1
|
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
|
2
|
Xia Z, Xiang H, Shi YM. Bacterial Secondary Metabolites Embedded in Producer Cell Membranes and Antibiotics Targeting Their Biosynthesis. ChemMedChem 2024; 19:e202400469. [PMID: 39287217 DOI: 10.1002/cmdc.202400469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/19/2024]
Abstract
The bacterial cell membrane primarily houses lipids, carbohydrates, and proteins forming a barrier and interface that maintains cellular integrity, supports homeostasis, and senses environmental changes. Compared to lipid components and excreted secondary metabolites, compounds embedded in the producer cell membrane are often overlooked due to their low abundance and niche-specific functions. The accumulation of findings has led to an increased appreciation of their crucial roles in bacterial cell biochemistry, physiology, and ecology, as well as their impact on mutualistic and pathogenic bacteria-eukaryote interactions. This review highlights the structures, biosynthesis, regulation, and ecological functions of membrane-embedded secondary metabolites. It also discusses antibiotics that target their biosynthetic pathways, aiming to inspire the development of antibiotics specific to pathogenic bacteria without harming human cells.
Collapse
Affiliation(s)
- Zhao Xia
- Key Laboratory of Quantitative Synthetic Biology, Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hao Xiang
- Key Laboratory of Quantitative Synthetic Biology, Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi-Ming Shi
- Key Laboratory of Quantitative Synthetic Biology, Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
3
|
Suárez Vázquez TA, López López N, Salinas Carmona MC. MASTer cell: chief immune modulator and inductor of antimicrobial immune response. Front Immunol 2024; 15:1360296. [PMID: 38638437 PMCID: PMC11024470 DOI: 10.3389/fimmu.2024.1360296] [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: 12/22/2023] [Accepted: 02/26/2024] [Indexed: 04/20/2024] Open
Abstract
Mast cells have long been recognized for their involvement in allergic pathology through the immunoglobulin E (IgE)-mediated degranulation mechanism. However, there is growing evidence of other "non-canonical" degranulation mechanisms activated by certain pathogen recognition receptors. Mast cells release several mediators, including histamine, cytokines, chemokines, prostaglandins, and leukotrienes, to initiate and enhance inflammation. The chemical nature of activating stimuli influences receptors, triggering mechanisms for the secretion of formed and new synthesized mediators. Mast cells have more than 30 known surface receptors that activate different pathways for direct and indirect activation by microbes. Different bacterial strains stimulate mast cells through various ligands, initiating the innate immune response, which aids in clearing the bacterial burden. Mast cell interactions with adaptative immune cells also play a crucial role in infections. Recent publications revealed another "non-canonical" degranulation mechanism present in tryptase and chymase mast cells in humans and connective tissue mast cells in mice, occurring through the activation of the Mas-related G protein-coupled receptor (MRGPRX2/b2). This receptor represents a new therapeutic target alongside antibiotic therapy. There is an urgent need to reconsider and redefine the biological role of these MASTer cells of innate immunity, extending beyond their involvement in allergic pathology.
Collapse
Affiliation(s)
| | | | - Mario César Salinas Carmona
- Department of Immunology, School of Medicine and Dr. Jose Eleuterio Gonzalez University Hospital, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| |
Collapse
|
4
|
Zhao H, Sun J, Yang Y. Research Progress of Photodynamic Therapy in Wound Healing: A Literature Review. J Burn Care Res 2023; 44:1327-1333. [PMID: 37747820 DOI: 10.1093/jbcr/irad146] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Indexed: 09/27/2023]
Abstract
Light is an efficient technique that has a significant influence on contemporary medicine. Photodynamic therapy (PDT), which involves the combined action of photosensitizers (PSs), oxygen, and light, has emerged as a therapeutically promising method for treating a broad variety of solid tumors and infectious diseases. Photodynamic therapy is minimally invasive, has few side effects, lightens scars, and reduces tissue loss while preserving organ structure and function. In particular, PDT has a high healing potential for wounds (PDT stimulates wound healing by enhancing re-epithelialization, promoting angiogenesis as well as modulating skin homeostasis). Wound healing involves interactions between many different processes, including coagulation, inflammation, angiogenesis, cellular migration, and proliferation. Poor wound healing with diabetes or extensive burns remains a difficult challenge. This review emphasizes PDT as a potential research field and summarizes PDT's role in wound healing, including normal wounds, chronic wounds, and aging wounds.
Collapse
Affiliation(s)
- Hongqing Zhao
- Nanbu County People's Hospital, Nanchong City, Sichuan Province 637300, China
- Department of Dermatology, Fourth Medical Center of Chinese PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing 100048, China
- Jinzhou Medical University, No. 40 Songpo Road, Taihe District, Jinzhou City, Liaoning Province 121001, China
| | - Jiachen Sun
- Department of Dermatology, Fourth Medical Center of Chinese PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing 100048, China
| | - Yuguang Yang
- Department of Dermatology, Fourth Medical Center of Chinese PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing 100048, China
| |
Collapse
|
5
|
Miralda I, Samanas NB, Seo AJ, Foronda JS, Sachen J, Hui Y, Morrison SD, Oskeritzian CA, Piliponsky AM. Siglec-9 is an inhibitory receptor on human mast cells in vitro. J Allergy Clin Immunol 2023; 152:711-724.e14. [PMID: 37100120 PMCID: PMC10524464 DOI: 10.1016/j.jaci.2023.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/04/2023] [Accepted: 04/13/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND Mast cell activation is critical for the development of allergic diseases. Ligation of sialic acid-binding immunoglobin-like lectins (Siglecs), such as Siglec-6, -7, and -8 as well as CD33, have been shown to inhibit mast cell activation. Recent studies showed that human mast cells express Siglec-9, an inhibitory receptor also expressed by neutrophils, monocytes, macrophages, and dendritic cells. OBJECTIVE We aimed to characterize Siglec-9 expression and function in human mast cells in vitro. METHODS We assessed the expression of Siglec-9 and Siglec-9 ligands on human mast cell lines and human primary mast cells by real-time quantitative PCR, flow cytometry, and confocal microscopy. We used a clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) gene editing approach to disrupt the SIGLEC9 gene. We evaluated Siglec-9 inhibitory activity on mast cell function by using native Siglec-9 ligands, glycophorin A (GlycA), and high-molecular-weight hyaluronic acid, a monoclonal antibody against Siglec-9, and coengagement of Siglec-9 with the high-affinity receptor for IgE (FcεRI). RESULTS Human mast cells express Siglec-9 and Siglec-9 ligands. SIGLEC9 gene disruption resulted in increased expression of activation markers at baseline and increased responsiveness to IgE-dependent and IgE-independent stimulation. Pretreatment with GlycA or high-molecular-weight hyaluronic acid followed by IgE-dependent or -independent stimulation had an inhibitory effect on mast cell degranulation. Coengagement of Siglec-9 with FcεRI in human mast cells resulted in reduced degranulation, arachidonic acid production, and chemokine release. CONCLUSIONS Siglec-9 and its ligands play an important role in limiting human mast cell activation in vitro.
Collapse
Affiliation(s)
- Irina Miralda
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash
| | - Nyssa B Samanas
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash
| | - Albert J Seo
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash
| | - Jake S Foronda
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash
| | - Josie Sachen
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash
| | - Yvonne Hui
- University of South Carolina School of Medicine, Columbia, SC
| | - Shane D Morrison
- Department of Surgery, Division of Plastic Surgery, Seattle Children's Hospital, Seattle, Wash
| | | | - Adrian M Piliponsky
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Wash; Department of Pediatrics, University of Washington School of Medicine, Seattle, Wash; Department of Pathology, University of Washington School of Medicine, Seattle, Wash; Department of Global Health, University of Washington School of Medicine, Seattle, Wash.
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Banafea GH, Bakhashab S, Alshaibi HF, Natesan Pushparaj P, Rasool M. The role of human mast cells in allergy and asthma. Bioengineered 2022; 13:7049-7064. [PMID: 35266441 PMCID: PMC9208518 DOI: 10.1080/21655979.2022.2044278] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Mast cells are tissue-inhabiting cells that play an important role in inflammatory diseases of the airway tract. Mast cells arise in the bone marrow as progenitor cells and complete their differentiation in tissues exposed to the external environment, such as the skin and respiratory tract, and are among the first to respond to bacterial and parasitic infections. Mast cells express a variety of receptors that enable them to respond to a wide range of stimulants, including the high-affinity FcεRI receptor. Upon initial contact with an antigen, mast cells are sensitized with IgE to recognize the allergen upon further contact. FcεRI-activated mast cells are known to release histamine and proteases that contribute to asthma symptoms. They release a variety of cytokines and lipid mediators that contribute to immune cell accumulation and tissue remodeling in asthma. Mast cell mediators trigger inflammation and also have a protective effect. This review aims to update the existing knowledge on the mediators released by human FcεRI-activated mast cells, and to unravel their pathological and protective roles in asthma and allergy. In addition, we highlight other diseases that arise from mast cell dysfunction, the therapeutic approaches used to address them, and fill the gaps in our current knowledge. Mast cell mediators not only trigger inflammation but may also have a protective effect. Given the differences between human and animal mast cells, this review focuses on the mediators released by human FcεRI-activated mast cells and the role they play in asthma and allergy.
Collapse
Affiliation(s)
- Ghalya H Banafea
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sherin Bakhashab
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Huda F Alshaibi
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Peter Natesan Pushparaj
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmood Rasool
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
9
|
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
|
10
|
Functional Insights into the High-Molecular-Mass Penicillin-Binding Proteins of Streptococcus agalactiae Revealed by Gene Deletion and Transposon Mutagenesis Analysis. J Bacteriol 2021; 203:e0023421. [PMID: 34124943 DOI: 10.1128/jb.00234-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
High-molecular-mass penicillin-binding proteins (PBPs) are enzymes that catalyze the biosynthesis of bacterial cell wall peptidoglycan. The Gram-positive bacterial pathogen Streptococcus agalactiae (group B streptococcus [GBS]) produces five high-molecular-mass PBPs, namely, PBP1A, PBP1B, PBP2A, PBP2B, and PBP2X. Among these, only PBP2X is essential for cell viability, whereas the other four PBPs are individually dispensable. The biological function of the four nonessential PBPs is poorly characterized in GBS. We deleted the pbp1a, pbp1b, pbp2a, and pbp2b genes individually from a genetically well-characterized serotype V GBS strain and studied the phenotypes of the four isogenic mutant strains. Compared to the wild-type parental strain, (i) none of the pbp isogenic mutant strains had a significant growth defect in Todd-Hewitt broth supplemented with 0.2% yeast extract (THY) rich medium, (ii) isogenic mutant Δpbp1a and Δpbp1b strains had significantly increased susceptibility to penicillin and ampicillin, and (iii) isogenic mutant Δpbp1a and Δpbp2b strains had significantly longer chain lengths. Using saturated transposon mutagenesis and transposon insertion site sequencing, we determined the genes essential for the viability of the wild-type GBS strain and each of the four isogenic pbp deletion mutant strains in THY rich medium. The pbp1a gene is essential for cell viability in the pbp2b deletion background. Reciprocally, pbp2b is essential in the pbp1a deletion background. Moreover, the gene encoding RodA, a peptidoglycan polymerase that works in conjunction with PBP2B, is also essential in the pbp1a deletion background. Together, our results suggest functional overlap between PBP1A and the PBP2B-RodA complex in GBS cell wall peptidoglycan biosynthesis. IMPORTANCE High-molecular-mass penicillin-binding proteins (HMM PBPs) are enzymes required for bacterial cell wall biosynthesis. Bacterial pathogen group B streptococcus (GBS) produces five distinct HMM PBPs. The biological functions of these proteins are not well characterized in GBS. In this study, we performed a comprehensive deletion analysis of genes encoding HMM PBPs in GBS. We found that deleting certain PBP-encoding genes altered bacterial susceptibility to beta-lactam antibiotics, cell morphology, and the essentiality of other enzymes involved in cell wall peptidoglycan synthesis. The results of our study shed new light on the biological functions of PBPs in GBS.
Collapse
|
11
|
Chin HS, Fu NY. Physiological Functions of Mcl-1: Insights From Genetic Mouse Models. Front Cell Dev Biol 2021; 9:704547. [PMID: 34336857 PMCID: PMC8322662 DOI: 10.3389/fcell.2021.704547] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/14/2021] [Indexed: 01/27/2023] Open
Abstract
The ability to regulate the survival and death of a cell is paramount throughout the lifespan of a multicellular organism. Apoptosis, a main physiological form of programmed cell death, is regulated by the Bcl-2 family proteins that are either pro-apoptotic or pro-survival. The in vivo functions of distinct Bcl-2 family members are largely unmasked by genetically engineered murine models. Mcl-1 is one of the two Bcl-2 like pro-survival genes whose germline deletion causes embryonic lethality in mice. Its requisite for the survival of a broad range of cell types has been further unraveled by using conditional and inducible deletion murine model systems in different tissues or cell lineages and at distinct developmental stages. Moreover, genetic mouse cancer models have also demonstrated that Mcl-1 is essential for the survival of multiple tumor types. The MCL-1 locus is commonly amplified across various cancer types in humans. Small molecule inhibitors with high affinity and specificity to human MCL-1 have been developed and explored for the treatment of certain cancers. To facilitate the pre-clinical studies of MCL-1 in cancer and other diseases, transgenic mouse models over-expressing human MCL-1 as well as humanized MCL-1 mouse models have been recently engineered. This review discusses the current advances in understanding the physiological roles of Mcl-1 based on studies using genetic murine models and its critical implications in pathology and treatment of human diseases.
Collapse
Affiliation(s)
- Hui San Chin
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Nai Yang Fu
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore.,Department of Physiology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
12
|
Genetic Basis Underlying the Hyperhemolytic Phenotype of Streptococcus agalactiae Strain CNCTC10/84. J Bacteriol 2020; 202:JB.00504-20. [PMID: 32958630 DOI: 10.1128/jb.00504-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/11/2020] [Indexed: 01/30/2023] Open
Abstract
Streptococcus agalactiae (group B streptococcus [GBS]) is a major cause of infections in newborns, pregnant women, and immunocompromised patients. GBS strain CNCTC10/84 is a clinical isolate that has high virulence in animal models of infection and has been used extensively to study GBS pathogenesis. Two unusual features of this strain are hyperhemolytic activity and hypo-CAMP factor activity. These two phenotypes are typical of GBS strains that are functionally deficient in the CovR-CovS two-component regulatory system. A previous whole-genome sequencing study found that strain CNCTC10/84 has intact covR and covS regulatory genes. We investigated CovR-CovS regulation in CNCTC10/84 and discovered that a single-nucleotide insertion in a homopolymeric tract in the covR promoter region underlies the strong hemolytic activity and weak CAMP activity of this strain. Using isogenic mutant strains, we demonstrate that this single-nucleotide insertion confers significantly decreased expression of covR and covS and altered expression of CovR-CovS-regulated genes, including that of genes encoding β-hemolysin and CAMP factor. This single-nucleotide insertion also confers significantly increased GBS survival in human whole blood ex vivo IMPORTANCE Group B streptococcus (GBS) is the leading cause of neonatal sepsis, pneumonia, and meningitis. GBS strain CNCTC10/84 is a highly virulent blood isolate that has been used extensively to study GBS pathogenesis for over 20 years. Strain CNCTC10/84 has an unusually strong hemolytic activity, but the genetic basis is unknown. In this study, we discovered that a single-nucleotide insertion in an intergenic homopolymeric tract is responsible for the elevated hemolytic activity of CNCTC10/84.
Collapse
|
13
|
Genome-Wide Assessment of Streptococcus agalactiae Genes Required for Survival in Human Whole Blood and Plasma. Infect Immun 2020; 88:IAI.00357-20. [PMID: 32747604 DOI: 10.1128/iai.00357-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/26/2020] [Indexed: 12/17/2022] Open
Abstract
Streptococcus agalactiae (group B streptococcus, or GBS) is a common cause of bacteremia and sepsis in newborns, pregnant women, and immunocompromised patients. The molecular mechanisms used by GBS to survive and proliferate in blood are not well understood. Here, using a highly virulent GBS strain and transposon-directed insertion site sequencing (TraDIS), we performed genome-wide screens to discover novel GBS genes required for bacterial survival in human whole blood and plasma. The screen identified 85 and 41 genes that are required for GBS growth in whole blood and plasma, respectively. A common set of 29 genes was required in both whole blood and plasma. Targeted gene deletion confirmed that (i) genes encoding methionine transporter (metP) and manganese transporter (mtsA) are crucial for GBS survival in whole blood and plasma, (ii) gene W903_1820, encoding a small multidrug export family protein, contributes significantly to GBS survival in whole blood, (iii) the shikimate pathway gene aroA is essential for GBS growth in whole blood and plasma, and (iv) deletion of srr1, encoding a fibrinogen-binding adhesin, increases GBS survival in whole blood. Our findings provide new insight into the GBS-host interactions in human blood.
Collapse
|
14
|
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
|
15
|
Chompunud Na Ayudhya C, Roy S, Thapaliya M, Ali H. Roles of a Mast Cell-Specific Receptor MRGPRX2 in Host Defense and Inflammation. J Dent Res 2020; 99:882-890. [PMID: 32392433 DOI: 10.1177/0022034520919107] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mast cells are multifunctional immune cells that are found most abundantly at host-environment interfaces, such as the skin, respiratory tract, and oral/gastrointestinal mucosa. Not surprisingly, mast cells act as sentinel cells that sense microbial attacks and initiate a protective immune response and promote healing. Although mast cells share many features with other innate immune effector cells, such as neutrophils and macrophages, they uniquely interact closely with blood vessels and release an extensive set of mediators for the recruitment of innate and adaptive immune cells. A novel human G protein-coupled receptor (GPCR), known as Mas-related GPCR-X2 (MRGPRX2, mouse ortholog, MrgprB2), has recently been identified, which is expressed on mast cells but not neutrophils and macrophages. Interestingly, activation of MrgprB2 by bacteria-derived quorum-sensing peptides inhibits bacterial growth, prevents biofilm formation, and leads to the recruitment of neutrophils to effectively clear bacteria. Furthermore, host defense antimicrobial peptides and small-molecule peptide mimetics also activate mast cells via MRGPRX2/B2. MrgprB2-mediated activation of local mast cells also clears cutaneous bacterial infection, promotes healing, and protects against reinfection. In addition to their role in host defense, mast cells contribute to a number of chronic inflammatory diseases such as periodontitis, neurogenic inflammation, and inflammatory pain likely via the activation of MRGPRX2. In this review, we discuss the roles of MRGPRX2/B2 in the clearance of bacterial infection, wound healing, periodontal disease, neurogenic inflammation, and inflammatory pain. We propose that harnessing mast cells' host defense and immunomodulatory properties via the activation of MRGPRX2 may lead to novel approaches for the treatment of drug-resistant bacterial infections. On the other hand, increased MRGPRX2 expression on mast cells and their inappropriate activation may contribute to periodontitis, neurogenic inflammation, and inflammatory pain. Thus, targeting MRGPRX2 could provide novel approaches to modulate these conditions.
Collapse
Affiliation(s)
- C Chompunud Na Ayudhya
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - S Roy
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M Thapaliya
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - H Ali
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
16
|
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
|
17
|
Patras KA, Coady A, Babu P, Shing SR, Ha AD, Rooholfada E, Brandt SL, Geriak M, Gallo RL, Nizet V. Host Cathelicidin Exacerbates Group B Streptococcus Urinary Tract Infection. mSphere 2020; 5:e00932-19. [PMID: 32321824 PMCID: PMC7178553 DOI: 10.1128/msphere.00932-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/06/2020] [Indexed: 12/12/2022] Open
Abstract
Group B Streptococcus (GBS) causes frequent urinary tract infection (UTI) in susceptible populations, including individuals with type 2 diabetes and pregnant women; however, specific host factors responsible for increased GBS susceptibility in these populations are not well characterized. Here, we investigate cathelicidin, a cationic antimicrobial peptide, known to be critical for defense during UTI with uropathogenic Escherichia coli (UPEC). We observed a loss of antimicrobial activity of human and mouse cathelicidins against GBS and UPEC in synthetic urine and no evidence for increased cathelicidin resistance in GBS urinary isolates. Furthermore, we found that GBS degrades cathelicidin in a protease-dependent manner. Surprisingly, in a UTI model, cathelicidin-deficient (Camp-/-) mice showed decreased GBS burdens and mast cell recruitment in the bladder compared to levels in wild-type (WT) mice. Pharmacologic inhibition of mast cells reduced GBS burdens and histamine release in WT but not Camp-/- mice. Streptozotocin-induced diabetic mice had increased bladder cathelicidin production and mast cell recruitment at 24 h postinfection with GBS compared to levels in nondiabetic controls. We propose that cathelicidin is an important immune regulator but ineffective antimicrobial peptide against GBS in urine. Combined, our findings may in part explain the increased frequency of GBS UTI in diabetic and pregnant individuals.IMPORTANCE Certain populations such as diabetic individuals are at increased risk for developing urinary tract infections (UTI), although the underlying reasons for this susceptibility are not fully known. Additionally, diabetics are more likely to become infected with certain types of bacteria, such as group B Streptococcus (GBS). In this study, we find that an antimicrobial peptide called cathelicidin, which is thought to protect the bladder from infection, is ineffective in controlling GBS and alters the type of immune cells that migrate to the bladder during infection. Using a mouse model of diabetes, we observe that diabetic mice are more susceptible to GBS infection even though they also have more infiltrating immune cells and increased production of cathelicidin. Taken together, our findings identify this antimicrobial peptide as a potential contributor to increased susceptibility of diabetic individuals to GBS UTI.
Collapse
Affiliation(s)
- Kathryn A Patras
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Alison Coady
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Priyanka Babu
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Samuel R Shing
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Albert D Ha
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Emma Rooholfada
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Stephanie L Brandt
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | | | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, La Jolla, California, USA
| | - Victor Nizet
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
18
|
Okahashi N, Nakata M, Hirose Y, Morisaki H, Kataoka H, Kuwata H, Kawabata S. Streptococcal H2O2 inhibits IgE-triggered degranulation of RBL-2H3 mast cell/basophil cell line by inducing cell death. PLoS One 2020; 15:e0231101. [PMID: 32302339 PMCID: PMC7164662 DOI: 10.1371/journal.pone.0231101] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/16/2020] [Indexed: 12/04/2022] Open
Abstract
Mast cells and basophils are central players in allergic reactions triggered by immunoglobulin E (IgE). They have intracellular granules containing allergic mediators (e.g., histamine, serotonin, inflammatory cytokines, proteases and β-hexosaminidase), and stimulation by IgE-allergen complex leads to the release of such allergic mediators from the granules, that is, degranulation. Mast cells are residents of mucosal surfaces, including those of nasal and oral cavities, and play an important role in the innate defense system. Members of the mitis group streptococci such as Streptococcus oralis, are primary colonizers of the human oral cavity. They produce hydrogen peroxide (H2O2) as a by-product of sugar metabolism. In this study, we investigated the effects of streptococcal infection on RBL-2H3 mast cell/basophil cell line. Infection by oral streptococci did not induce degranulation of the cells. Stimulation of the RBL-2H3 cells with anti-dinitrophenol (DNP) IgE and DNP-conjugated human serum albumin triggers degranulation with the release of β-hexosaminidase. We found that S. oralis and other mitis group streptococci inhibited the IgE-triggered degranulation of RBL-2H3 cells. Since mitis group streptococci produce H2O2, we examined the effect of S. oralis mutant strain deficient in producing H2O2, and found that they lost the ability to suppress the degranulation. Moreover, H2O2 alone inhibited the IgE-induced degranulation. Subsequent analysis suggested that the inhibition of degranulation was related to the cytotoxicity of streptococcal H2O2. Activated RBL-2H3 cells produce interleukin-4 (IL-4); however, IL-4 production was not induced by streptococcal H2O2. Furthermore, an in vivo study using the murine pollen-induced allergic rhinitis model suggested that the streptococcal H2O2 reduces nasal allergic reaction. These findings reveal that H2O2 produced by oral mitis group streptococci inhibits IgE-stimulated degranulation by inducing cell death. Consequently, streptococcal H2O2 can be considered to modulate the allergic reaction in mucosal surfaces.
Collapse
Affiliation(s)
- Nobuo Okahashi
- Center for Frontier Oral Science, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Masanobu Nakata
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Yujiro Hirose
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Hirobumi Morisaki
- Department of Oral Microbiology and Immunology, School of Dentistry, Showa University, Shinagawa, Tokyo, Japan
| | - Hideo Kataoka
- Department of Oral Microbiology, Asahi University School of Dentistry, Mizuho, Gifu, Japan
| | - Hirotaka Kuwata
- Department of Oral Microbiology and Immunology, School of Dentistry, Showa University, Shinagawa, Tokyo, Japan
| | - Shigetada Kawabata
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
Siemens N, Oehmcke-Hecht S, Hoßmann J, Skorka SB, Nijhuis RHT, Ruppen C, Skrede S, Rohde M, Schultz D, Lalk M, Itzek A, Pieper DH, van den Bout CJ, Claas ECJ, Kuijper EJ, Mauritz R, Sendi P, Wunderink HF, Norrby-Teglund A. Prothrombotic and Proinflammatory Activities of the β-Hemolytic Group B Streptococcal Pigment. J Innate Immun 2019; 12:291-303. [PMID: 31743913 DOI: 10.1159/000504002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 10/06/2019] [Indexed: 12/29/2022] Open
Abstract
A prominent feature of severe streptococcal infections is the profound inflammatory response that contributes to systemic toxicity. In sepsis the dysregulated host response involves both immunological and nonimmunological pathways. Here, we report a fatal case of an immunocompetent healthy female presenting with toxic shock and purpura fulminans caused by group B streptococcus (GBS; serotype III, CC19). The strain (LUMC16) was pigmented and hyperhemolytic. Stimulation of human primary cells with hyperhemolytic LUMC16 and STSS/NF-HH strains and pigment toxin resulted in a release of proinflammatory mediators, including tumor necrosis factor, interleukin (IL)-1β, and IL-6. In addition, LUMC16 induced blood clotting and showed factor XII activity on its surface, which was linked to the presence of the pigment. The expression of pigment was not linked to a mutation within the CovR/S region. In conclusion, our study shows that the hemolytic lipid toxin contributes to the ability of GBS to cause systemic hyperinflammation and interferes with the coagulation system.
Collapse
Affiliation(s)
- Nikolai Siemens
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden, .,Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, University of Greifswald, Greifswald, Germany,
| | - Sonja Oehmcke-Hecht
- Institute of Medical Microbiology, Virology, and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Jörn Hoßmann
- Microbial Interactions and Processes, Helmholtz Centre for Infection Research - HZI, Braunschweig, Germany
| | - Sebastian B Skorka
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, University of Greifswald, Greifswald, Germany
| | - Roel H T Nijhuis
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Medical Microbiology and Medical Immunology, Meander Medical Center, Amersfoort, The Netherlands
| | - Corinne Ruppen
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Steinar Skrede
- Department of Medicine, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research - HZI, Braunschweig, Germany
| | - Daniel Schultz
- Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Michael Lalk
- Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Andreas Itzek
- Microbial Interactions and Processes, Helmholtz Centre for Infection Research - HZI, Braunschweig, Germany
| | - Dietmar H Pieper
- Microbial Interactions and Processes, Helmholtz Centre for Infection Research - HZI, Braunschweig, Germany
| | | | - Eric C J Claas
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ed J Kuijper
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert Mauritz
- Department of Intensive Care Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Parham Sendi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Herman F Wunderink
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anna Norrby-Teglund
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
22
|
Repeated Vaginal Exposures to the Common Cosmetic and Household Preservative Methylisothiazolinone Induce Persistent, Mast Cell-Dependent Genital Pain in ND4 Mice. Int J Mol Sci 2019; 20:ijms20215361. [PMID: 31661848 PMCID: PMC6862067 DOI: 10.3390/ijms20215361] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/14/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022] Open
Abstract
A history of allergies doubles the risk of vulvodynia—a chronic pain condition of unknown etiology often accompanied by increases in numbers of vulvar mast cells. We previously established the biological plausibility of this relationship in mouse models where repeated exposures to the allergens oxazolone or dinitrofluorobenzene on the labiar skin or inside the vaginal canal of ND4 Swiss Webster outbred mice led to persistent tactile sensitivity and local increases in mast cells. In these models, depletion of mast cells alleviated pain. While exposure to cleaning chemicals has been connected to elevated vulvodynia risk, no single agent has been linked to adverse outcomes. We sensitized female mice to methylisothiazolinone (MI)—a biocide preservative ubiquitous in cosmetics and cleaners—dissolved in saline on their flanks, and subsequently challenged them with MI or saline for ten consecutive days in the vaginal canal. MI-challenged mice developed persistent tactile sensitivity, increased vaginal mast cells and eosinophils, and had higher serum Immunoglobulin E. Therapeutic and preventive intra-vaginal administration of Δ9-tetrahydrocannabinol reduced mast cell accumulation and tactile sensitivity. MI is known to cause skin and airway irritation in humans, and here we provide the first pre-clinical evidence that repeated MI exposures can also provoke allergy-driven genital pain.
Collapse
|
23
|
Vornhagen J, Quach P, Santana-Ufret V, Alishetti V, Brokaw A, Armistead B, Qing Tang H, MacDonald JW, Bammler TK, Adams Waldorf KM, Uldbjerg N, Rajagopal L. Human Cervical Mucus Plugs Exhibit Insufficiencies in Antimicrobial Activity Towards Group B Streptococcus. J Infect Dis 2019; 217:1626-1636. [PMID: 29425317 DOI: 10.1093/infdis/jiy076] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 02/05/2018] [Indexed: 01/31/2023] Open
Abstract
Preterm birth is a leading cause of neonatal mortality and lacks an effective therapy. Ascending microbial infections from the lower genital tract lead to infection of the placenta, amniotic fluid, and fetus causing preterm birth or stillbirth. Directly in the path of an ascending infection is the cervical mucus plug (CMP), a dense mucoid structure in the cervical canal with potential antimicrobial properties. In this study, we aimed to define the components of CMP responsible for antimicrobial activity against a common lower genital tract organism associated with preterm birth and stillbirths, namely, group B streptococcus (GBS). Using a quantitative proteomic approach, we identified antimicrobial factors in CMPs that were collected from healthy human pregnancies. However, we noted that the concentration of antimicrobial peptides present in the human CMPs were insufficient to directly kill GBS, and antimicrobial activity, when observed, was due to antibiotics retained in the CMPs. Despite this insufficiency, CMP proteins were able to activate leukocytes in whole blood resulting in increased rates of bacterial killing, suggesting a role for the CMP in enhancing complement-mediated killing or leukocyte activation. This study provides new insight into how the human CMP may limit ascending bacterial infection.
Collapse
Affiliation(s)
- Jay Vornhagen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington.,Department of Global Health, University of Washington, Seattle
| | - Phoenicia Quach
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington
| | - Verónica Santana-Ufret
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington
| | - Varchita Alishetti
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington
| | - Alyssa Brokaw
- Department of Global Health, University of Washington, Seattle
| | - Blair Armistead
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington.,Department of Pediatrics, University of Washington, Seattle.,Department of Global Health, University of Washington, Seattle
| | - Hai Qing Tang
- Department of Obstetrics and Gynecology, Aarhus University Hospital, Skejby, Denmark
| | - James W MacDonald
- Environmental and Occupational Health Sciences, University of Washington, Seattle
| | - Theo K Bammler
- Environmental and Occupational Health Sciences, University of Washington, Seattle
| | - Kristina M Adams Waldorf
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington.,Department of Obstetrics and Gynecology, University of Washington, Seattle.,Center for Innate Immunity and Immune Disease, University of Washington, Seattle.,Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Niels Uldbjerg
- Department of Obstetrics and Gynecology, Aarhus University Hospital, Skejby, Denmark
| | - Lakshmi Rajagopal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington.,Center for Innate Immunity and Immune Disease, University of Washington, Seattle.,Department of Pediatrics, University of Washington, Seattle.,Department of Global Health, University of Washington, Seattle
| |
Collapse
|
24
|
Gendrin C, Merillat S, Vornhagen J, Coleman M, Armistead B, Ngo L, Aggarwal A, Quach P, Berrigan J, Rajagopal L. Diminished Capsule Exacerbates Virulence, Blood-Brain Barrier Penetration, Intracellular Persistence, and Antibiotic Evasion of Hyperhemolytic Group B Streptococci. J Infect Dis 2019; 217:1128-1138. [PMID: 29301010 DOI: 10.1093/infdis/jix684] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/28/2017] [Indexed: 12/21/2022] Open
Abstract
Group B streptococci (GBS) are encapsulated, β-hemolytic bacteria that are a common cause of infections in human newborns and certain adults. Two factors important for GBS virulence are the sialic acid capsular polysaccharide that promotes immune evasion and the hemolytic pigment that induces host cell cytotoxcity. These virulence factors are often oppositely regulated by the CovR/CovS two-component system. Clinical GBS strains exhibiting hyperhemolysis and low capsule due to pathoadaptive covR/S mutations have been isolated from patients. Given the importance of capsule to GBS virulence, we predicted that a decrease or loss of capsule would attenuate the virulence of covR/S mutants. Surprisingly, hyperhemolytic GBS with low or no capsule exhibit increased virulence, intracellular persistence, and blood-brain barrier penetration, which was independent of a Trojan horse mechanism of barrier penetration. Additionally, intracellular persistence enabled both hemolytic and hyperhemolytic GBS to evade antibiotics routinely used to treat these infections. The finding that diminished capsule expression promotes GBS virulence, intracellular persistence, and antibiotic evasion has important implications for sustained antibiotic therapy and efficacy of capsule-based vaccines.
Collapse
Affiliation(s)
- Claire Gendrin
- Department of Pediatrics, University of Washington.,Center for Global Infections Disease Research, Seattle Children's Research Institute
| | - Sean Merillat
- Center for Global Infections Disease Research, Seattle Children's Research Institute
| | - Jay Vornhagen
- Department of Pediatrics, University of Washington.,Center for Global Infections Disease Research, Seattle Children's Research Institute.,Department of Global Health, University of Washington, Seattle
| | - Michelle Coleman
- Department of Pediatrics, University of Washington.,Center for Global Infections Disease Research, Seattle Children's Research Institute
| | - Blair Armistead
- Department of Pediatrics, University of Washington.,Center for Global Infections Disease Research, Seattle Children's Research Institute.,Department of Global Health, University of Washington, Seattle
| | - Lisa Ngo
- Center for Global Infections Disease Research, Seattle Children's Research Institute
| | - Anjali Aggarwal
- Center for Global Infections Disease Research, Seattle Children's Research Institute
| | - Phoenicia Quach
- Center for Global Infections Disease Research, Seattle Children's Research Institute
| | - Jacob Berrigan
- Center for Global Infections Disease Research, Seattle Children's Research Institute
| | - Lakshmi Rajagopal
- Department of Pediatrics, University of Washington.,Center for Global Infections Disease Research, Seattle Children's Research Institute.,Department of Global Health, University of Washington, Seattle
| |
Collapse
|
25
|
Piliponsky AM, Acharya M, Shubin NJ. Mast Cells in Viral, Bacterial, and Fungal Infection Immunity. Int J Mol Sci 2019; 20:ijms20122851. [PMID: 31212724 PMCID: PMC6627964 DOI: 10.3390/ijms20122851] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/31/2019] [Accepted: 06/11/2019] [Indexed: 01/01/2023] Open
Abstract
Mast cells are granule-rich immune cells that are distributed throughout the body in areas where microorganisms typically reside, such as mucosal tissues and the skin, as well as connective tissues. It is well known that mast cells have significant roles in IgE-mediated conditions, such as anaphylaxis, but, because of their location, it is also thought that mast cells act as innate immune cells against pathogens and initiate defensive immune responses. In this review, we discuss recent studies focused on mast cell interactions with flaviviruses and Candida albicans, and mast cell function in the cecal ligation and puncture model of sepsis. We selected these studies because they are clear examples of how mast cells can either promote host resistance to infection, as previously proposed, or contribute to a dysregulated host response that can increase host morbidity and mortality. Importantly, we can distill from these studies that the contribution of mast cells to infection outcomes depends in part on the infection model, including the genetic approach used to assess the influence of mast cells on host immunity, the species in which mast cells are studied, and the differential contribution of mast cell subtypes to immunity. Accordingly, we think that this review highlights the complexity of mast cell biology in the context of innate immune responses.
Collapse
Affiliation(s)
- Adrian M Piliponsky
- Departments of Pediatrics and Pathology, University of Washington, Seattle, WA 98195, USA.
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA.
| | - Manasa Acharya
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA.
| | - Nicholas J Shubin
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA.
| |
Collapse
|
26
|
Varricchi G, Rossi FW, Galdiero MR, Granata F, Criscuolo G, Spadaro G, de Paulis A, Marone G. Physiological Roles of Mast Cells: Collegium Internationale Allergologicum Update 2019. Int Arch Allergy Immunol 2019; 179:247-261. [PMID: 31137021 DOI: 10.1159/000500088] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 04/02/2019] [Indexed: 11/19/2022] Open
Abstract
Mast cells are immune cells which have a widespread distribution in nearly all tissues. These cells and their mediators are canonically viewed as primary effector cells in allergic disorders. However, in the last years, mast cells have gained recognition for their involvement in several physiological and pathological conditions. They are highly heterogeneous immune cells displaying a constellation of surface receptors and producing a wide spectrum of inflammatory and immunomodulatory mediators. These features enable the cells to act as sentinels in harmful situations as well as respond to metabolic and immune changes in their microenvironment. Moreover, they communicate with many immune and nonimmune cells implicated in several immunological responses. Although mast cells contribute to host responses in experimental infections, there is no satisfactory model to study how they contribute to infection outcome in humans. Mast cells modulate physiological and pathological angiogenesis and lymphangiogenesis, but their role in tumor initiation and development is still controversial. Cardiac mast cells store and release several mediators that can exert multiple effects in the homeostatic control of different cardiometabolic functions. Although mast cells and their mediators have been simplistically associated with detrimental roles in allergic disorders, there is increasing evidence that they can also have homeostatic or protective roles in several pathophysiological processes. These findings may reflect the functional heterogeneity of different subsets of mast cells.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences (DiSMeT), Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy
| | - Francesca Wanda Rossi
- Department of Translational Medical Sciences (DiSMeT), Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences (DiSMeT), Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy
| | - Francescopaolo Granata
- Department of Translational Medical Sciences (DiSMeT), Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy
| | - Gjada Criscuolo
- Department of Translational Medical Sciences (DiSMeT), Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences (DiSMeT), Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences (DiSMeT), Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences (DiSMeT), Naples, Italy, .,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy, .,World Allergy Organization (WAO) Center of Excellence, Naples, Italy, .,Institute of Endocrinology and Experimental Oncology (IEOS), CNR, Naples, Italy,
| |
Collapse
|
27
|
Armistead B, Oler E, Adams Waldorf K, Rajagopal L. The Double Life of Group B Streptococcus: Asymptomatic Colonizer and Potent Pathogen. J Mol Biol 2019; 431:2914-2931. [PMID: 30711542 DOI: 10.1016/j.jmb.2019.01.035] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 12/22/2022]
Abstract
Group B streptococcus (GBS) is a β-hemolytic gram-positive bacterium that colonizes the lower genital tract of approximately 18% of women globally as an asymptomatic member of the gastrointestinal and/or vaginal flora. If established in other host niches, however, GBS is highly pathogenic. During pregnancy, ascending GBS infection from the vagina to the intrauterine space is associated with preterm birth, stillbirth, and fetal injury. In addition, vertical transmission of GBS during or after birth results in life-threatening neonatal infections, including pneumonia, sepsis, and meningitis. Although the mechanisms by which GBS traffics from the lower genital tract to vulnerable host niches are not well understood, recent advances have revealed that many of the same bacterial factors that promote asymptomatic vaginal carriage also facilitate dissemination and virulence. Furthermore, highly pathogenic GBS strains have acquired unique factors that enhance survival in invasive niches. Several host factors also exist that either subdue GBS upon vaginal colonization or alternatively permit invasive infection. This review summarizes the GBS and host factors involved in GBS's state as both an asymptomatic colonizer and an invasive pathogen. Gaining a better understanding of these mechanisms is key to overcoming the challenges associated with vaccine development and identification of novel strategies to mitigate GBS virulence.
Collapse
Affiliation(s)
- Blair Armistead
- Department of Global Health, University of Washington, Seattle 98195, WA, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle 98101, WA, USA
| | - Elizabeth Oler
- Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle 98195, WA, USA
| | - Kristina Adams Waldorf
- Department of Global Health, University of Washington, Seattle 98195, WA, USA; Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle 98195, WA, USA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle 98109, WA, USA; Sahlgrenska Academy, Gothenburg University, Gothenburg 413 90, Sweden
| | - Lakshmi Rajagopal
- Department of Global Health, University of Washington, Seattle 98195, WA, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle 98101, WA, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle 98195, WA, USA.
| |
Collapse
|
28
|
Arifuzzaman M, Mobley YR, Choi HW, Bist P, Salinas CA, Brown ZD, Chen SL, Staats HF, Abraham SN. MRGPR-mediated activation of local mast cells clears cutaneous bacterial infection and protects against reinfection. SCIENCE ADVANCES 2019; 5:eaav0216. [PMID: 30613778 PMCID: PMC6314830 DOI: 10.1126/sciadv.aav0216] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 11/27/2018] [Indexed: 05/22/2023]
Abstract
Mast cells (MCs) are strategically distributed at barrier sites and prestore various immunocyte-recruiting cytokines, making them ideal targets for selective activation to treat peripheral infections. Here, we report that topical treatment with mastoparan, a peptide MC activator (MCA), enhances clearance of Staphylococcus aureus from infected mouse skins and accelerates healing of dermonecrotic lesions. Mastoparan functions by activating connective tissue MCs (CTMCs) via the MRGPRX2 (Mas-related G protein-coupled receptor member X2) receptor. Peripheral CTMC activation, in turn, enhances recruitment of bacteria-clearing neutrophils and wound-healing CD301b+ dendritic cells. Consistent with MCs playing a master coordinating role, MC activation also augmented migration of various antigen-presenting dendritic cells to draining lymph nodes, leading to stronger protection against a second infection challenge. MCAs therefore orchestrate both the innate and adaptive immune arms, which could potentially be applied to combat peripheral infections by a broad range of pathogens.
Collapse
Affiliation(s)
- Mohammad Arifuzzaman
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Yuvon R. Mobley
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Hae Woong Choi
- Department of Pathology, Duke University, Durham, NC 27710, USA
| | - Pradeep Bist
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore 169857, Singapore
| | | | - Zachary D. Brown
- Undergraduate Program in Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Swaine L. Chen
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Infectious Diseases Group, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Herman F. Staats
- Department of Pathology, Duke University, Durham, NC 27710, USA
- Department of Immunology, Duke University, Durham, NC 27710, USA
- Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Soman N. Abraham
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
- Department of Pathology, Duke University, Durham, NC 27710, USA
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore 169857, Singapore
- Department of Immunology, Duke University, Durham, NC 27710, USA
- Corresponding author.
| |
Collapse
|
29
|
Vornhagen J, Armistead B, Santana-Ufret V, Gendrin C, Merillat S, Coleman M, Quach P, Boldenow E, Alishetti V, Leonhard-Melief C, Ngo LY, Whidbey C, Doran KS, Curtis C, Waldorf KMA, Nance E, Rajagopal L. Group B streptococcus exploits vaginal epithelial exfoliation for ascending infection. J Clin Invest 2018; 128:1985-1999. [PMID: 29629904 DOI: 10.1172/jci97043] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 02/27/2018] [Indexed: 12/11/2022] Open
Abstract
Thirteen percent of pregnancies result in preterm birth or stillbirth, accounting for fifteen million preterm births and three and a half million deaths annually. A significant cause of these adverse pregnancy outcomes is in utero infection by vaginal microorganisms. To establish an in utero infection, vaginal microbes enter the uterus by ascending infection; however, the mechanisms by which this occurs are unknown. Using both in vitro and murine models of vaginal colonization and ascending infection, we demonstrate how a vaginal microbe, group B streptococcus (GBS), which is frequently associated with adverse pregnancy outcomes, uses vaginal exfoliation for ascending infection. GBS induces vaginal epithelial exfoliation by activation of integrin and β-catenin signaling. However, exfoliation did not diminish GBS vaginal colonization as reported for other vaginal microbes. Rather, vaginal exfoliation increased bacterial dissemination and ascending GBS infection, and abrogation of exfoliation reduced ascending infection and improved pregnancy outcomes. Thus, for some vaginal bacteria, exfoliation promotes ascending infection rather than preventing colonization. Our study provides insight into mechanisms of ascending infection by vaginal microbes.
Collapse
Affiliation(s)
- Jay Vornhagen
- Department of Global Health, University of Washington, Seattle, Washington, USA.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - 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
| | - Verónica Santana-Ufret
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Claire Gendrin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Sean Merillat
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Michelle Coleman
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA.,Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Phoenicia Quach
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Erica Boldenow
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA.,Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Varchita Alishetti
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | | | - Lisa Y Ngo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Christopher Whidbey
- Department of Global Health, University of Washington, Seattle, Washington, USA.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Kelly S Doran
- Department of Microbiology and Immunology, University of Colorado Denver, Aurora, Colorado, USA
| | | | - Kristina M Adams Waldorf
- Department of Global Health, University of Washington, Seattle, Washington, USA.,Department of Obstetrics and Gynecology, and.,Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington, USA.,Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | | | - 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, Seattle, Washington, USA.,Center for Innate Immunity and Immune Disease, University of Washington, Seattle, Washington, USA
| |
Collapse
|
30
|
Mitchell T, MacDonald JW, Srinouanpranchanh S, Bammler TK, Merillat S, Boldenow E, Coleman M, Agnew K, Baldessari A, Stencel-Baerenwald JE, Tisoncik-Go J, Green RR, Gale MJ, Rajagopal L, Adams Waldorf KM. Evidence of cardiac involvement in the fetal inflammatory response syndrome: disruption of gene networks programming cardiac development in nonhuman primates. Am J Obstet Gynecol 2018; 218:438.e1-438.e16. [PMID: 29475580 PMCID: PMC6070341 DOI: 10.1016/j.ajog.2018.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/22/2017] [Accepted: 01/04/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Most early preterm births are associated with intraamniotic infection and inflammation, which can lead to systemic inflammation in the fetus. The fetal inflammatory response syndrome describes elevations in the fetal interleukin-6 level, which is a marker for inflammation and fetal organ injury. An understanding of the effects of inflammation on fetal cardiac development may lead to insight into the fetal origins of adult cardiovascular disease. OBJECTIVE The purpose of this study was to determine whether the fetal inflammatory response syndrome is associated with disruptions in gene networks that program fetal cardiac development. STUDY DESIGN We obtained fetal cardiac tissue after necropsy from a well-described pregnant nonhuman primate model (pigtail macaque, Macaca nemestrina) of intrauterine infection (n=5) and controls (n=5). Cases with the fetal inflammatory response syndrome (fetal plasma interleukin-6 >11 pg/mL) were induced by either choriodecidual inoculation of a hypervirulent group B streptococcus strain (n=4) or intraamniotic inoculation of Escherichia coli (n=1). RNA and protein were extracted from fetal hearts and profiled by microarray and Luminex (Millipore, Billerica, MA) for cytokine analysis, respectively. Results were validated by quantitative reverse transcriptase polymerase chain reaction. Statistical and bioinformatics analyses included single gene analysis, gene set analysis, Ingenuity Pathway Analysis (Qiagen, Valencia, CA), and Wilcoxon rank sum. RESULTS Severe fetal inflammation developed in the context of intraamniotic infection and a disseminated bacterial infection in the fetus. Interleukin-6 and -8 in fetal cardiac tissues were elevated significantly in fetal inflammatory response syndrome cases vs controls (P<.05). A total of 609 probe sets were expressed differentially (>1.5-fold change, P<.05) in the fetal heart (analysis of variance). Altered expression of select genes was validated by quantitative reverse transcriptase polymerase chain reaction that included several with known functions in cardiac injury, morphogenesis, angiogenesis, and tissue remodeling (eg, angiotensin I converting enzyme 2, STEAP family member 4, natriuretic peptide A, and secreted frizzled-related protein 4; all P<.05). Multiple gene sets and pathways that are involved in cardiac morphogenesis and vasculogenesis were downregulated significantly by gene set and Ingenuity Pathway Analysis (hallmark transforming growth factor beta signaling, cellular morphogenesis during differentiation, morphology of cardiovascular system; all P<.05). CONCLUSION Disruption of gene networks for cardiac morphogenesis and vasculogenesis occurred in the preterm fetal heart of nonhuman primates with preterm labor, intraamniotic infection, and severe fetal inflammation. Inflammatory injury to the fetal heart in utero may contribute to the development of heart disease later in life. Development of preterm labor therapeutics must also target fetal inflammation to lessen organ injury and potential long-term effects on cardiac function.
Collapse
Affiliation(s)
- Timothy Mitchell
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | | | - Theodor K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Sean Merillat
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Erica Boldenow
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | | | - Kathy Agnew
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA
| | - Audrey Baldessari
- Washington National Primate Research Center, University of Washington, Seattle, WA
| | - Jennifer E Stencel-Baerenwald
- Department of Immunology, University of Washington, Seattle, WA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA
| | - Jennifer Tisoncik-Go
- Department of Immunology, University of Washington, Seattle, WA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA
| | - Richard R Green
- Department of Immunology, University of Washington, Seattle, WA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA
| | - Michael J Gale
- Department of Immunology, University of Washington, Seattle, WA; Department of Global Health, University of Washington, Seattle, WA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA
| | - Lakshmi Rajagopal
- Department of Pediatrics, University of Washington, Seattle, WA; Department of Global Health, University of Washington, Seattle, WA; 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 & Gynecology, University of Washington, Seattle, WA; Department of Global Health, University of Washington, Seattle, WA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA; Sahlgrenska Academy, Gothenburg, Sweden.
| |
Collapse
|
31
|
Lannon SMR, Adams Waldorf KM, Fiedler T, Kapur RP, Agnew K, Rajagopal L, Gravett MG, Fredricks DN. Parallel detection of lactobacillus and bacterial vaginosis-associated bacterial DNA in the chorioamnion and vagina of pregnant women at term. J Matern Fetal Neonatal Med 2018; 32:2702-2710. [PMID: 29478370 DOI: 10.1080/14767058.2018.1446208] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND The majority of early preterm births are associated with intrauterine infections, which are thought to occur when microbes traffic into the uterus from the lower genital tract and seed the placenta. Bacterial vaginosis (BV) is associated with heterogeneous bacterial communities in the vagina and is linked to preterm birth. The extent to which trafficking into the uterus of normal and BV-associated vaginal bacteria occurs is unknown. The study objective was to characterize in parallel the distribution and quantities of bacteria in the vagina, uterus, and placental compartments. METHODS Pregnant women at term (≥37 weeks) presenting for delivery were recruited prospectively. Swabs were collected in parallel from the vagina, chorioamnion. Choriodecidual swabs were collected if a cesarean section was performed. Samples were analyzed by culture, broad-range 16S rRNA gene PCR, and bacterial species-specific quantitative PCR (qPCR) for DNA from Lactobacillus and a panel of BV-associated bacteria. Results were correlated with placental histopathology. RESULTS Of the 23 women enrolled, 15 were delivered by cesarean section (N = 10 without labor; N = 5 in labor) and eight were delivered vaginally. BV was diagnosed in two women not in labor. Placental histopathology identified chorioamnionitis or funisitis in six cases [1/10 (10%) not in labor; 5/13 (38%) in labor]. Among non-laboring women, broad-range 16S qPCR detected bacteria in the chorioamnion and the choriodecidua (4/10; 40%). Among laboring women, Lactobacillus species were frequently detected in the chorioamnion by qPCR (4/13; 31%). In one case, mild chorioamnionitis was associated with qPCR detection of similar microbes in the chorioamnion and vagina (e.g. Leptotrichia/Sneathia, Megasphaera), along a quantitative gradient. CONCLUSIONS Microbial trafficking of lactobacilli and fastidious bacteria into the chorioamniotic membranes and choriodecidua occurs at term in normal pregnancies. In one case, we demonstrated a quantitative gradient between multiple bacterial species in the lower genital tract and placenta. Not all bacterial colonization is associated with placental inflammation and clinical sequelae. Further studies of the role of placental colonization with Lactobacillus in normal pregnancy and fastidious bacteria in chorioamnionitis may improve prevention and treatment approaches for preterm labor.
Collapse
Affiliation(s)
- Sophia M R Lannon
- a Department of Obstetrics & Gynecology , University of Washington , Seattle, Washington , DC , USA.,b Northwest Perinatal Associates , Portland , OR , USA
| | - Kristina M Adams Waldorf
- a Department of Obstetrics & Gynecology , University of Washington , Seattle, Washington , DC , USA
| | - Tina Fiedler
- c Fred Hutchinson Cancer Research Center , Seattle, Washington , DC , USA
| | - Raj P Kapur
- d Department of Pathology , University of Washington , Seattle, Washington , DC , USA.,e Department of Pathology , Seattle Children's Hospital , Seattle, Washington , DC , USA
| | - Kathy Agnew
- a Department of Obstetrics & Gynecology , University of Washington , Seattle, Washington , DC , USA
| | - Lakshmi Rajagopal
- f Department of Pediatrics , University of Washington , Seattle, Washington , DC , USA.,g Center for Global Infectious Disease Research, Seattle Children's Research Institute , Seattle , Washington , DC , USA
| | - Michael G Gravett
- a Department of Obstetrics & Gynecology , University of Washington , Seattle, Washington , DC , USA
| | - David N Fredricks
- c Fred Hutchinson Cancer Research Center , Seattle, Washington , DC , USA
| |
Collapse
|
32
|
Abstract
Mast cells are hematopoietic progenitor-derived, granule-containing immune cells that are widely distributed in tissues that interact with the external environment, such as the skin and mucosal tissues. It is well-known that mast cells are significantly involved in IgE-mediated allergic reactions, but because of their location, it has also been long hypothesized that mast cells can act as sentinel cells that sense pathogens and initiate protective immune responses. Using mast cell or mast cell protease-deficient murine models, recent studies by our groups and others indicate that mast cells have pleiotropic regulatory roles in immunological responses against pathogens. In this review, we discuss studies that demonstrate that mast cells can either promote host resistance to infections caused by bacteria and fungi or contribute to dysregulated immune responses that can increase host morbidity and mortality. Overall, these studies indicate that mast cells can influence innate immune responses against bacterial and fungal infections via multiple mechanisms. Importantly, the contribution of mast cells to infection outcomes depends in part on the infection model, including the genetic approach used to assess the influence of mast cells on host immunity, hence highlighting the complexity of mast cell biology in the context of innate immune responses.
Collapse
Affiliation(s)
- Adrian M Piliponsky
- Departments of Pediatrics and Pathology, University of Washington, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Luigina Romani
- Pathology Section, Department of Experimental Medicine, University of Perugia, Perugia, Italy
- Center of functional genomics (C.U.R.Ge.F.), Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
33
|
Protective effect of Group B Streptococcus type-III polysaccharide conjugates against maternal colonization, ascending infection and neonatal transmission in rodent models. Sci Rep 2018; 8:2593. [PMID: 29416049 PMCID: PMC5803199 DOI: 10.1038/s41598-018-20609-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/18/2018] [Indexed: 12/25/2022] Open
Abstract
Group B Streptococcus (GBS) is a normal inhabitant of recto-vaginal mucosae in up to 30% of healthy women. Colonization is a major risk factor for perinatal infection which can lead to severe complications such as stillbirth and neonatal invasive disease. Intra-partum antibiotic prophylaxis in colonized women is a safe and cost-effective preventive measure against early-onset disease in the first days of life, but has no effect on late-onset manifestations or on early maternal infection. Maternal immunization with capsular polysaccharide-based vaccines shows promise for the prevention of both early-onset and late-onset neonatal infections, although ability to prevent maternal colonization and ascending infection has been less studied. Here we investigated the effect of a GBS glycoconjugate vaccine since the very early stage of maternal GBS acquisition to neonatal outcome by rodent models of vaginal colonization and ascending infection. Immunization of female mice and rats with a type III glycoconjugate reduced vaginal colonization, infection of chorioamniotic/ placental membranes and bacterial transmission to fetuses and pups. Type III specific antibodies were detected in the blood and vagina of vaccinated mothers and their offspring. The obtained data support a potential preventive effect of GBS glycoconjugate vaccines during the different stages of pregnancy.
Collapse
|
34
|
Lee JH, Kang S, Ahn M, Jang H, Min DH. Development of Dual-Pore Coexisting Branched Silica Nanoparticles for Efficient Gene-Chemo Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 13. [PMID: 29251426 DOI: 10.1002/smll.201602363] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/12/2016] [Indexed: 05/15/2023]
Abstract
Various strategies for combination therapy to overcome current limitations in cancer therapy have been actively investigated. Among them, simultaneous delivery of multiple drugs is a subject of high interest due to anticipated synergistic effect, but there have been difficulties in designing and developing effective nanomaterials for this purpose. In this work, dual-pore coexisting hybrid porous silica nanoparticles are developed through Volmer-Weber growth pathway for efficient co-delivery of gene and anticancer drug. Based on the different pore sizes (2-3 and 40-45 nm) and surface modifications of the core and branch domains, loading and controlled release of gene and drug are achieved by appropriate strategies for each environment. With excellent loading capacity and low cytotoxicity of the present platform, the combinational cancer therapy is successfully demonstrated against human cervical cancer cell line. Through a series of quantitative analyses, the excellent gene-chemo combinational therapeutic efficiency is successfully demonstrated. It is expected that the present nanoparticle will be applicable to various biomedical fields that require co-delivery of small molecule and nucleic acid.
Collapse
Affiliation(s)
- Jong-Hwan Lee
- Department of Chemistry, Seoul National University, Center for RNA Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Seounghun Kang
- Department of Chemistry, Seoul National University, Center for RNA Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Minchul Ahn
- Department of Chemistry, Seoul National University, Center for RNA Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Hongje Jang
- Department of Chemistry, Kwangwoon University, 20 Gwangwoon-ro, Nowon-gu, Seoul, 01897, Republic of Korea
| | - Dal-Hee Min
- Department of Chemistry, Seoul National University, Center for RNA Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Institute of Nanobio Convergence Technology, Lemonex Inc., Seoul, 08826, Republic of Korea
| |
Collapse
|
35
|
Patras KA, Nizet V. Group B Streptococcal Maternal Colonization and Neonatal Disease: Molecular Mechanisms and Preventative Approaches. Front Pediatr 2018; 6:27. [PMID: 29520354 PMCID: PMC5827363 DOI: 10.3389/fped.2018.00027] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Group B Streptococcus (GBS) colonizes the gastrointestinal and vaginal epithelium of a significant percentage of healthy women, with potential for ascending intrauterine infection or transmission during parturition, creating a risk of serious disease in the vulnerable newborn. This review highlights new insights on the bacterial virulence determinants, host immune responses, and microbiome interactions that underpin GBS vaginal colonization, the proximal step in newborn infectious disease pathogenesis. From the pathogen perspective, the function GBS adhesins and biofilms, β-hemolysin/cytolysin toxin, immune resistance factors, sialic acid mimicry, and two-component transcriptional regulatory systems are reviewed. From the host standpoint, pathogen recognition, cytokine responses, and the vaginal mucosal and placental immunity to the pathogen are detailed. Finally, the rationale, efficacy, and potential unintended consequences of current universal recommended intrapartum antibiotic prophylaxis are considered, with updates on new developments toward a GBS vaccine or alternative approaches to reducing vaginal colonization.
Collapse
Affiliation(s)
- Kathryn A Patras
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States
| | - Victor Nizet
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
36
|
Gendrin C, Shubin NJ, Boldenow E, Merillat S, Clauson M, Power D, Doran KS, Abrink M, Pejler G, Rajagopal L, Piliponsky AM. Mast cell chymase decreases the severity of group B Streptococcus infections. J Allergy Clin Immunol 2017; 142:120-129.e6. [PMID: 28916188 DOI: 10.1016/j.jaci.2017.07.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 07/06/2017] [Accepted: 07/24/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Group B Streptococcus (GBS) or Streptococcus agalactiae are β-hemolytic gram-positive bacteria that colonize the lower genital tracts of women and are frequently associated with infections during pregnancy. Innate immune defenses are critical for controlling GBS dissemination and systemic infection. Mast cells are resident sentinel cells that come into contact with pathogens early during colonization and infection. OBJECTIVE We aimed to investigate the contribution of chymase to systemic GBS infection and rates of preterm birth. METHODS Pharmacologic and genetic approaches using mice deficient in mast cell protease (MCPT) 4, the mouse functional homologue of human chymase, were used. RESULTS Our studies show that mast cells release a protease with chymotrypsin-like cleavage specificity in response to GBS. Additionally, increased GBS systemic infection and preterm births were observed in MCPT4-deficient mice versus MCPT4-sufficient mice. Furthermore, we observed that proteolytic cleavage of the host extracellular matrix protein fibronectin by peritoneal cell-derived mast cell lysates diminished GBS adherence. Consistent with this observation, the increase in GBS dissemination and preterm births observed in MCPT4-deficient mice was abolished when GBS was deficient in expression of the fibronectin-binding protein SfbA. CONCLUSIONS Taken together, our results suggest that the protective effect of MCPT4 against GBS dissemination and preterm labor can be attributed in part to MCPT4-mediated proteolysis of fibronectin. Our studies reveal a novel role of mast cells in defense against bacterial infections.
Collapse
Affiliation(s)
- Claire Gendrin
- Department of Pediatric Infectious Diseases, University of Washington, Seattle, Wash; Seattle Children's Research Institute, Seattle, Wash
| | | | | | - Sean Merillat
- Seattle Children's Research Institute, Seattle, Wash
| | | | - Danial Power
- Seattle Children's Research Institute, Seattle, Wash
| | - Kelly S Doran
- Department of Biology and Center for Microbial Sciences, San Diego State University, San Diego, Calif; Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, Calif
| | - Magnus Abrink
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University for Agricultural Sciences, Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Department of Anatomy, Physiology and Biochemistry, the Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Lakshmi Rajagopal
- Department of Pediatric Infectious Diseases, University of Washington, Seattle, Wash; Seattle Children's Research Institute, Seattle, Wash; Department of Global Health, University of Washington, Seattle, Wash.
| | - Adrian M Piliponsky
- Department of Pediatric Infectious Diseases, University of Washington, Seattle, Wash; Seattle Children's Research Institute, Seattle, Wash.
| |
Collapse
|
37
|
Vornhagen J, Adams Waldorf KM, Rajagopal L. Perinatal Group B Streptococcal Infections: Virulence Factors, Immunity, and Prevention Strategies. Trends Microbiol 2017. [PMID: 28633864 DOI: 10.1016/j.tim.2017.05.013] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Group B streptococcus (GBS) or Streptococcus agalactiae is a β-hemolytic, Gram-positive bacterium that is a leading cause of neonatal infections. GBS commonly colonizes the lower gastrointestinal and genital tracts and, during pregnancy, neonates are at risk of infection. Although intrapartum antibiotic prophylaxis during labor and delivery has decreased the incidence of early-onset neonatal infection, these measures do not prevent ascending infection that can occur earlier in pregnancy leading to preterm births, stillbirths, or late-onset neonatal infections. Prevention of GBS infection in pregnancy is complex and is likely influenced by multiple factors, including pathogenicity, host factors, vaginal microbiome, false-negative screening, and/or changes in antibiotic resistance. A deeper understanding of the mechanisms of GBS infections during pregnancy will facilitate the development of novel therapeutics and vaccines. Here, we summarize and discuss important advancements in our understanding of GBS vaginal colonization, ascending infection, and preterm birth.
Collapse
Affiliation(s)
- Jay Vornhagen
- Department of Global Health, University of Washington, Seattle, WA, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Kristina M Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Lakshmi Rajagopal
- Department of Global Health, University of Washington, Seattle, WA, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
38
|
Abstract
Streptococcus agalactiae (group B Streptococcus, GBS), is a Gram-positive, asymptomatic colonizer of the human gastrointestinal tract and vaginal tract of 10 - 30% of adults. In immune-compromised individuals, including neonates, pregnant women, and the elderly, GBS may switch to an invasive pathogen causing sepsis, arthritis, pneumonia, and meningitis. Because GBS is a leading bacterial pathogen of neonates, current prophylaxis is comprised of late gestation screening for GBS vaginal colonization and subsequent peripartum antibiotic treatment of GBS-positive mothers. Heavy GBS vaginal burden is a risk factor for both neonatal disease and colonization. Unfortunately, little is known about the host and bacterial factors that promote or permit GBS vaginal colonization. This protocol describes a technique for establishing persistent GBS vaginal colonization using a single β-estradiol pre-treatment and daily sampling to determine bacterial load. It further details methods to administer additional therapies or reagents of interest and to collect vaginal lavage fluid and reproductive tract tissues. This mouse model will further the understanding of the GBS-host interaction within the vaginal environment, which will lead to potential therapeutic targets to control maternal vaginal colonization during pregnancy and to prevent transmission to the vulnerable newborn. It will also be of interest to increase our understanding of general bacterial-host interactions in the female vaginal tract.
Collapse
Affiliation(s)
- Kathryn A Patras
- Department of Pediatrics, Division of Host-Microbe Systems & Therapeutics, University of California San Diego School of Medicine
| | - Kelly S Doran
- Department of Pediatrics, Division of Host-Microbe Systems & Therapeutics, University of California San Diego School of Medicine; Department of Biology and Center for Microbial Sciences, San Diego State University;
| |
Collapse
|
39
|
Boldenow E, Gendrin C, Ngo L, Bierle C, Vornhagen J, Coleman M, Merillat S, Armistead B, Whidbey C, Alishetti V, Santana-Ufret V, Ogle J, Gough M, Srinouanprachanh S, MacDonald JW, Bammler TK, Bansal A, Liggitt HD, Rajagopal L, Adams Waldorf KM. Group B Streptococcus circumvents neutrophils and neutrophil extracellular traps during amniotic cavity invasion and preterm labor. Sci Immunol 2016; 1:1/4/eaah4576. [PMID: 27819066 DOI: 10.1126/sciimmunol.aah4576] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Preterm birth is a leading cause of neonatal morbidity and mortality. Although microbial invasion of the amniotic cavity (MIAC) is associated with the majority of early preterm births, the temporal events that occur during MIAC and preterm labor are not known. Group B Streptococci (GBS) are β-hemolytic, gram-positive bacteria, which commonly colonize the vagina but have been recovered from the amniotic fluid in preterm birth cases. To understand temporal events that occur during MIAC, we utilized a unique chronically catheterized nonhuman primate model that closely emulates human pregnancy. This model allows monitoring of uterine contractions, timing of MIAC and immune responses during pregnancy-associated infections. Here, we show that adverse outcomes such as preterm labor, MIAC, and fetal sepsis were observed more frequently during infection with hemolytic GBS when compared to nonhemolytic GBS. Although MIAC was associated with systematic progression in chorioamnionitis beginning with chorionic vasculitis and progressing to neutrophilic infiltration, the ability of the GBS hemolytic pigment toxin to induce neutrophil cell death and subvert killing by neutrophil extracellular traps (NETs) in placental membranes in vivo facilitated MIAC and fetal injury. Furthermore, compared to maternal neutrophils, fetal neutrophils exhibit decreased neutrophil elastase activity and impaired phagocytic functions to GBS. Collectively, our studies demonstrate how a unique bacterial hemolytic lipid toxin enables GBS to circumvent neutrophils and NETs in placental membranes to induce fetal injury and preterm labor.
Collapse
Affiliation(s)
- Erica Boldenow
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Claire Gendrin
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Lisa Ngo
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Craig Bierle
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Jay Vornhagen
- Department of Pediatric Infectious Diseases, University of Washington and 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
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Sean Merillat
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Blair Armistead
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America; Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Christopher Whidbey
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America; Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Varchita Alishetti
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Veronica Santana-Ufret
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Jason Ogle
- Washington National Primate Center, University of Washington, Seattle, Washington, United States of America
| | - Michael Gough
- Washington National Primate Center, University of Washington, Seattle, Washington, United States of America
| | - Sengkeo Srinouanprachanh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Aasthaa Bansal
- Department of Pharmacy, University of Washington, Seattle, Washington, United States of America
| | - H Denny Liggitt
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Lakshmi Rajagopal
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America; Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Kristina M Adams Waldorf
- Department of Obstetrics & Gynecology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
40
|
Abstract
UNLABELLED Preterm birth increases the risk of adverse birth outcomes and is the leading cause of neonatal mortality. A significant cause of preterm birth is in utero infection with vaginal microorganisms. These vaginal microorganisms are often recovered from the amniotic fluid of preterm birth cases. A vaginal microorganism frequently associated with preterm birth is group B streptococcus (GBS), or Streptococcus agalactiae However, the molecular mechanisms underlying GBS ascension are poorly understood. Here, we describe the role of the GBS hyaluronidase in ascending infection and preterm birth. We show that clinical GBS strains associated with preterm labor or neonatal infections have increased hyaluronidase activity compared to commensal strains obtained from rectovaginal swabs of healthy women. Using a murine model of ascending infection, we show that hyaluronidase activity was associated with increased ascending GBS infection, preterm birth, and fetal demise. Interestingly, hyaluronidase activity reduced uterine inflammation but did not impact placental or fetal inflammation. Our study shows that hyaluronidase activity enables GBS to subvert uterine immune responses, leading to increased rates of ascending infection and preterm birth. These findings have important implications for the development of therapies to prevent in utero infection and preterm birth. IMPORTANCE GBS are a family of bacteria that frequently colonize the vagina of pregnant women. In some cases, GBS ascend from the vagina into the uterine space, leading to fetal injury and preterm birth. Unfortunately, little is known about the mechanisms underlying ascending GBS infection. In this study, we show that a GBS virulence factor, HylB, shows higher activity in strains isolated from cases of preterm birth than those isolates from rectovaginal swabs of healthy women. We discovered that GBS rely on HylB to avoid immune detection in uterine tissue, but not placental tissue, which leads to increased rates of fetal injury and preterm birth. These studies provide novel insight into the underlying mechanisms of ascending infection.
Collapse
|
41
|
Whidbey C, Burnside K, Martinez RM, Gendrin C, Vornhagen J, Frando A, Harrell MI, McAdams R, Rajagopal L. A Hyperhemolytic/Hyperpigmented Group B Streptococcus Strain with a CovR Mutation Isolated from an Adolescent Patient with Sore Throat. CLINICAL RESEARCH IN INFECTIOUS DISEASES 2015; 2:1018. [PMID: 26913295 PMCID: PMC4762654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Group B Streptococci (GBS) are ß-hemolytic, gram-positive bacteria that are typically associated with infections in human newborns or immunocompromised adults. However, mutation in the two-component regulator CovR/S relieves repression of hemolysin, potentially increasing virulence of GBS. We report the isolation of hyperhemolytic/hyperpigmented GBS strain from an adolescent patient who presented to the University of Washington clinic with symptoms of sore throat. While the patient also tested positive for mononucleosis, a GBS strain with increased hemolysis was isolated from the throat swab obtained from the patient. As hyperhemolytic/hyperpigmented GBS strains are typically associated with mutations in the regulator CovR/CovS, we sequenced the covR/S loci in the clinical isolate. An adenine to cytosine mutation resulting in a change in amino acid coding sequence from glutamine at position 120 to proline in CovR (Q120P) was identified. Introduction of the Q120P amino acid substitution in a CovR complementation plasmid abolished complementation of a ΔcovR mutant derived from the wild type GBS serotype Ia strain A909; these results confirm that the hyperhemolysis observed in the clinical isolate is due to the Q120P substitution in CovR. Antibiotic was prescribed and the patient's symptoms resolved without reported complications. This study represents the first report of the isolation of a hyperhemolytic/hyperpigmented GBS strain due to a covR/S mutation from an adolescent patient with persistent sore throat who was also diagnosed with mononucleosis. The isolation of GBS CovR/S mutants indicates their presence in settings of co-infections and includes adolescents.
Collapse
Affiliation(s)
- Christopher Whidbey
- Department of Pediatrics, University of Washington School of Medicine, USA
- Department of Global Health, University of Washington School of Public Health, USA
| | - Kellie Burnside
- Department of Pediatrics, University of Washington School of Medicine, USA
| | - Raquel M. Martinez
- Department of Laboratory Medicine, University of Washington School of Medicine, USA
| | | | - Jay Vornhagen
- Department of Pediatrics, University of Washington School of Medicine, USA
- Department of Global Health, University of Washington School of Public Health, USA
| | - Andrew Frando
- Department of Global Health, University of Washington School of Public Health, USA
| | | | - Ryan McAdams
- Department of Pediatrics, University of Washington School of Medicine, USA
| | - Lakshmi Rajagopal
- Seattle Children's Research Institute, USA
- Department of Global Health, University of Washington School of Public Health, USA
| |
Collapse
|