrkal J. Novel Potential Probiotic Lactobacilli for Prevention and Treatment of Vulvovaginal Infections. Probiotics Antimicrob Proteins 2021; 13:163-172. [PMID: 32583131 DOI: 10.1007/s12602-020-09675-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Lactobacilli in the vaginal tract are essential to protect against microbial infections. We therefore focused on isolating vaginal lactobacilli from pregnant women and testing their functional properties. Lactobacilli were isolated from 50 vaginal swabs and the purified isolates were identified by MALDI-TOF MS. Functional properties (antimicrobial activity, organic acids and hydrogen peroxide production, antibiotic susceptibility, auto-aggregation, and hydrophobicity) of selected isolates were tested. Lactobacilli (41 strains) were identified in 58% of swabs with a predominance of Lactobacillus crispatus (48%) followed by L. jensenii (21%), L. rhamnosus (14%), L. fermentum (10%), and L. gasseri (7%). The highest antibacterial activity was determined for L. fermentum and L. rhamnosus. Strong anti-Candida activity was observed for strains L. crispatus, L. fermentum, and L. rhamnosus. Strain L. jensenii 58C possessed the highest production of hydrogen peroxide (6.32 ± 0.60 mg/l). The best lactic acid producer was strain L. rhamnosus 72A (11.6 ± 0.2 g/l). All strains were resistant to fluconazole and metronidazole. The highest auto-aggregation was observed for strain L. crispatus 51A (98.8 ± 0.1% after 24 h). Strain L. rhamnosus 68A showed the highest hydrophobicity (69.1 ± 1.4%). Strains L. fermentum and L. rhamnosus showed high antibacterial activity and hydrophobicity, and strains L. crispatus possessed high auto-aggregation and anti-Candida activity. Thus, these strains alone or in a mix could be used for the preparation of probiotic products for treatment and prevention of vulvovaginal infections of pregnant and non-pregnant women.
Collapse
Affiliation(s)
- Monika Kumherová
- Department of Dairy, Fat and Cosmetics, University of Chemistry and Technology, Technická 5, 166 28, Prague 6, Czech Republic.
| | - Kristina Veselá
- Department of Dairy, Fat and Cosmetics, University of Chemistry and Technology, Technická 5, 166 28, Prague 6, Czech Republic
| | - Michaela Kosová
- Department of Dairy, Fat and Cosmetics, University of Chemistry and Technology, Technická 5, 166 28, Prague 6, Czech Republic
| | - Jaromír Mašata
- Department of Obstetrics and Gynecology, First Faculty of Medicine, Charles University and General University Hospital, Apolinářská 18, 128 51, Prague 2, Czech Republic
| | - Šárka Horáčková
- Department of Dairy, Fat and Cosmetics, University of Chemistry and Technology, Technická 5, 166 28, Prague 6, Czech Republic
| | - Jan Šmidrkal
- Department of Dairy, Fat and Cosmetics, University of Chemistry and Technology, Technická 5, 166 28, Prague 6, Czech Republic
| |
Collapse
|
36
|
RNA Sequencing Reveals Distinct Immune Responses in the Chorioamniotic Membranes of Women with Preterm Labor and Microbial or Sterile Intra-amniotic Inflammation. Infect Immun 2021; 89:IAI.00819-20. [PMID: 33558326 DOI: 10.1128/iai.00819-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/03/2021] [Indexed: 12/31/2022] Open
Abstract
Preterm labor precedes premature birth, the leading cause of neonatal morbidity and mortality worldwide. Preterm labor can occur in the context of either microbe-associated intra-amniotic inflammation (i.e., intra-amniotic infection) or intra-amniotic inflammation in the absence of detectable microorganisms (i.e., sterile intra-amniotic inflammation). Both intra-amniotic infection and sterile intra-amniotic inflammation trigger local immune responses that have deleterious effects on fetal life. Yet, the extent of such immune responses in the fetal tissues surrounding the amniotic cavity (i.e., the chorioamniotic membranes) is poorly understood. By using RNA sequencing (RNA seq) as a discovery approach, we found that there were significant transcriptomic differences involving host response to pathogens in the chorioamniotic membranes of women with intra-amniotic infection compared to those from women without inflammation. In addition, the sterile or microbial nature of intra-amniotic inflammation was associated with distinct transcriptomic profiles in the chorioamniotic membranes. Moreover, the immune response in the chorioamniotic membranes of women with sterile intra-amniotic inflammation was milder in nature than that induced by microbes and involved the upregulation of alarmins and inflammasome-related molecules. Lastly, the presence of maternal and fetal inflammatory responses in the placenta was associated with the upregulation of immune processes in the chorioamniotic membranes. Collectively, these findings provide insight into the immune responses against microbes or alarmins that take place in the fetal tissues surrounding the amniotic cavity, shedding light on the immunobiology of preterm labor and birth.
Collapse
|
37
|
Tong M, Hanna SE, Abrahams VM. Polymicrobial stimulation of human fetal membranes induce neutrophil activation and neutrophil extracellular trap release. J Reprod Immunol 2021; 145:103306. [PMID: 33725528 DOI: 10.1016/j.jri.2021.103306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/07/2021] [Accepted: 03/01/2021] [Indexed: 11/28/2022]
Abstract
Preterm birth is a major contributor to neonatal mortality and morbidity. While the causes of preterm birth remain incompletely understood, infection is a major risk factor, and chorioamnionitis is commonly observed. Chorioamnionitis is characterized by inflammation and neutrophil infiltration of the fetal membranes (FM). We recently reported that human FMs which had been exposed to low levels of bacterial lipopolysaccharide (LPS) recruit neutrophils and activate them, increasing their secretion of pro-inflammatory cytokines, degranulation of myeloperoxidase (MPO), and release of neutrophil extracellular traps (NETs). Herein, we demonstrate that conditioned media (CM) from viral dsRNA (Poly(I:C))-stimulated FMs also increased neutrophil migration, and induced the secretion of inflammatory IL-8 and the release of NETs. Furthermore, CM from FMs stimulated by a combination of bacterial LPS and Poly(I:C) augmented neutrophil NET release, compared to CM from FMs stimulated with either Poly(I:C) or LPS alone. NETs induced by FMs exposed to Poly(I:C), with or without LPS, were released and degraded quicker than those induced by resting or LPS-stimulated FM-CM. These findings indicate that FMs exposed to viral dsRNA promote neutrophil recruitment, activation and NET formation, similar to FMs exposed to bacterial LPS alone. However, in response to FM polymicrobial stimulation the levels and kinetics of NET release are augmented. This work builds upon our understanding of how infections at the maternal-fetal interface may affect neutrophil function.
Collapse
Affiliation(s)
- Mancy Tong
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Sarah E Hanna
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Vikki M Abrahams
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
38
|
Hyaluronidase Impairs Neutrophil Function and Promotes Group B Streptococcus Invasion and Preterm Labor in Nonhuman Primates. mBio 2021; 12:mBio.03115-20. [PMID: 33402537 PMCID: PMC8545101 DOI: 10.1128/mbio.03115-20] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Invasive bacterial infections during pregnancy are a major risk factor for preterm birth, stillbirth, and fetal injury. Group B streptococci (GBS) are Gram-positive bacteria that asymptomatically colonize the lower genital tract but infect the amniotic fluid and induce preterm birth or stillbirth. Experimental models that closely emulate human pregnancy are pivotal for the development of successful strategies to prevent these adverse pregnancy outcomes. Using a unique nonhuman primate model that mimics human pregnancy and informs temporal events surrounding amniotic cavity invasion and preterm labor, we show that the animals inoculated with hyaluronidase (HylB)-expressing GBS consistently exhibited microbial invasion into the amniotic cavity, fetal bacteremia, and preterm labor. Although delayed cytokine responses were observed at the maternal-fetal interface, increased prostaglandin and matrix metalloproteinase levels in these animals likely mediated preterm labor. HylB-proficient GBS dampened reactive oxygen species production and exhibited increased resistance to neutrophils compared to an isogenic mutant. Together, these findings demonstrate how a bacterial enzyme promotes GBS amniotic cavity invasion and preterm labor in a model that closely resembles human pregnancy.
Collapse
|
39
|
Gomez-Lopez N, Romero R, Varrey A, Leng Y, Miller D, Done B, Xu Y, Bhatti G, Motomura K, Gershater M, Pique-Regi R, Tarca AL. RNA Sequencing Reveals Diverse Functions of Amniotic Fluid Neutrophils and Monocytes/Macrophages in Intra-Amniotic Infection. J Innate Immun 2020; 13:63-82. [PMID: 33152737 DOI: 10.1159/000509718] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/25/2020] [Indexed: 12/17/2022] Open
Abstract
Intra-amniotic infection, the invasion of microbes into the amniotic cavity resulting in inflammation, is a clinical condition that can lead to adverse pregnancy outcomes for the mother and fetus as well as severe long-term neonatal morbidities. Despite much research focused on the consequences of intra-amniotic infection, there remains little knowledge about the innate immune cells that respond to invading microbes. We performed RNA-seq of sorted amniotic fluid neutrophils and monocytes/macrophages from women with intra-amniotic infection to determine the transcriptomic differences between these innate immune cells. Further, we sought to identify specific transcriptomic pathways that were significantly altered by the maternal or fetal origin of amniotic fluid neutrophils and monocytes/macrophages, the presence of a severe fetal inflammatory response, and pregnancy outcome (i.e., preterm or term delivery). We show that significant transcriptomic differences exist between amniotic fluid neutrophils and monocytes/macrophages from women with intra-amniotic infection, indicating the distinct roles these cells play. The transcriptome of amniotic fluid immune cells varies based on their maternal or fetal origin, and the significant transcriptomic differences between fetal and maternal monocytes/macrophages imply that those of fetal origin exhibit impaired functions. Notably, transcriptomic changes in amniotic fluid monocytes/macrophages suggest that these immune cells collaborate with neutrophils in the trafficking of fetal leukocytes throughout the umbilical cord (i.e., funisitis). Finally, amniotic fluid neutrophils and monocytes/macrophages from preterm deliveries display enhanced transcriptional activity compared to those from term deliveries, highlighting the protective role of these cells during this vulnerable period. Collectively, these findings demonstrate the underlying complexity of local innate immune responses in women with intra-amniotic infection and provide new insights into the functions of neutrophils and monocytes/macrophages in the amniotic cavity.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA, .,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA, .,Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA,
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA.,Detroit Medical Center, Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Florida International University, Miami, Florida, USA
| | - Aneesha Varrey
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yaozhu Leng
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Bogdan Done
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Kenichiro Motomura
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Meyer Gershater
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roger Pique-Regi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| |
Collapse
|
40
|
Singh N, Herbert B, Sooranna G, Das A, Sooranna SR, Yellon SM, Johnson MR. Distinct preterm labor phenotypes have unique inflammatory signatures and contraction associated protein profiles†. Biol Reprod 2020; 101:1031-1045. [PMID: 31411323 DOI: 10.1093/biolre/ioz144] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/05/2019] [Accepted: 06/29/2019] [Indexed: 01/02/2023] Open
Abstract
Preterm labor (PTL) is the predominant cause of childhood morbidity and mortality. It has several phenotypes, each with a distinct etiology often involving inflammation. Here, in samples of reproductive tissues obtained in early PTL from women with phenotypically defined PTL, we examined the presence and distribution of inflammation and its relationship with prolabor gene expression. In chorioamnionitis (CA-PTL), cytokine protein concentrations were increased across all tissues; in idiopathic (I-PTL), the inflammatory changes were limited to the choriodecidua; inflammation was not a feature of placental abruption (PA-PTL). CA-PTL was associated with activation of p65 in the myometrium and AP-1 in the choriodecidua, and PA-PTL with CREB in the choriodecidua. In the myometrium, PGHS-2 mRNA level was increased in CA- and I-PTL; in the amnion, PGHS-2 mRNA level was higher in PA- and I-PTL, while in CA-PTL, OT, OTR mRNA, and CX-43 expression were increased. In the choriodecidua, PGHS-2 mRNA level was unchanged, but in CA and I-PTL, OT mRNA level were increased and OTR was reduced. These data show that CA-PTL is associated with widespread inflammation and prolabor gene expression. In contrast, in I-PTL, inflammation is limited to the choriodecidua, with discrete increases in PGHS-2 in the amnion and OT in the choriodecidua. Inflammation is not a feature of PA-PTL, which is associated with increased OT and OTR in the amnion.
Collapse
Affiliation(s)
- Natasha Singh
- Chelsea and Westminster Hospital, London, United Kingdom.,Institute of Reproductive and Developmental Biology, London, United Kingdom
| | - Bronwen Herbert
- Institute of Reproductive and Developmental Biology, London, United Kingdom
| | - Gavin Sooranna
- Chelsea and Westminster Hospital, London, United Kingdom.,Institute of Reproductive and Developmental Biology, London, United Kingdom
| | - Anya Das
- Institute of Reproductive and Developmental Biology, London, United Kingdom
| | - Suren R Sooranna
- Chelsea and Westminster Hospital, London, United Kingdom.,Institute of Reproductive and Developmental Biology, London, United Kingdom
| | - Steven M Yellon
- Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Mark R Johnson
- Chelsea and Westminster Hospital, London, United Kingdom.,Institute of Reproductive and Developmental Biology, London, United Kingdom
| |
Collapse
|
41
|
Armistead B, Quach P, Snyder JM, Santana-Ufret V, Furuta A, Brokaw A, Rajagopal L. Hemolytic Membrane Vesicles of Group B Streptococcus Promote Infection. J Infect Dis 2020; 223:1488-1496. [PMID: 32861213 DOI: 10.1093/infdis/jiaa548] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/25/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Group B streptococci (GBS) are β-hemolytic, Gram-positive bacteria associated with fetal injury, preterm birth, spontaneous abortion, and neonatal infections. A key factor promoting GBS virulence is the β-hemolysin/cytolysin, a pigmented ornithine rhamnolipid (also known as granadaene) associated with the bacterial surface. METHODS A previous study indicated that GBS produce small structures known as membrane vesicles (MVs), which contain virulence-associated proteins. In this study, we show that GBS MVs are pigmented and hemolytic, indicating that granadaene is functionally active in MVs. RESULTS In addition, MVs from hyperhemolytic GBS induced greater cell death of neutrophils, T cells, and B cells compared with MVs from isogenic nonhemolytic GBS, implicating MVs as a potential mechanism for granadaene-mediated virulence. Finally, hemolytic MVs reduced oxidative killing of GBS and aggravated morbidity and mortality of neonatal mice infected with GBS. CONCLUSIONS These studies, taken together, reveal a novel mechanism by which GBS deploy a crucial virulence factor to promote bacterial dissemination and pathogenesis.
Collapse
Affiliation(s)
- Blair Armistead
- Department of Global Health, University of Washington, Seattle, Washington, USA.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Phoenicia Quach
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Jessica M Snyder
- Department of Comparative Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Verónica Santana-Ufret
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Anna Furuta
- Department of Global Health, University of Washington, Seattle, Washington, USA.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Alyssa Brokaw
- Department of Global Health, University of Washington, Seattle, Washington, USA.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Lakshmi Rajagopal
- Department of Global Health, University of Washington, Seattle, Washington, USA.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA.,Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
42
|
Harris SM, Boldenow E, Domino SE, Loch-Caruso R. Toxicant Disruption of Immune Defenses: Potential Implications for Fetal Membranes and Pregnancy. Front Physiol 2020; 11:565. [PMID: 32547423 PMCID: PMC7272693 DOI: 10.3389/fphys.2020.00565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/07/2020] [Indexed: 11/22/2022] Open
Abstract
In addition to providing a physical compartment for gestation, the fetal membranes (FM) are an active immunological barrier that provides defense against pathogenic microorganisms that ascend the gravid reproductive tract. Pathogenic infection of the gestational tissues (FM and placenta) is a leading known cause of preterm birth (PTB). Some environmental toxicants decrease the capacity for organisms to mount an immune defense against pathogens. For example, the immunosuppressive effects of the widespread environmental contaminant trichloroethylene (TCE) are documented for lung infection with Streptococcus zooepidemicus. Group B Streptococcus (GBS; Streptococcus agalactiae) is a bacterial pathogen that is frequently found in the female reproductive tract and can colonize the FM in pregnant women. Work in our laboratory has demonstrated that a bioactive TCE metabolite, S-(1, 2-dichlorovinyl)-L-cysteine (DCVC), potently inhibits innate immune responses to GBS in human FM in culture. Despite these provocative findings, little is known about how DCVC and other toxicants modify the risk for pathogenic infection of FM. Infection of the gestational tissues (FM and placenta) is a leading known cause of PTB, therefore toxicant compromise of FM ability to fight off infectious microorganisms could significantly contribute to PTB risk. This Perspective provides the current status of understanding of toxicant-pathogen interactions in FM, highlighting knowledge gaps, challenges, and opportunities for research that can advance protections for maternal and fetal health.
Collapse
Affiliation(s)
- Sean M. Harris
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Erica Boldenow
- Department of Biology, Calvin College, Grand Rapids, MI, United States
| | - Steven E. Domino
- Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Rita Loch-Caruso
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
43
|
Coleman M, Orvis A, Wu TY, Dacanay M, Merillat S, Ogle J, Baldessari A, Kretzer NM, Munson J, Boros-Rausch AJ, Shynlova O, Lye S, Rajagopal L, Adams Waldorf KM. A Broad Spectrum Chemokine Inhibitor Prevents Preterm Labor but Not Microbial Invasion of the Amniotic Cavity or Neonatal Morbidity in a Non-human Primate Model. Front Immunol 2020; 11:770. [PMID: 32425945 PMCID: PMC7203489 DOI: 10.3389/fimmu.2020.00770] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022] Open
Abstract
Leukocyte activation within the chorioamniotic membranes is strongly associated with inflammation and preterm labor (PTL). We hypothesized that prophylaxis with a broad-spectrum chemokine inhibitor (BSCI) would downregulate the inflammatory microenvironment induced by Group B Streptococcus (GBS, Streptococcus agalactiae) to suppress PTL and microbial invasion of the amniotic cavity (MIAC). To correlate BSCI administration with PTL and MIAC, we used a unique chronically catheterized non-human primate model of Group B Streptococcus (GBS)-induced PTL. In the early third trimester (128–138 days gestation; ~29–32 weeks human pregnancy), animals received choriodecidual inoculations of either: (1) saline (N = 6), (2) GBS, 1–5 × 108 colony forming units (CFU)/ml; N = 5), or (3) pre-treatment and daily infusions of a BSCI (10 mg/kg intravenous and intra-amniotic) with GBS (1–5 × 108 CFU/ml; N = 4). We measured amniotic cavity pressure (uterine contraction strength) and sampled amniotic fluid (AF) and maternal blood serially and cord blood at delivery. Cesarean section was performed 3 days post-inoculation or earlier for PTL. Data analysis used Fisher's exact test, Wilcoxon rank sum and one-way ANOVA with Bonferroni correction. Saline inoculation did not induce PTL or infectious sequelae. In contrast, GBS inoculation typically induced PTL (4/5, 80%), MIAC and fetal bacteremia (3/5; 60%). Remarkably, PTL did not occur in the BSCI+GBS group (0/4, 0%; p = 0.02 vs. GBS), despite MIAC and fetal bacteremia in all cases (4/4; 100%). Compared to the GBS group, BSCI prophylaxis was associated with significantly lower cytokine levels including lower IL-8 in amniotic fluid (p = 0.03), TNF-α in fetal plasma (p < 0.05), IFN-α and IL-7 in the fetal lung (p = 0.02) and IL-18, IL-2, and IL-7 in the fetal brain (p = 0.03). Neutrophilic chorioamnionitis was common in the BSCI and GBS groups, but was more severe in the BSCI+GBS group with greater myeloperoxidase staining (granulocyte marker) in the amnion and chorion (p < 0.05 vs. GBS). Collectively, these observations indicate that blocking the chemokine response to infection powerfully suppressed uterine contractility, PTL and the cytokine response, but did not prevent MIAC and fetal pneumonia. Development of PTL immunotherapies should occur in tandem with evaluation for AF microbes and consideration for antibiotic therapy.
Collapse
Affiliation(s)
- Michelle Coleman
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Austyn Orvis
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Tsung-Yen Wu
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
| | - Matthew Dacanay
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
| | - Sean Merillat
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Jason Ogle
- Washington National Primate Center, University of Washington, Seattle, WA, United States
| | - Audrey Baldessari
- Washington National Primate Center, University of Washington, Seattle, WA, United States
| | - Nicole M Kretzer
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
| | - Jeff Munson
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States
| | | | - Oksana Shynlova
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics & Gynaecology, University of Toronto, Toronto, ON, Canada
| | - Stephen Lye
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics & Gynaecology, University of Toronto, Toronto, ON, Canada
| | - Lakshmi Rajagopal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Kristina M Adams Waldorf
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
44
|
Carreras-Abad C, Ramkhelawon L, Heath PT, Le Doare K. A Vaccine Against Group B Streptococcus: Recent Advances. Infect Drug Resist 2020; 13:1263-1272. [PMID: 32425562 PMCID: PMC7196769 DOI: 10.2147/idr.s203454] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 03/10/2020] [Indexed: 01/12/2023] Open
Abstract
Group B streptococcus (GBS) causes a high burden of neonatal and infant disease globally. Implementing a vaccine for pregnant women is a promising strategy to prevent neonatal and infant GBS disease and has been identified as a priority by the World Health Organisation (WHO). GBS serotype-specific polysaccharide – protein conjugate vaccines are at advanced stages of development, but a large number of participants would be required to undertake Phase III clinical efficacy trials. Efforts are therefore currently focused on establishing serocorrelates of protection in natural immunity studies as an alternative pathway for licensure of a GBS vaccine, followed by Phase IV studies to evaluate safety and effectiveness. Protein vaccines are in earlier stages of development but are highly promising as they might confer protection irrespective of serotype. Further epidemiological, immunological and health economic studies are required to enable the vaccine to reach its target population as soon as possible.
Collapse
Affiliation(s)
- Clara Carreras-Abad
- Paediatric Infectious Diseases Research Group and Vaccine Institute, Institute for Infection and Immunity, St George's, University of London, London, UK.,Department of Paediatrics, Obstetrics and Gynecology and Preventive Medicine and Public Health, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laxmee Ramkhelawon
- Paediatric Infectious Diseases Research Group and Vaccine Institute, Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Paul T Heath
- Paediatric Infectious Diseases Research Group and Vaccine Institute, Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Kirsty Le Doare
- Paediatric Infectious Diseases Research Group and Vaccine Institute, Institute for Infection and Immunity, St George's, University of London, London, UK.,Pathogen Immunity Group, Public Health England, Porton Down,UK.,Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| |
Collapse
|
45
|
Cappelletti M, Presicce P, Kallapur SG. Immunobiology of Acute Chorioamnionitis. Front Immunol 2020; 11:649. [PMID: 32373122 PMCID: PMC7177011 DOI: 10.3389/fimmu.2020.00649] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 03/23/2020] [Indexed: 12/19/2022] Open
Abstract
Acute chorioamnionitis is characterized by neutrophilic infiltration and inflammation at the maternal fetal interface. It is a relatively common complication of pregnancy and can have devastating consequences including preterm labor, maternal infections, fetal infection/inflammation, fetal lung, brain, and gastrointestinal tract injury. In this review, we will discuss current understanding of the pathogenesis, immunobiology, and mechanisms of this condition. Most commonly, acute chorioamnionitis is a result of ascending infection with relatively low-virulence organisms such as the Ureaplasma species. Furthermore, recent vaginal microbiome studies suggest that there is a link between vaginal dysbiosis, vaginal inflammation, and ascending infection. Although less common, microorganisms invading the maternal-fetal interface via hematogenous route (e.g., Zika virus, Cytomegalovirus, and Listeria) can cause placental villitis and severe fetal inflammation and injury. We will provide an overview of the knowledge gleaned from different animal models of acute chorioamnionitis and the role of different immune cells in different maternal-fetal compartments. Lastly, we will discuss how infectious agents can break the maternal tolerance of fetal allograft during pregnancy and highlight the novel future therapeutic approaches.
Collapse
Affiliation(s)
- Monica Cappelletti
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Suhas G Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
46
|
Presicce P, Cappelletti M, Senthamaraikannan P, Ma F, Morselli M, Jackson CM, Mukherjee S, Miller LA, Pellegrini M, Jobe AH, Chougnet CA, Kallapur SG. TNF-Signaling Modulates Neutrophil-Mediated Immunity at the Feto-Maternal Interface During LPS-Induced Intrauterine Inflammation. Front Immunol 2020; 11:558. [PMID: 32308656 PMCID: PMC7145904 DOI: 10.3389/fimmu.2020.00558] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/11/2020] [Indexed: 01/10/2023] Open
Abstract
Accumulation of activated neutrophils at the feto-maternal interface is a defining hallmark of intrauterine inflammation (IUI) that might trigger an excessive immune response during pregnancy. Mechanisms responsible of this massive neutrophil recruitment are poorly investigated. We have previously showed that intraamniotic injection of LPS in rhesus macaques induced a neutrophil predominant inflammatory response similar to that seen in human IUI. Here, we demonstrate that anti-TNF antibody (Adalimumab) inhibited ~80% of genes induced by LPS involved in inflammatory signaling and innate immunity in chorio-decidua neutrophils. Consistent with the gene expression data, TNF-blockade decreased LPS-induced neutrophil accumulation and activation at the feto-maternal interface. We also observed a reduction in IL-6 and other pro-inflammatory cytokines but not prostaglandins concentrations in the amniotic fluid. Moreover, TNF-blockade decreased mRNA expression of inflammatory cytokines in the chorio-decidua but not in the uterus, suggesting that inhibition of TNF-signaling decreased the inflammation in a tissue-specific manner within the uterine compartment. Taken together, our results demonstrate a predominant role for TNF-signaling in modulating the neutrophilic infiltration at the feto-maternal interface during IUI and suggest that blockade of TNF-signaling could be considered as a therapeutic approach for IUI, the major leading cause of preterm birth.
Collapse
Affiliation(s)
- Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Monica Cappelletti
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Paranthaman Senthamaraikannan
- Division of Neonatology/Pulmonary Biology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Feiyang Ma
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Institute for Quantitative and Computational Biosciences-Collaboratory, University of California, Los Angeles, Los Angeles, CA, United States
| | - Courtney M Jackson
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Shibabrata Mukherjee
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Lisa A Miller
- California National Primate Research Center, University of California, Davis, Davis, CA, United States.,Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Institute for Quantitative and Computational Biosciences-Collaboratory, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alan H Jobe
- Division of Neonatology/Pulmonary Biology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Claire A Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Suhas G Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
47
|
Lipid analogs reveal features critical for hemolysis and diminish granadaene mediated Group B Streptococcus infection. Nat Commun 2020; 11:1502. [PMID: 32198389 PMCID: PMC7083881 DOI: 10.1038/s41467-020-15282-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 02/27/2020] [Indexed: 12/17/2022] Open
Abstract
Although certain microbial lipids are toxins, the structural features important for cytotoxicity remain unknown. Increased functional understanding is essential for developing therapeutics against toxic microbial lipids. Group B Streptococci (GBS) are bacteria associated with preterm births, stillbirths, and severe infections in neonates and adults. GBS produce a pigmented, cytotoxic lipid, known as granadaene. Despite its importance to all manifestations of GBS disease, studies towards understanding granadaene’s toxic activity are hindered by its instability and insolubility in purified form. Here, we report the synthesis and screening of lipid derivatives inspired by granadaene, which reveal features central to toxin function, namely the polyene chain length. Furthermore, we show that vaccination with a non-toxic synthetic analog confers the production of antibodies that inhibit granadaene-mediated hemolysis ex vivo and diminish GBS infection in vivo. This work provides unique structural and functional insight into granadaene and a strategy to mitigate GBS infection, which will be relevant to other toxic lipids encoded by human pathogens. Granadaene, produced by Group B Streptococcus (GBS), is a long polyene lipid involved in cellular toxicity and hemolytic activity. Here, the authors synthesize and characterize granadaene-like compounds and show that a non-toxic analog diminishes GBS infection in mice when incorporated into a vaccine formulation.
Collapse
|
48
|
Armistead B, Whidbey C, Iyer LM, Herrero-Foncubierta P, Quach P, Haidour A, Aravind L, Cuerva JM, Jaspan HB, Rajagopal L. The cyl Genes Reveal the Biosynthetic and Evolutionary Origins of the Group B Streptococcus Hemolytic Lipid, Granadaene. Front Microbiol 2020; 10:3123. [PMID: 32038561 PMCID: PMC6985545 DOI: 10.3389/fmicb.2019.03123] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 12/24/2019] [Indexed: 01/31/2023] Open
Abstract
Group B Streptococcus (GBS) is a β-hemolytic, Gram-positive bacterium that commonly colonizes the female lower genital tract and is associated with fetal injury, preterm birth, spontaneous abortion, and neonatal infections. A major factor promoting GBS virulence is the β-hemolysin/cytolysin, which is cytotoxic to several host cells. We recently showed that the ornithine rhamnolipid pigment, Granadaene, produced by the gene products of the cyl operon, is hemolytic. Here, we demonstrate that heterologous expression of the GBS cyl operon conferred hemolysis, pigmentation, and cytoxicity to Lactococcus lactis, a model non-hemolytic Gram-positive bacterium. Similarly, pigment purified from L. lactis is hemolytic, cytolytic, and identical in structure to Granadaene extracted from GBS, indicating the cyl operon is sufficient for Granadaene production in a heterologous host. Using a systematic survey of phyletic patterns and contextual associations of the cyl genes, we identify homologs of the cyl operon in physiologically diverse Gram-positive bacteria and propose undescribed functions of cyl gene products. Together, these findings bring greater understanding to the biosynthesis and evolutionary foundations of a key GBS virulence factor and suggest that such potentially toxic lipids may be encoded by other bacteria.
Collapse
Affiliation(s)
- Blair Armistead
- Department of Global Health, University of Washington, Seattle, WA, United States.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Christopher Whidbey
- Department of Global Health, University of Washington, Seattle, WA, United States.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Lakshminarayan M Iyer
- Computational Biology Branch, National Center for Biotechnology Information, National Institutes of Health, Bethesda, MD, United States
| | | | - Phoenicia Quach
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Ali Haidour
- Department of Organic Chemistry, University of Granada, Granada, Spain
| | - L Aravind
- Computational Biology Branch, National Center for Biotechnology Information, National Institutes of Health, Bethesda, MD, United States
| | | | - Heather B Jaspan
- Department of Global Health, University of Washington, Seattle, WA, United States.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States.,Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Lakshmi Rajagopal
- Department of Global Health, University of Washington, Seattle, WA, United States.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
49
|
Abstract
Preterm birth is a serious global health problem that affects 5-18% of pregnancies worldwide. In addition to being the major cause of neonatal mortality and morbidity, preterm birth is associated with short term and long term complications in the offspring. Despite this, the causes and pathogenesis of preterm birth remain unclear. Neutrophils are innate immune cells that infiltrate the maternal-fetal interface during normal parturition and their accumulation is dramatically increased during preterm birth, especially in the presence of an infection. Indeed, a defining feature of chorioamnionitis (inflammation of the chorioamnionic fetal membranes) that is associated with more than 40% of preterm births, is neutrophil accumulation. While these cells may play an important role during normal term parturition as well as preterm birth, their functions at the maternal-fetal interface are unclear. This review will provide a broad overview of the relevant studies to enable a better understanding of the roles of neutrophils during normal parturition and preterm birth.
Collapse
|
50
|
Romero R, Gomez-Lopez N, Winters AD, Jung E, Shaman M, Bieda J, Panaitescu B, Pacora P, Erez O, Greenberg JM, Ahmad MM, Hsu CD, Theis KR. Evidence that intra-amniotic infections are often the result of an ascending invasion - a molecular microbiological study. J Perinat Med 2019; 47:915-931. [PMID: 31693497 PMCID: PMC7147941 DOI: 10.1515/jpm-2019-0297] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 08/18/2019] [Indexed: 12/15/2022]
Abstract
Background Microbial invasion of the amniotic cavity resulting in intra-amniotic infection is associated with obstetrical complications such as preterm labor with intact or ruptured membranes, cervical insufficiency, as well as clinical and histological chorioamnionitis. The most widely accepted pathway for intra-amniotic infection is the ascension of microorganisms from the lower genital tract. However, hematogenous dissemination of microorganisms from the oral cavity or intestine, retrograde seeding from the peritoneal cavity through the fallopian tubes, and introduction through invasive medical procedures have also been suggested as potential pathways for intra-amniotic infection. The primary reason that an ascending pathway is viewed as most common is that the microorganisms most often detected in the amniotic fluid are those that are typical inhabitants of the vagina. However, thus far, no studies have shown that microorganisms in the amniotic cavity are simultaneously present in the vagina of the woman from which they were isolated. The objective of the study was to determine the frequency with which microorganisms isolated from women with intra-amniotic infection are also present in the lower genital tract. Methods This was a cross-sectional study of women with intra-amniotic infection with intact membranes. Intra-amniotic infection was defined as a positive culture and elevated concentrations of interleukin-6 (IL-6) (>2.6 ng/mL) in amniotic fluid and/or acute histologic chorioamnionitis and funisitis. Microorganisms isolated from bacterial cultures of amniotic fluid were taxonomically identified through matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF) and 16S ribosomal RNA (rRNA) gene sequencing. Vaginal swabs were obtained at the time of amniocentesis for the identification of microorganisms in the lower genital tract. The overall bacterial profiles of amniotic fluids and vaginal swabs were characterized through 16S rRNA gene sequencing. The bacterial profiles of vaginal swabs were interrogated for the presence of bacteria cultured from amniotic fluid and for the presence of prominent (>1% average relative abundance) operational taxonomic units (OTUs) within the overall 16S rRNA gene bacterial profiles of amniotic fluid. Results (1) A total of 75% (6/8) of women had bacteria cultured from their amniotic fluid that are typical residents of the vaginal ecosystem. (2) A total of 62.5% (5/8) of women with bacteria cultured from their amniotic fluid also had these bacteria present in their vagina. (3) The microorganisms cultured from amniotic fluid and also detected in the vagina were Ureaplasma urealyticum, Escherichia coli, and Streptococcus agalactiae. (4) 16S rRNA gene sequencing revealed that the amniotic fluid of women with intra-amniotic infection had bacterial profiles dominated by Sneathia, Ureaplasma, Prevotella, Lactobacillus, Escherichia, Gardnerella, Peptostreptococcus, Peptoniphilus, and Streptococcus, many of which had not been cultured from the amniotic fluid samples. (5) Seventy percent (7/10) of the prominent (>1% average relative abundance) OTUs found in amniotic fluid were also prominent in the vagina. Conclusion The majority of women with intra-amniotic infection had bacteria cultured from their amniotic fluid that were typical vaginal commensals, and these bacteria were detected within the vagina at the time of amniocentesis. Molecular microbiological interrogation of amniotic fluid from women with intra-amniotic infection revealed that the bacterial profiles of amniotic fluid were largely consistent with those of the vagina. These findings indicate that ascension from the lower genital tract is the primary pathway for intra-amniotic infection.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
- Detroit Medical Center, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Florida International University, Miami, FL, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
- Perinatal Research Initiative in Maternal, Perinatal and Child Health, Wayne State University School of Medicine, Detroit, MI, USA
| | - Andrew D. Winters
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Majid Shaman
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Janine Bieda
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bogdan Panaitescu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Percy Pacora
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Maternity Department “D,” Division of Obstetrics and Gynecology, Soroka University Medical Center School of Medicine, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Jonathan M. Greenberg
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Madison M. Ahmad
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Chaur-Dong Hsu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Perinatal Research Initiative in Maternal, Perinatal and Child Health, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kevin R. Theis
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
- Perinatal Research Initiative in Maternal, Perinatal and Child Health, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|