1
|
Alexander NG, Cutts WD, Hooven TA, Kim BJ. Mechanisms and Manifestations of Group B Streptococcus Meningitis in Newborns. J Pediatric Infect Dis Soc 2025; 14:piae103. [PMID: 39927629 PMCID: PMC11808573 DOI: 10.1093/jpids/piae103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/02/2024] [Indexed: 02/11/2025]
Abstract
Group B Streptococcus (GBS; Streptococcus agalactiae) is a gram-positive colonizer of the healthy intestinal and genitourinary microbiota. During and shortly after birth, neonates and infants can be opportunistically infected leading to sepsis, pneumonia, or meningitis among other illnesses. GBS is the leading cause of neonatal meningitis globally, and while prophylactic treatments have been successful for reducing early-onset disease, no decrease in the incidence of late-onset disease has occurred and no vaccine is currently available. In this review, we describe GBS both from a clinical and molecular standpoint. We first describe the history of GBS perinatal disease and its clinical presentation and treatment, as well as patient outcomes. We then present recently discovered GBS interactions at the blood-brain barrier that contribute to disease and inflammatory responses, and efforts to develop a broadly effective GBS vaccine.
Collapse
Affiliation(s)
- Natalie G Alexander
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - William D Cutts
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Thomas A Hooven
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Richard King Mellon Institute for Pediatric Research, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Brandon J Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa, Alabama, USA
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, Alabama, USA
| |
Collapse
|
2
|
Chen Y, Qu J, Wang S, Tang M, Liao S, Liu Y, Li L, Wu B. Streptococcus agalactiae infection of a reconstructed human respiratory epithelium revealed infection and host response characteristics by different serotypes. Microb Pathog 2025; 199:107243. [PMID: 39708978 DOI: 10.1016/j.micpath.2024.107243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Streptococcus agalactiae poses a significant threat to neonatal health, causing morbidity and mortality when transmitted from the maternal vagina to the newborn's respiratory tract. Among its various strains, serotype III is predominant in severe neonatal infections in Asia. However, the mechanisms of pathogenesis and host responses underlying serotype-specific disease outcomes remain poorly understood. METHODS This study employed 2D airway epithelial organoids as a host model to investigate the progression of S. agalactiae respiratory tract infections across four serotypes. RNA and ATAC analyses were used to identify differentially expressed genes and related pathways. RESULTS S. agalactiae infection triggered inflammatory responses in differentiated airway epithelia, particularly increasing the expression of genes linked to innate immunity. Serotype III elicited stronger immune responses compared to other serotypes. The SLC2A3 gene demonstrated upregulated expression and increased chromatin accessibility specific to serotype III infection. CONCLUSIONS Neonates predominantly clear S. agalactiae infections through immune detection and opsonophagocytosis. These findings highlight the importance of patient triage based on serotype variations. Developing therapeutic candidates targeting invasive serotypes to enhance OPK titers may be crucial. Furthermore, serotype III infection may induce apoptosis via Ferroptosis.
Collapse
Affiliation(s)
- Yujia Chen
- Department of Oncology, Shenzhen Guangming District People's Hospital, Shenzhen, Guangdong Province, 518107, China
| | - Jing Qu
- Department of Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong Province, 518055, China
| | - Senyang Wang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100000, China
| | - Mingxing Tang
- Department of Otolaryngology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong Province, 518052, China
| | - Shumin Liao
- Department of Otolaryngology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| | - Yingzi Liu
- Department of Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, 518000, China
| | - Liang Li
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, 518055, China.
| | - Benqing Wu
- Department of Neonatology, Shenzhen Guangming District People's Hospital, Shenzhen, Guangdong Province, 518107, China.
| |
Collapse
|
3
|
Payen S, Giroux MC, Gisch N, Schombel U, Fittipaldi N, Segura M, Gottschalk M. Lipoteichoic acids influence cell shape and bacterial division of Streptococcus suis serotype 2, but play a limited role in the pathogenesis of the infection. Vet Res 2024; 55:34. [PMID: 38504299 PMCID: PMC10953176 DOI: 10.1186/s13567-024-01287-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/01/2024] [Indexed: 03/21/2024] Open
Abstract
Streptococcus suis serotype 2 is a major swine pathogen and a zoonotic agent, causing meningitis in both swine and humans, responsible for substantial economic losses to the swine industry worldwide. The pathogenesis of infection and the role of bacterial cell wall components in virulence have not been fully elucidated. Lipoproteins, peptidoglycan, as well as lipoteichoic acids (LTA) have all been proposed to contribute to virulence. In the present study, the role of the LTA in the pathogenesis of the infection was evaluated through the characterisation of a mutant of the S. suis serotype 2 strain P1/7 lacking the LtaS enzyme, which mediates the polymerization of the LTA poly-glycerolphosphate chain. The ltaS mutant was confirmed to completely lack LTA and displayed significant morphological defects. Although the bacterial growth of this mutant was not affected, further results showed that LTA is involved in maintaining S. suis bacterial fitness. However, its role in the pathogenesis of the infection appears limited. Indeed, LTA presence reduces self-agglutination, biofilm formation and even dendritic cell activation, which are important aspects of the pathogenesis of the infection caused by S. suis. In addition, it does not seem to play a critical role in virulence using a systemic mouse model of infection.
Collapse
Affiliation(s)
- Servane Payen
- Research Group On Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, J2S 2M2, Canada
| | - Marie-Christine Giroux
- Research Group On Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, J2S 2M2, Canada
| | - Nicolas Gisch
- Division of Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Ursula Schombel
- Division of Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Nahuel Fittipaldi
- Research Group On Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, J2S 2M2, Canada
| | - Mariela Segura
- Research Group On Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, J2S 2M2, Canada
| | - Marcelo Gottschalk
- Research Group On Infectious Diseases in Production Animals (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, J2S 2M2, Canada.
| |
Collapse
|
4
|
Payen S, Roy D, Okura M, Segura M, Gottschalk M. Study of the Role of Lipoprotein Maturation Enzymes in the Pathogenesis of the Infection Caused by the Streptococcus suis Serotype 2 Sequence Type 25 North American Prototype Strain. Pathogens 2023; 12:1325. [PMID: 38003790 PMCID: PMC10675726 DOI: 10.3390/pathogens12111325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Streptococcus suis serotype 2 is an important swine bacterial pathogen causing sudden death, septic shock, and meningitis. However, serotype 2 strains are phenotypically and genotypically heterogeneous and composed of a multitude of sequence types (STs) whose distributions greatly vary worldwide. It has been previously shown that the lipoprotein (LPP) maturation enzymes diacylglyceryl transferase (Lgt) and signal peptidase (Lsp) significantly modulate the inflammatory host response and play a differential role in virulence depending on the genetic background of the strain. Differently from Eurasian ST1/ST7 strains, the capsular polysaccharide of a North American S. suis serotype 2 ST25 representative strain only partially masks sub-capsular domains and bacterial wall components. Thus, our hypothesis is that since LPPs would be more surface exposed in ST25 strains than in their ST1 or ST7 counterparts, the maturation enzymes would play a more important role in the pathogenesis of the infection caused by the North American strain. Using isogenic Δlgt and Δlsp mutants derived from the wild-type ST25 strain, our studies suggest that these enzymes do not seem to play a role in the interaction between S. suis and epithelial and endothelial cells, regardless of the genetics background of the strain used. However, a role in the formation of biofilms (also independently of the STs) has been demonstrated. Moreover, the involvement of LPP dendritic cell activation in vitro seems to be somehow more pronounced with the ST25 strain. Finally, the Lgt enzyme seems to play a more important role in the virulence of the ST25 strain. Although some differences between STs could be observed, our original hypothesis that LPPs would be significantly more important in ST25 strains due to a better bacterial surface exposition could not be confirmed.
Collapse
Affiliation(s)
- Servane Payen
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (M.S.)
| | - David Roy
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Masatoshi Okura
- Division of Transboundary Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Kagoshima 891-0105, Japan;
| | - Mariela Segura
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (M.S.)
| | - Marcelo Gottschalk
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP) and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (M.S.)
| |
Collapse
|
5
|
Vancolen S, Ayash T, Allard MJ, Sébire G. Sex-Specific Dysconnective Brain Injuries and Neuropsychiatric Conditions such as Autism Spectrum Disorder Caused by Group B Streptococcus-Induced Chorioamnionitis. Int J Mol Sci 2023; 24:14090. [PMID: 37762401 PMCID: PMC10531534 DOI: 10.3390/ijms241814090] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Global health efforts have increased against infectious diseases, but issues persist with pathogens like Group B Streptococcus (GBS). Preclinical studies have elaborated on the mechanistic process of GBS-induced chorioamnionitis and its impact on the fetal programming of chronic neuropsychiatric diseases. GBS inoculation in rodents demonstrated the following: (i) silent and self-limited placental infection, similar to human chorioamnionitis; (ii) placental expression of chemokines attracting polymorphonuclear (PMN) cells; (iii) in vitro cytokine production; (iv) PMN infiltration in the placenta (histologic hallmark of human chorioamnionitis), linked to neurobehavioral impairments like cerebral palsy and autism spectrum disorders (ASD); (v) upregulation of interleukin-1β (IL-1β) in the placenta and fetal blood, associated with higher ASD risk in humans; (vi) sex-specific effects, with higher IL-1β release and PMN recruitment in male placenta; (vii) male offspring exhibiting ASD-like traits, while female offspring displayed attention deficit and hyperactivity disorder (ADHD)-like traits; (viii) IL-1 and/or NF-kB blockade alleviate placental and fetal inflammation, as well as subsequent neurobehavioral impairments. These findings offer potential therapeutic avenues, including sex-adapted anti-inflammatory treatment (e.g., blocking IL-1; repurposing of FDA-approved IL-1 receptor antagonist (IL-1Ra) treatment). Blocking the IL-1 pathway offers therapeutic potential to alleviate chorioamnionitis-related disabilities, presenting an opportunity for a human phase II RCT that uses IL-1 blockade added to the classic antibiotic treatment of chorioamnionitis.
Collapse
Affiliation(s)
- Seline Vancolen
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada;
- Department of Pediatrics, Research Institute of the McGill University Health Center, Montreal, QC H3G 1Y6, Canada
| | - Taghreed Ayash
- Department of Pediatrics, Research Institute of the McGill University Health Center, Montreal, QC H3G 1Y6, Canada
| | - Marie-Julie Allard
- Department of Pediatrics, Research Institute of the McGill University Health Center, Montreal, QC H3G 1Y6, Canada
| | - Guillaume Sébire
- Department of Pediatrics, Research Institute of the McGill University Health Center, Montreal, QC H3G 1Y6, Canada
| |
Collapse
|
6
|
Domon H, Hirayama S, Isono T, Sasagawa K, Takizawa F, Maekawa T, Yanagihara K, Terao Y. Macrolides Decrease the Proinflammatory Activity of Macrolide-Resistant Streptococcus pneumoniae. Microbiol Spectr 2023; 11:e0014823. [PMID: 37191519 PMCID: PMC10269745 DOI: 10.1128/spectrum.00148-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/25/2023] [Indexed: 05/17/2023] Open
Abstract
Over the past 2 decades, the prevalence of macrolide-resistant Streptococcus pneumoniae (MRSP) has increased considerably, due to widespread macrolide use. Although macrolide usage has been proposed to be associated with treatment failure in patients with pneumococcal diseases, macrolides may be clinically effective for treating these diseases, regardless of the susceptibility of the causative pneumococci to macrolides. As we previously demonstrated that macrolides downregulate the transcription of various genes in MRSP, including the gene encoding the pore-forming toxin pneumolysin, we hypothesized that macrolides affect the proinflammatory activity of MRSP. Using HEK-Blue cell lines, we found that the supernatants from macrolide-treated MRSP cultures induced decreased NF-κB activation in cells expressing Toll-like receptor 2 and nucleotide-binding oligomerization domain 2 compared to the supernatants from untreated MRSP cells, suggesting that macrolides inhibit the release of these ligands from MRSP. Real-time PCR analysis revealed that macrolides significantly downregulated the transcription of various genes encoding peptidoglycan synthesis-, lipoteichoic acid synthesis-, and lipoprotein synthesis-related molecules in MRSP cells. The silkworm larva plasma assay demonstrated that the peptidoglycan concentrations in the supernatants from macrolide-treated MRSP cultures were significantly lower than those from untreated MRSP cultures. Triton X-114 phase separation revealed that lipoprotein expression decreased in macrolide-treated MRSP cells compared to the lipoprotein expression in untreated MRSP cells. Consequently, macrolides may decrease the expression of bacterial ligands of innate immune receptors, resulting in the decreased proinflammatory activity of MRSP. IMPORTANCE To date, the clinical efficacy of macrolides in pneumococcal disease is assumed to be linked to their ability to inhibit the release of pneumolysin. However, our previous study demonstrated that oral administration of macrolides to mice intratracheally infected with macrolide-resistant Streptococcus pneumoniae resulted in decreased levels of pneumolysin and proinflammatory cytokines in bronchoalveolar lavage fluid samples compared to the levels in samples from untreated infected control mice, without affecting the bacterial load in the fluid. This finding suggests that additional mechanisms by which macrolides negatively regulate proinflammatory cytokine production may be involved in their efficacy in vivo. Furthermore, in this study, we demonstrated that macrolides downregulated the transcription of various proinflammatory-component-related genes in S. pneumoniae, which provides an additional explanation for the clinical benefits of macrolides.
Collapse
Affiliation(s)
- Hisanori Domon
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Satoru Hirayama
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Toshihito Isono
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Karin Sasagawa
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Fumio Takizawa
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tomoki Maekawa
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
7
|
Segklia K, Matsas R, Papastefanaki F. Brain Infection by Group B Streptococcus Induces Inflammation and Affects Neurogenesis in the Adult Mouse Hippocampus. Cells 2023; 12:1570. [PMID: 37371040 DOI: 10.3390/cells12121570] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Central nervous system infections caused by pathogens crossing the blood-brain barrier are extremely damaging and trigger cellular alterations and neuroinflammation. Bacterial brain infection, in particular, is a major cause of hippocampal neuronal degeneration. Hippocampal neurogenesis, a continuous multistep process occurring throughout life in the adult brain, could compensate for such neuronal loss. However, the high rates of cognitive and other sequelae from bacterial meningitis/encephalitis suggest that endogenous repair mechanisms might be severely affected. In the current study, we used Group B Streptococcus (GBS) strain NEM316, to establish an adult mouse model of brain infection and determine its impact on adult neurogenesis. Experimental encephalitis elicited neurological deficits and death, induced inflammation, and affected neurogenesis in the dentate gyrus of the adult hippocampus by suppressing the proliferation of progenitor cells and the generation of newborn neurons. These effects were specifically associated with hippocampal neurogenesis while subventricular zone neurogenesis was not affected. Overall, our data provide new insights regarding the effect of GBS infection on adult brain neurogenesis.
Collapse
Affiliation(s)
- Katerina Segklia
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Neurobiology Department, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Neurobiology Department, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Florentia Papastefanaki
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Neurobiology Department, Hellenic Pasteur Institute, 11521 Athens, Greece
| |
Collapse
|
8
|
Barks JD, Liu Y, Dopp IA, Silverstein FS. Azithromycin reduces inflammation-amplified hypoxic-ischemic brain injury in neonatal rats. Pediatr Res 2022; 92:415-423. [PMID: 34625655 PMCID: PMC8989723 DOI: 10.1038/s41390-021-01747-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/07/2021] [Accepted: 06/16/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Systemic inflammation amplifies neonatal hypoxic-ischemic (HI) brain injury. Azithromycin (AZ), an antibiotic with anti-inflammatory properties, improves sensorimotor function and reduces tissue damage after neonatal rat HI brain injury. The objective of this study was to determine if AZ is neuroprotective in two neonatal rat models of inflammation-amplified HI brain injury. DESIGN/METHODS Seven-day-old (P7) rats received injections of toll-like receptor agonists lipopolysaccharide (LPS) or Pam3Cys-Ser-(Lys)4 (PAM) prior to right carotid ligation followed by 50 min (LPS + HI) or 60 min (PAM + HI) in 8% oxygen. Outcomes included contralateral forelimb function (forepaw placing; grip strength), survival, %Intact right hemisphere (brain damage), and a composite score incorporating these measures. We compared postnatal day 35 outcomes in controls and groups treated with three or five AZ doses. Then, we compared P21 outcomes when the first (of five) AZ doses were administered 1, 2, or 4 h after HI. RESULTS In both LPS + HI and PAM + HI models, AZ improved sensorimotor function, survival, brain tissue preservation, and composite scores. Benefits increased with five- vs. three-dose AZ and declined with longer initiation delay. CONCLUSIONS Perinatal systemic infection is a common comorbidity of neonatal asphyxia brain injury and contributes to adverse outcomes. These data support further evaluation of AZ as a candidate treatment for neonatal neuroprotection. IMPACT AZ treatment decreases sensorimotor impairment and severity of brain injury, and improves survival, after inflammation-amplified HI brain injury, and this can be achieved even with a 2 h delay in initiation. This neuroprotective benefit is seen in models of inflammation priming by both Gram-negative and Gram-positive infections. This extends our previous findings that AZ treatment is neuroprotective after HI brain injury in neonatal rats.
Collapse
Affiliation(s)
- John D.E. Barks
- Department of Pediatrics, University of Michigan Medical School, The University of Michigan, Ann Arbor, MI
| | - Yiqing Liu
- Department of Pediatrics, University of Michigan Medical School, The University of Michigan, Ann Arbor, MI
| | - Ian A. Dopp
- Department of Pediatrics, University of Michigan Medical School, The University of Michigan, Ann Arbor, MI
| | - Faye S. Silverstein
- Department of Pediatrics, University of Michigan Medical School, The University of Michigan, Ann Arbor, MI,Department of Neurology, University of Michigan Medical School, The University of Michigan, Ann Arbor, MI
| |
Collapse
|
9
|
Kurian NK, Modi D. Mechanisms of group B Streptococcus-mediated preterm birth: lessons learnt from animal models. REPRODUCTION AND FERTILITY 2022; 3:R109-R120. [PMID: 35794927 PMCID: PMC9254271 DOI: 10.1530/raf-21-0105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 11/22/2022] Open
Abstract
Group B Streptococcus (GBS) is an opportunistic pathogenic bacterium which upon colonization in the female reproductive tract can cause preterm births, fetal injury, and demise. Several determinants for GBS pathogenesis have been explored so far through the studies using animal models ranging from mice to non-human primates. The results from these experimental data have identified outer membrane vesicles, β-hemolysin, hyaluronidase, and Cas9 of GBS as major virulence factors leading to preterm births. Most of these factors drive inflammation through activation of NLRP3 and elevated production of IL1-β. However, the absence of one of the factors from the pathogen reduces but does not completely abolish the pathogenesis of GBS suggesting the involvement of more than one factor in causing preterm birth. This makes further exploration of other virulence factors of GBS pathogenesis important in gaining an insight into the mechanistic basis of GBS-mediated preterm births. Lay summary Group B Streptococcus (GBS) is a pathogenic bacteria whose infection in the reproductive tract during pregnancy can cause premature delivery. This bacterial infection is one of the major causes of death of mother and baby during pregnancy, and the bacteria is prevalent in all parts of the world. This makes the research on GBS so important and many of the mechanisms behind GBS infection during pregnancy still remain unexplored. In this review, we have outlined how various animal models contributed in finding the mechanism of GBS pathogenesis. The review also focuses on compiling various virulence factors which makes GBS pathogenic in the vulnerable. Understanding the mechanisms of infection by GBS will be crucial in developing drugs and vaccines to protect against the harmful effects of the bacteria.
Collapse
Affiliation(s)
- Noble K Kurian
- Department of Microbiology, Atmiya University, Rajkot, Gujarat, India
| | - Deepak Modi
- Molecular and Cellular Biology Laboratory, ICMR-National Institute for Research in Reproductive Health and Child Health (NIRRCH), Indian Council of Medical Research (ICMR), Mumbai, India
| |
Collapse
|
10
|
Ederer KU, Holzinger JM, Maier KT, Zeller L, Werner M, Toelge M, Gessner A, Bülow S. A Polymorphism of Bactericidal/Permeability-Increasing Protein Affects Its Neutralization Efficiency towards Lipopolysaccharide. Int J Mol Sci 2022; 23:ijms23031324. [PMID: 35163248 PMCID: PMC8836039 DOI: 10.3390/ijms23031324] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/05/2023] Open
Abstract
Gram-negative sepsis driven by lipopolysaccharide (LPS) has detrimental outcomes, especially in neonates. The neutrophil-derived bactericidal/permeability-increasing protein (BPI) potently neutralizes LPS. Interestingly, polymorphism of the BPI gene at position 645 (rs4358188) corresponds to a favorable survival rate of these patients in the presence of at least one allele 645 A as opposed to 645 G. When we exploited the existing X-ray crystal structure, the corresponding amino acid at position 216 was revealed as surface exposed and proximal to the lipid-binding pocket in the N-terminal domain of BPI. Our further analysis predicted a shift in surface electrostatics by a positively charged lysine (BPI216K) exchanging a negatively charged glutamic acid (BPI216E). To investigate differences in interaction with LPS, we expressed both BPI variants recombinantly. The amino acid exchange neither affected affinity towards LPS nor altered bactericidal activity. However, when stimulating human peripheral blood mononuclear cells, BPI216K exhibited a superior LPS-neutralizing capacity (IC50 12.0 ± 2.5 pM) as compared to BPI216E (IC50 152.9 ± 113.4 pM, p = 0.0081) in respect to IL-6 secretion. In conclusion, we provide a functional correlate to a favorable outcome of sepsis in the presence of BPI216K.
Collapse
|
11
|
Ayash TA, Vancolen SY, Segura M, Allard MJ, Sebire G. Protective Effects of Interleukin-1 Blockade on Group B Streptococcus-Induced Chorioamnionitis and Subsequent Neurobehavioral Impairments of the Offspring. Front Endocrinol (Lausanne) 2022; 13:833121. [PMID: 35846278 PMCID: PMC9283950 DOI: 10.3389/fendo.2022.833121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/26/2022] [Indexed: 11/24/2022] Open
Abstract
Group B Streptococcus (GBS) is one of the most common bacteria isolated in human chorioamnionitis. Placental infection due to GBS is a major risk factor for fetal organ injuries, preterm birth, perinatal morbidity and mortality, and life-long multiorgan morbidities. Preclinical and clinical studies have shown that GBS-induced infection drives polymorphonuclear (PMN) cell infiltration within the placenta, the hallmark of human chorioamnionitis. In preclinical and clinical studies, the upregulation of interleukin(IL)-1β in the placenta and maternal/fetal blood was associated with a high risk of neurodevelopmental impairments in the progeny. We hypothesized that targeted IL-1 blockade administered to the dam alleviates GBS-induced chorioamnionitis and the downstream fetal inflammatory response syndrome (FIRS). IL-1 receptor antagonist (IL-1Ra) improved the gestational weight gain of GBS-infected dams and did not worsen the infectious manifestations. IL-1Ra reduced the IL-1β titer in the maternal sera of GBS-infected dams. IL-1Ra decreased the levels of IL-1β, IL-6, chemokine (C-X-C motif) ligand 1 (CXCL1), and polymorphonuclear (PMN) infiltration in GBS-infected placenta. IL-1Ra treatment reduced the IL-1β titer in the fetal sera of GBS-exposed fetuses. IL-1 blockade also alleviated GBS-induced FIRS and subsequent neurobehavioral impairments of the offspring without worsening the outcome of GBS infection. Altogether, these results showed that IL-1 plays a key role in the physiopathology of live GBS-induced chorioamnionitis and consequent neurobehavioral impairments.
Collapse
Affiliation(s)
| | | | - Mariela Segura
- Faculty of Veterinary Medicine, Université de Montreal, St-Hyacinthe, QC, Canada
| | | | - Guillaume Sebire
- Department of Pediatrics, McGill University, Montreal, QC, Canada
- *Correspondence: Guillaume Sebire,
| |
Collapse
|
12
|
Payen S, Roy D, Boa A, Okura M, Auger JP, Segura M, Gottschalk M. Role of Maturation of Lipoproteins in the Pathogenesis of the Infection Caused by Streptococcus suis Serotype 2. Microorganisms 2021; 9:microorganisms9112386. [PMID: 34835511 PMCID: PMC8621357 DOI: 10.3390/microorganisms9112386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/03/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022] Open
Abstract
Streptococcus suis serotype 2 is an important porcine bacterial pathogen associated with multiple pathologies in piglets. Bacterial lipoproteins (LPPs) have been described as playing important roles in the pathogenesis of the infection of other Gram-positive bacteria as adhesins, pro-inflammatory cell activators and/or virulence factors. In the current study, we aimed to evaluate the role of the prolipoprotein diacylglyceryl transferase (Lgt) and lipoprotein signal peptidase (Lsp) enzymes, which are responsible for LPP maturation, on the pathogenesis of the infection caused by two different sequence types (STs) of S. suis serotype 2 strains (virulent ST1 and highly virulent ST7). Through the use of isogenic Δlgt, Δlsp and double Δlgt/Δlsp mutants, it was shown that lack of these enzymes did not influence S. suis adhesion/invasion to porcine respiratory epithelial cells. However, in the absence of the Lsp and/or Lgt, a significant reduction in the capacity of S. suis to activate phagocytic cells and induce pro-inflammatory mediators (in vitro and in vivo) was observed. In general, results obtained with the double mutant did not differ in comparison to single mutants, indicating lack of an additive effect. Finally, our data suggest that these enzymes play a differential role in virulence, depending on the genetic background of the strain and being more important for the highly virulent ST7 strain.
Collapse
Affiliation(s)
- Servane Payen
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (D.R.); (A.B.); (J.-P.A.); (M.S.)
| | - David Roy
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (D.R.); (A.B.); (J.-P.A.); (M.S.)
| | - Anaïs Boa
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (D.R.); (A.B.); (J.-P.A.); (M.S.)
| | - Masatoshi Okura
- Division of Bacterial and Parasitic Disease, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba 305-0856, Japan;
| | - Jean-Philippe Auger
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (D.R.); (A.B.); (J.-P.A.); (M.S.)
| | - Mariela Segura
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (D.R.); (A.B.); (J.-P.A.); (M.S.)
| | - Marcelo Gottschalk
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (S.P.); (D.R.); (A.B.); (J.-P.A.); (M.S.)
- Correspondence:
| |
Collapse
|
13
|
Redline RW. Placental pathology: Pathways leading to or associated with perinatal brain injury in experimental neurology, special issue: Placental mediated mechanisms of perinatal brain injury. Exp Neurol 2021; 347:113917. [PMID: 34748755 DOI: 10.1016/j.expneurol.2021.113917] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/25/2021] [Accepted: 11/02/2021] [Indexed: 11/04/2022]
Abstract
Perinatal brain injury is a multifactorial process. In utero placental physiology plays a major role in neuroprotection and the normal development of the fetal central nervous system. Advances in placental pathology have clarified several specific mechanisms of injury and the histologic lesions most strongly associated with them. This review provides an updated summary of the relevant placental anatomy and physiology, the specific placental pathways leading to brain injury, the revised Amsterdam classification system for placental pathology, and the known associations of specific placental lesions with subtypes of adverse neurologic outcomes.
Collapse
Affiliation(s)
- Raymond W Redline
- Case Western Reserve University School of Medicine, Departments of Pathology and Reproductive Biology, University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, Cleveland, OH 44106, United States of America.
| |
Collapse
|
14
|
Abhilasha KV, Sumanth MS, Thyagarajan A, Sahu RP, Kemparaju K, Marathe GK. Reversible cross-tolerance to platelet-activating factor signaling by bacterial toxins. Platelets 2021; 32:960-967. [PMID: 32835559 DOI: 10.1080/09537104.2020.1810652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Bacterial toxins signaling through Toll-like receptors (TLRs) are implicated in the pathogenesis of many inflammatory diseases. Among the toxins, lipopolysaccharide (LPS) exerts its action via TLR-4 while lipoteichoic acid (LTA) and bacterial lipoproteins such as Braun lipoprotein (BLP) or its synthetic analogue Pam3CSK4 act through TLR-2. Part of the TLR mediated pathogenicity is believed to stem from endogenously biosynthesized platelet-activating factor (PAF)- a potent inflammatory phospholipid acting through PAF-receptor (PAF-R). However, the role of PAF in inflammatory diseases like endotoxemia is controversial. In order to test the direct contribution of PAF in TLR-mediated pathogenicity, we intraperitoneally injected PAF to Wistar albino mice in the presence or absence of bacterial toxins. Intraperitoneal injection of PAF (5 μg/mouse) causes sudden death of mice, that can be delayed by simultaneously or pre-treating the animals with high doses of bacterial toxins- a phenomenon known as endotoxin cross-tolerance. The bacterial toxins- induced tolerance to PAF can be reversed by increasing the concentration of PAF suggesting the reversibility of cross-tolerance. We did similar experiments using human platelets that express both canonical PAF-R and TLRs. Although bacterial toxins did not induce human platelet aggregation, they inhibited PAF-induced platelet aggregation in a reversible manner. Using rabbit platelets that are ultrasensitive to PAF, we found bacterial toxins (LPS and LTA) and Pam3CSK4 causing rabbit platelet aggregation via PAF-R dependent way. The physical interaction of PAF-R and bacterial toxins is also demonstrated in a human epidermal cell line having stable PAF-R expression. Thus, we suggest the possibility of direct physical interaction of bacterial toxins with PAF-R leading to cross-tolerance.
Collapse
Affiliation(s)
| | | | - Anita Thyagarajan
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Ravi Prakash Sahu
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Kempaiah Kemparaju
- Department of Studies in Biochemistry, University of Mysore, Mysuru, India.,Department of Studies in Molecular Biology, University of Mysore, Mysuru, India
| | - Gopal Kedihithlu Marathe
- Department of Studies in Biochemistry, University of Mysore, Mysuru, India.,Department of Studies in Molecular Biology, University of Mysore, Mysuru, India
| |
Collapse
|
15
|
Parra-Izquierdo I, Lakshmanan HHS, Melrose AR, Pang J, Zheng TJ, Jordan KR, Reitsma SE, McCarty OJT, Aslan JE. The Toll-Like Receptor 2 Ligand Pam2CSK4 Activates Platelet Nuclear Factor-κB and Bruton's Tyrosine Kinase Signaling to Promote Platelet-Endothelial Cell Interactions. Front Immunol 2021; 12:729951. [PMID: 34527000 PMCID: PMC8435771 DOI: 10.3389/fimmu.2021.729951] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/12/2021] [Indexed: 12/24/2022] Open
Abstract
Circulating platelets establish a variety of immunological programs and orchestrate inflammatory responses at the endothelium. Platelets express the innate immunity family of Toll-like receptors (TLRs). While TLR2/TLR1 ligands are known to activate platelets, the effects of TLR2/TLR6 ligands on platelet function remain unclear. Here, we aim to determine whether the TLR2/TLR6 agonists Pam2CSK4 and FSL-1 activate human platelets. In addition, human umbilical vein endothelial cells (HUVECs) and platelets were co-cultured to analyze the role of platelet TLR2/TLR6 on inflammation and adhesion to endothelial cells. Pam2CSK4, but not FSL-1, induced platelet granule secretion and integrin αIIbβ3 activation in a concentration-dependent manner. Moreover, Pam2CSK4 promoted platelet aggregation and increased platelet adhesion to collagen-coated surfaces. Mechanistic studies with blocking antibodies and pharmacologic inhibitors demonstrated that the TLR2/Nuclear factor-κB axis, Bruton’s-tyrosine kinase, and a secondary ADP feedback loop are involved in Pam2CSK4-induced platelet functional responses. Interestingly, Pam2CSK4 showed cooperation with immunoreceptor tyrosine-based activation motif (ITAM)-mediated signaling to enhance platelet activation. Finally, the presence of platelets increased inflammatory responses in HUVECs treated with Pam2CSK4, and platelets challenged with Pam2CSK4 showed increased adhesion to HUVECs under static and physiologically relevant flow conditions. Herein, we define a functional role for platelet TLR2-mediated signaling, which may represent a druggable target to dampen excessive platelet activation in thrombo-inflammatory diseases.
Collapse
Affiliation(s)
- Iván Parra-Izquierdo
- Knight Cardiovascular Institute and Division of Cardiology, School of Medicine, Oregon Health & Science University, Portland, OR, United States.,Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Hari Hara Sudhan Lakshmanan
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Alexander R Melrose
- Knight Cardiovascular Institute and Division of Cardiology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Jiaqing Pang
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Tony J Zheng
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Kelley R Jordan
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Stéphanie E Reitsma
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Owen J T McCarty
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, United States.,Division of Hematology and Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Joseph E Aslan
- Knight Cardiovascular Institute and Division of Cardiology, School of Medicine, Oregon Health & Science University, Portland, OR, United States.,Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, United States.,Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
16
|
Venkataranganayaka Abhilasha K, Kedihithlu Marathe G. Bacterial lipoproteins in sepsis. Immunobiology 2021; 226:152128. [PMID: 34488139 DOI: 10.1016/j.imbio.2021.152128] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 07/09/2021] [Accepted: 08/10/2021] [Indexed: 01/05/2023]
Abstract
Bacterial lipoproteins are membrane proteins derived from both gram-negative and gram-positive bacteria. They seem to have diverse functions not only on bacterial growth, but also play an important role in host's virulence. Bacterial lipoproteins exert their action on host immune cells via TLR2/1 or TLR2/6. Therefore, bacterial lipoproteins also need to be considered while addressing bacterial pathogenicity besides classical bacterial endotoxin like LPS and other microbial associated molecular patterns such as LTA, and peptidoglycans. In this mini-review, we provide an overview of general bacterial lipoprotein biosynthesis and the need to understand the lipoprotein-mediated pathogenicity in diseases like sepsis.
Collapse
Affiliation(s)
- Kandahalli Venkataranganayaka Abhilasha
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru 570006, Karnataka, India; Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Gopal Kedihithlu Marathe
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru 570006, Karnataka, India; Department of Studies in Molecular Biology, University of Mysore, Manasagangothri, Mysuru 570006, Karnataka, India.
| |
Collapse
|
17
|
Rajpoot S, Wary KK, Ibbott R, Liu D, Saqib U, Thurston TLM, Baig MS. TIRAP in the Mechanism of Inflammation. Front Immunol 2021; 12:697588. [PMID: 34305934 PMCID: PMC8297548 DOI: 10.3389/fimmu.2021.697588] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/23/2021] [Indexed: 12/15/2022] Open
Abstract
The Toll-interleukin-1 Receptor (TIR) domain-containing adaptor protein (TIRAP) represents a key intracellular signalling molecule regulating diverse immune responses. Its capacity to function as an adaptor molecule has been widely investigated in relation to Toll-like Receptor (TLR)-mediated innate immune signalling. Since the discovery of TIRAP in 2001, initial studies were mainly focused on its role as an adaptor protein that couples Myeloid differentiation factor 88 (MyD88) with TLRs, to activate MyD88-dependent TLRs signalling. Subsequent studies delineated TIRAP’s role as a transducer of signalling events through its interaction with non-TLR signalling mediators. Indeed, the ability of TIRAP to interact with an array of intracellular signalling mediators suggests its central role in various immune responses. Therefore, continued studies that elucidate the molecular basis of various TIRAP-protein interactions and how they affect the signalling magnitude, should provide key information on the inflammatory disease mechanisms. This review summarizes the TIRAP recruitment to activated receptors and discusses the mechanism of interactions in relation to the signalling that precede acute and chronic inflammatory diseases. Furthermore, we highlighted the significance of TIRAP-TIR domain containing binding sites for several intracellular inflammatory signalling molecules. Collectively, we discuss the importance of the TIR domain in TIRAP as a key interface involved in protein interactions which could hence serve as a therapeutic target to dampen the extent of acute and chronic inflammatory conditions.
Collapse
Affiliation(s)
- Sajjan Rajpoot
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Kishore K Wary
- Department of Pharmacology and Regenerative Medicine, The University of Illinois at Chicago, Chicago, IL, United States
| | - Rachel Ibbott
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Dongfang Liu
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, United States.,School of Graduate Studies, Rutgers Biomedical and Health Sciences, Newark, NJ, United States.,Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
| | - Uzma Saqib
- Discipline of Chemistry, Indian Institute of Technology Indore (IITI), Indore, India
| | - Teresa L M Thurston
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| |
Collapse
|
18
|
Craig-Mueller N, Hammad R, Elling R, Alzubi J, Timm B, Kolter J, Knelangen N, Bednarski C, Gläser B, Ammann S, Ivics Z, Fischer J, Speckmann C, Schwarz K, Lachmann N, Ehl S, Moritz T, Henneke P, Cathomen T. Modeling MyD88 Deficiency In Vitro Provides New Insights in Its Function. Front Immunol 2021; 11:608802. [PMID: 33424861 PMCID: PMC7786022 DOI: 10.3389/fimmu.2020.608802] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/18/2020] [Indexed: 11/24/2022] Open
Abstract
Inherited defects in MyD88 and IRAK4, two regulators in Toll-like receptor (TLR) signaling, are clinically highly relevant, but still incompletely understood. MyD88- and IRAK4-deficient patients are exceedingly susceptible to a narrow spectrum of pathogens, with ∼50% lethality in the first years of life. To better understand the underlying molecular and cellular characteristics that determine disease progression, we aimed at modeling the cellular response to pathogens in vitro. To this end, we determined the immunophenotype of monocytes and macrophages derived from MyD88- and IRAK4-deficient patients. We recognized that macrophages derived from both patients were particularly poorly activated by streptococci, indicating that both signaling intermediates are essential for the immune response to facultative pathogens. To characterize this defect in more detail, we generated induced pluripotent stem cells (iPSCs) of fibroblasts derived from an MyD88-deficient patient. The underlying genetic defect was corrected using Sleeping Beauty transposon vectors encoding either the long (L) or the short (S) MYD88 isoform, respectively. Macrophages derived from these iPSC lines (iMacs) expressed typical macrophage markers, stably produced either MyD88 isoform, and showed robust phagocytic activity. Notably, iMacs expressing MyD88-L, but not MyD88-S, exhibited similar responses to external stimuli, including cytokine release patterns, as compared to genetically normal iMacs. Thus, the two MyD88 isoforms assume distinct functions in signaling. In conclusion, iPSC technology, in combination with efficient myeloid differentiation protocols, provides a valuable and inexhaustible source of macrophages, which can be used for disease modeling. Moreover, iPSC-derived macrophages may eventually aid in stabilizing MyD88-deficient patients during pyogenic infections.
Collapse
Affiliation(s)
- Nils Craig-Mueller
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Freiburg, Germany.,MD Program, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ruba Hammad
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Freiburg, Germany.,PhD Program, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Roland Elling
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Freiburg, Germany.,Institute for Immunodeficiency, Medical Center-University of Freiburg, Freiburg, Germany.,Center for Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Freiburg, Germany
| | - Jamal Alzubi
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Freiburg, Germany
| | - Barbara Timm
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Freiburg, Germany
| | - Julia Kolter
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Freiburg, Germany.,Institute for Immunodeficiency, Medical Center-University of Freiburg, Freiburg, Germany
| | - Nele Knelangen
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Freiburg, Germany.,Institute for Immunodeficiency, Medical Center-University of Freiburg, Freiburg, Germany
| | - Christien Bednarski
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Freiburg, Germany
| | - Birgitta Gläser
- Institute of Human Genetics, Medical Center-University of Freiburg, Freiburg, Germany
| | - Sandra Ammann
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Freiburg, Germany.,Institute for Immunodeficiency, Medical Center-University of Freiburg, Freiburg, Germany
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul-Ehrlich Institute, Langen, Germany
| | - Judith Fischer
- Institute of Human Genetics, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carsten Speckmann
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Freiburg, Germany.,Center for Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus Schwarz
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service Baden-Württemberg-Hessen, and Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Nico Lachmann
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster for Regenerative and Translational Medicine, Hannover, Germany
| | - Stephan Ehl
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Freiburg, Germany.,Institute for Immunodeficiency, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thomas Moritz
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster for Regenerative and Translational Medicine, Hannover, Germany
| | - Philipp Henneke
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Freiburg, Germany.,Institute for Immunodeficiency, Medical Center-University of Freiburg, Freiburg, Germany.,Center for Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
19
|
Benmimoun B, Papastefanaki F, Périchon B, Segklia K, Roby N, Miriagou V, Schmitt C, Dramsi S, Matsas R, Spéder P. An original infection model identifies host lipoprotein import as a route for blood-brain barrier crossing. Nat Commun 2020; 11:6106. [PMID: 33257684 PMCID: PMC7704634 DOI: 10.1038/s41467-020-19826-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
Pathogens able to cross the blood-brain barrier (BBB) induce long-term neurological sequelae and death. Understanding how neurotropic pathogens bypass this strong physiological barrier is a prerequisite to devise therapeutic strategies. Here we propose an innovative model of infection in the developing Drosophila brain, combining whole brain explants with in vivo systemic infection. We find that several mammalian pathogens are able to cross the Drosophila BBB, including Group B Streptococcus (GBS). Amongst GBS surface components, lipoproteins, and in particular the B leucine-rich Blr, are important for BBB crossing and virulence in Drosophila. Further, we identify (V)LDL receptor LpR2, expressed in the BBB, as a host receptor for Blr, allowing GBS translocation through endocytosis. Finally, we show that Blr is required for BBB crossing and pathogenicity in a murine model of infection. Our results demonstrate the potential of Drosophila for studying BBB crossing by pathogens and identify a new mechanism by which pathogens exploit the machinery of host barriers to generate brain infection.
Collapse
Affiliation(s)
- Billel Benmimoun
- Institut Pasteur, Brain Plasticity in Response to the Environment, CNRS, UMR3738, Paris, France
| | - Florentia Papastefanaki
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Bruno Périchon
- Unité de Biologie des Bactéries Pathogènes à Gram-positif, Institut Pasteur, CNRS, UMR 2001, Paris, France
| | - Katerina Segklia
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Nicolas Roby
- Institut Pasteur, Brain Plasticity in Response to the Environment, CNRS, UMR3738, Paris, France
| | - Vivi Miriagou
- Laboratory of Bacteriology, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Christine Schmitt
- Ultrastructure UTechS Ultrastructural Bioimaging Platform, Institut Pasteur, Paris, France
| | - Shaynoor Dramsi
- Unité de Biologie des Bactéries Pathogènes à Gram-positif, Institut Pasteur, CNRS, UMR 2001, Paris, France
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Pauline Spéder
- Institut Pasteur, Brain Plasticity in Response to the Environment, CNRS, UMR3738, Paris, France.
| |
Collapse
|
20
|
Sikora AE, Gomez C, Le Van A, Baarda BI, Darnell S, Martinez FG, Zielke RA, Bonventre JA, Jerse AE. A novel gonorrhea vaccine composed of MetQ lipoprotein formulated with CpG shortens experimental murine infection. Vaccine 2020; 38:8175-8184. [PMID: 33162204 DOI: 10.1016/j.vaccine.2020.10.077] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/15/2020] [Accepted: 10/23/2020] [Indexed: 11/18/2022]
Abstract
Bacterial surface lipoproteins are emerging as attractive vaccine candidates due to their biological importance and the feasibility of their large-scale production for vaccine manufacturing. The global prevalence of gonorrhea, resistance to antibiotics, and serious consequences to reproductive and neonatal health necessitate development of effective vaccines. Reverse vaccinology identified the surface-displayed L-methionine binding lipoprotein MetQ (NGO2139) and its homolog GNA1946 (NMB1946) as gonococcal and meningococcal vaccine candidates, respectively. Here, we assessed the suitability of MetQ for inclusion in a gonorrhea vaccine by examining MetQ conservation, its function inNeisseria gonorrhoeae (Ng) pathogenesis, and its ability to induce protective immune responses using a female murine model of lower genital tract infection. In-depth bioinformatics, phylogenetics and mapping the most prevalent Ng polymorphic amino acids to the GNA1946 crystal structure revealed remarkable MetQ conservation: ~97% Ng isolates worldwide possess a single MetQ variant. Mice immunized with rMetQ-CpG (n = 40), a vaccine containing a tag-free version of MetQ formulated with CpG, exhibited robust, antigen-specific antibody responses in serum and at the vaginal mucosae including IgA. Consistent with the activity of CpG as a Th1-stimulating adjuvant, the serum IgG1/IgG2a ratio of 0.38 suggested a Th1 bias. Combined data from two independent challenge experiments demonstrated that rMetQ-CpG immunized mice cleared infection faster than control animals (vehicle, p < 0.0001; CpG, p = 0.002) and had lower Ng burden (vehicle, p = 0.03; CpG, p < 0.0001). We conclude rMetQ-CpG induces a protective immune response that accelerates bacterial clearance from the murine lower genital tract and represents an attractive component of a gonorrhea subunit vaccine.
Collapse
Affiliation(s)
- Aleksandra E Sikora
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97330, United States; Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States.
| | - Carolina Gomez
- Department of Microbiology and Immunology, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | - Adriana Le Van
- Department of Microbiology and Immunology, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | - Benjamin I Baarda
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97330, United States
| | - Stephen Darnell
- Department of Microbiology and Immunology, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | - Fabian G Martinez
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97330, United States
| | - Ryszard A Zielke
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97330, United States
| | - Josephine A Bonventre
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97330, United States
| | - Ann E Jerse
- Department of Microbiology and Immunology, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States.
| |
Collapse
|
21
|
Lee IC, van Swam II, Boeren S, Vervoort J, Meijerink M, Taverne N, Starrenburg M, Bron PA, Kleerebezem M. Lipoproteins Contribute to the Anti-inflammatory Capacity of Lactobacillus plantarum WCFS1. Front Microbiol 2020; 11:1822. [PMID: 32849426 PMCID: PMC7403179 DOI: 10.3389/fmicb.2020.01822] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022] Open
Abstract
Bacterial lipoproteins are well-recognized microorganism-associated molecular patterns, which interact with Toll-like receptor (TLR) 2, an important pattern recognition receptor of the host innate immune system. Lipoproteins are conjugated with two- or three-acyl chains (di- or tri-acyl), which is essential for appropriate anchoring in the cell membrane as well as for the interaction with TLR2. Lipoproteins have mostly been studied in pathogens and have established roles in various biological processes, such as nutrient import, cell wall cross-linking and remodeling, and host-cell interaction. By contrast, information on the role of lipoproteins in the physiology and host interaction of probiotic bacteria is scarce. By deletion of lgt, encoding prolipoprotein diacylglyceryl transferase, responsible for lipidation of lipoprotein precursors, we investigated the roles of the collective group of lipoproteins in the physiology of the probiotic model strain Lactobacillus plantarum WCFS1 using proteomic analysis of secreted proteins. To investigate the consequences of the lgt mutation in host-cell interaction, the capacity of mutant and wild-type bacteria to stimulate TLR2 signaling and inflammatory responses was compared using (reporter-) cell-based models. These experiments exemplified the critical contribution of the acyl chains of lipoproteins in immunomodulation. To the best of our knowledge, this is the first study that investigated collective lipoprotein functions in a model strain for probiotic lactobacilli, and we show that the lipoproteins in L. plantarum WCFS1 are critical drivers of anti-inflammatory host responses toward this strain.
Collapse
Affiliation(s)
- I-Chiao Lee
- Host-Microbe Interactomics Group, Wageningen University & Research, Wageningen, Netherlands.,TiFN Food & Nutrition, Wageningen, Netherlands.,NIZO Food Research, Ede, Netherlands
| | - Iris I van Swam
- TiFN Food & Nutrition, Wageningen, Netherlands.,NIZO Food Research, Ede, Netherlands
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University & Research, Wageningen, Netherlands
| | - Jacques Vervoort
- Laboratory of Biochemistry, Wageningen University & Research, Wageningen, Netherlands
| | - Marjolein Meijerink
- Host-Microbe Interactomics Group, Wageningen University & Research, Wageningen, Netherlands
| | - Nico Taverne
- Host-Microbe Interactomics Group, Wageningen University & Research, Wageningen, Netherlands.,TiFN Food & Nutrition, Wageningen, Netherlands
| | | | - Peter A Bron
- TiFN Food & Nutrition, Wageningen, Netherlands.,NIZO Food Research, Ede, Netherlands
| | - Michiel Kleerebezem
- Host-Microbe Interactomics Group, Wageningen University & Research, Wageningen, Netherlands.,TiFN Food & Nutrition, Wageningen, Netherlands
| |
Collapse
|
22
|
Diaz-Dinamarca DA, Manzo RA, Soto DA, Avendaño-Valenzuela MJ, Bastias DN, Soto PI, Escobar DF, Vasquez-Saez V, Carrión F, Pizarro-Ortega MS, Wilson CAM, Berrios J, Kalergis AM, Vasquez AE. Surface Immunogenic Protein of Streptococcus Group B is an Agonist of Toll-Like Receptors 2 and 4 and a Potential Immune Adjuvant. Vaccines (Basel) 2020; 8:vaccines8010029. [PMID: 31963234 PMCID: PMC7157747 DOI: 10.3390/vaccines8010029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/20/2019] [Accepted: 01/02/2020] [Indexed: 12/19/2022] Open
Abstract
Vaccine-induced protection against pathogens, especially subunit-based vaccines, are related to antigen properties but mainly in their ability to stimulate the immune system by the use of an adjuvant. Modern vaccines are formulated with a high level of antigen purity, where an efficient adjuvant is necessary. In this context, the use of protein Toll-Like Receptor (TLR) agonists as vaccine adjuvants has been highlighted because of their optimal immunogenicity and minimal toxicity. The Surface Immunogenic Protein (SIP) from Group B Streptococcus (GBS) has gained importance as a new potential protein-based vaccine. Recently, we reported that recombinant SIP (rSIP) expressed by E. coli and purified by High Performance Liquid Chromatography (HPLC) alone induces a protective humoral immune response. In this study, we present the immunomodulatory properties of rSIP as a protein-based adjuvant, as an agonist of TLR. To this end, we showed that C57BL/6 bone marrow-derived dendritic cells pulsed by rSIP resulted in enhanced CD40, CD80, CD86, and Major Histocompatibility Complex (MHC) class II as well as increased secretion proinflammatory cytokines Interleukin (IL)-6, Interferon (IFN)-γ, Tumor Necrosis Factor (TNF)-α, and IL-10. Next, we investigated the in vivo effect of rSIP in the absence or presence of ovalbumin (OVA) on antigen-specific antibody secretion in C57BL/6 mice. Immunization with rSIP plus OVA showed that anti-OVA IgG2a and IgG1a increased significantly compared with OVA alone in C57BL/6 mice. Also, the immunization of rSIP plus OVA generates increased serum cytokines levels characterized by IL-12p70, IL-10, IL-4, and IFN-γ. Interestingly, we observed that rSIP stimulate Toll Like Receptor (TLR)2 and TLR4, individually expressed by Human embryonic kidney (HEK) 293-derived TLR reporter cells. These findings suggest that rSIP is a new potential protein TLR agonist adjuvant and may be employed in the development of new vaccines.
Collapse
Affiliation(s)
- Diego A. Diaz-Dinamarca
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.S.P.-O.); (A.M.K.)
| | - Ricardo A. Manzo
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
| | - Daniel A. Soto
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
| | - María José Avendaño-Valenzuela
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.S.P.-O.); (A.M.K.)
| | - Diego N. Bastias
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.S.P.-O.); (A.M.K.)
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Santo Tomas, Santiago 8320000, Chile
| | - Paulina I. Soto
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
| | - Daniel F. Escobar
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
| | - Valeria Vasquez-Saez
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
| | - Flavio Carrión
- Programa de Inmunología Traslacional, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 8320000, Chile;
| | - Magdalena S. Pizarro-Ortega
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.S.P.-O.); (A.M.K.)
| | - Christian A. M. Wilson
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 8320000, Chile;
| | - Julio Berrios
- Escuela de Ingeniería en Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340000, Chile;
| | - Alexis M. Kalergis
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (M.S.P.-O.); (A.M.K.)
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| | - Abel E. Vasquez
- Seccion de Biotecnologia, Instituto de Salud Publica de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (R.A.M.); (D.A.S.); (M.J.A.-V.); (D.N.B.); (P.I.S.); (D.F.E.); (V.V.-S.)
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Santo Tomas, Santiago 8320000, Chile
- Facultad de Ciencia, Universidad San Sebastián, Providencia, Santiago 8320000, Chile
- Correspondence: ; Tel.: +56-2-2575-5513
| |
Collapse
|
23
|
Lentini G, Famà A, Biondo C, Mohammadi N, Galbo R, Mancuso G, Iannello D, Zummo S, Giardina M, De Gaetano GV, Teti G, Beninati C, Midiri A. Neutrophils Enhance Their Own Influx to Sites of Bacterial Infection via Endosomal TLR-Dependent Cxcl2 Production. THE JOURNAL OF IMMUNOLOGY 2019; 204:660-670. [PMID: 31852751 DOI: 10.4049/jimmunol.1901039] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/19/2019] [Indexed: 12/27/2022]
Abstract
The influx of neutrophils to infection sites is a fundamental step in host defenses against the frequent human pathogen group B Streptococcus (GBS) and other extracellular bacteria. Using a mouse model of GBS-induced peritonitis, we show in this study that the chemokines Cxcl1 and Cxcl2 play distinctive roles in enhancing the recruitment and the antibacterial activities of neutrophils in a manner that is linked to differences in the cellular sources of these mediators. Cell depletion experiments demonstrated that neutrophils make a significant contribution to the in vivo production of Cxcl2 but not Cxcl1. In vitro, neutrophils responded weakly to LPS but released high levels of Cxcl2 after stimulation with GBS or other bacteria. Neutrophil-derived Cxcl2 acted in an autocrinous manner to increase its own production and to enhance antibacterial activities, including the release of oxygen radicals. In both neutrophils and macrophages, the production of Cxcl1/2 largely required the presence of functional UNC93B1, a chaperone protein involved in signaling by endosomal TLRs. Moreover, the phenotype of UNC93B1-defective phagocytes could be recapitulated by the simultaneous absence of TLR7, 9, and 13 but not by the absence of individual TLRs. Collectively, our data show that neutrophils recognize Gram-positive and Gram-negative bacteria by means of multiple phagosomal TLRs, resulting in de novo synthesis of Cxcl2, amplification of neutrophil recruitment, and potentiation of their antibacterial activities. These data may be useful to devise alternative therapeutic strategies aimed at enhancing the recruitment and the functional activities of polymorphonuclear leukocytes during infections caused by antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Germana Lentini
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Agata Famà
- Charybdis Vaccines Srl, 98125 Messina, Italy
| | - Carmelo Biondo
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Nastaran Mohammadi
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Roberta Galbo
- Department of Chemical, Biological, Pharmaceutical Sciences and Environmental Sciences, University of Messina, 98166 Messina, Italy; and
| | - Giuseppe Mancuso
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Daniela Iannello
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Sebastiana Zummo
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Miriam Giardina
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | | | | | - Concetta Beninati
- Department of Human Pathology, University of Messina, 98125 Messina, Italy.,Scylla Biotech SRL, 98125 Messina, Italy
| | - Angelina Midiri
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| |
Collapse
|
24
|
Feuerstein R, Gres V, Elias Perdigó N, Baasch S, Freudenhammer M, Elling R, Henneke P. Macrophages Are a Potent Source of Streptococcus-Induced IFN-β. THE JOURNAL OF IMMUNOLOGY 2019; 203:3416-3426. [PMID: 31732532 DOI: 10.4049/jimmunol.1900542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 10/14/2019] [Indexed: 11/19/2022]
Abstract
IFN-β essentially modulates the host response against mucocutaneous colonizers and potential pathogens, such as group B Streptococcus (GBS). It has been reported that the dominant signaling cascade driving IFN-β in macrophages (MΦ) in streptococcal infection is the cGAS-STING pathway, whereas conventional dendritic cells (DC) exploit endosomal recognition by intracellular TLRs. In this study, we revisited this issue by precisely monitoring the phenotypic dynamics in mixed mouse MΦ/DC cultures with GM-CSF, which requires snapshot definition of cellular identities. We identified four mononuclear phagocyte populations, of which two were transcriptionally and morphologically distinct MΦ-DC-like subsets, and two were transitional types. Notably, GBS induced a TLR7-dependent IFN-β signal only in MΦ-like but not in DC-like cells. IFN-β induction did not require live bacteria (i.e., the formation of cytolytic toxins), which are essential for IFN-β induction via cGAS-STING. In contrast to IFN-β, GBS induced TNF-α independently of TLR7. Subsequent to the interaction with streptococci, MΦ changed their immunophenotype and gained some typical DC markers and DC-like morphology. In summary, we identify IFN-β formation as part of the antistreptococcal repertoire of GM-CSF differentiated MΦ in vitro and in vivo and delineate their plasticity.
Collapse
Affiliation(s)
- Reinhild Feuerstein
- Institute for Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; and
| | - Vitka Gres
- Institute for Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; and
| | - Núria Elias Perdigó
- Institute for Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; and
| | - Sebastian Baasch
- Institute for Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; and
| | - Mirjam Freudenhammer
- Institute for Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; and.,Center for Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Roland Elling
- Institute for Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; and.,Center for Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Philipp Henneke
- Institute for Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; and .,Center for Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
25
|
Hikita N, Cho Y, Tachibana D, Hamazaki T, Koyama M, Tokuhara D. Cell surface antigens of neonatal monocytes are selectively impaired in basal expression, but hyperresponsive to lipopolysaccharide and zymosan. J Reprod Immunol 2019; 136:102614. [PMID: 31606697 DOI: 10.1016/j.jri.2019.102614] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/22/2019] [Accepted: 09/17/2019] [Indexed: 11/27/2022]
Abstract
Toll-like receptors (TLRs) are important components of the innate immune system, but how neonatal TLR-mediated immune responses differ from those of adults is unknown. We aimed to clarify the TLR-mediated expression profiles of cell surface antigens related to antigen presentation in neonates. CD14-positive monocytes were isolated from human cord blood and adult peripheral blood and then stimulated with lipopolysaccharide (LPS; TLR4 agonist) or zymosan (TLR2/6 agonist) or left unstimulated. Expression levels of the surface antigens major histocompatibility (MHC)-class II, CD80, CD86, CD11b, CD11c, CD14, and CD16 were then evaluated by flow cytometry. Cord blood CD14+CD16high monocytes (CBM) showed significantly lower basal levels of MHC-class II, CD80, and CD11b than adult blood CD14+CD16intermediate monocytes (ABM) (P < 0.01, P < 0.001, P < 0.001, respectively). LPS stimulation enhanced expression of MHC class II, CD80, and CD11b significantly more in CBM than in ABM (P < 0.001, P < 0.01, P < 0.01, respectively), resulting in levels that did not differ between CBM and ABM. Zymosan stimulation also enhanced expression of MHC class II, CD86, CD11b, and CD11c significantly more in CBM than in ABM (P < 0.001, P < 0.01, P < 0.001, P < 0.01, respectively), resulting in levels of CD86 and CD11c that did not differ in CBM and ABM. However, MHC class II, CD80, and CD11b remained significantly higher in ABM than in CBM (P < 0.05, P < 0.01, P < 0.05, respectively). These data indicate that CBM and ABM have distinct phenotypes and responses to stimulation.
Collapse
Affiliation(s)
- Norikatsu Hikita
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yuki Cho
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Daisuke Tachibana
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Takashi Hamazaki
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masayasu Koyama
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Daisuke Tokuhara
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan; Department of Medicine, University of California San Diego, San Diego, CA, USA.
| |
Collapse
|
26
|
Sex-specific maternofetal innate immune responses triggered by group B Streptococci. Sci Rep 2019; 9:8587. [PMID: 31197179 PMCID: PMC6565749 DOI: 10.1038/s41598-019-45029-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/29/2019] [Indexed: 01/27/2023] Open
Abstract
Group B Streptococcus (GBS) is one of the most common bacteria isolated in human chorioamnionitis, which is a major risk factor for premature birth and brain injuries. Males are at greater risk than females for developing lifelong neurobehavioural disorders, although the origins of this sex bias remain poorly understood. We previously showed that end-gestational inflammation triggered by GBS led to early neurodevelopmental impairments mainly in the male rat progeny. Identifying key inflammatory players involved in maternofetal immune activation by specific pathogens is critical to develop appropriate novel therapeutic interventions. We aimed to map out the GBS-induced profile of innate immune biomarkers in the maternal-placental-fetal axis, and to compare this immune profile between male and female tissues. We describe here that the GBS-induced immune signalling involved significantly higher levels of interleukin (IL)-1β, cytokine-induced neutrophil chemoattractant-1 (CINC-1/CXCL1) and polymorphonuclear cells (PMNs) infiltration in male compared to female maternofetal tissues. Although male - but not female - fetuses presented increased levels of IL-1β, fetuses from both sexes in-utero exposed to GBS had increased levels of TNF-α in their circulation. Levels of IL-1β detected in fetal sera correlated positively with the levels found in maternal circulation. Here, we report for the first time that the maternofetal innate immune signalling induced by GBS presents a sexually dichotomous profile, with more prominent inflammation in males than females. These sex-specific placental and fetal pro-inflammatory responses are in keeping with the higher susceptibility of the male population for preterm birth, brain injuries and neurodevelopmental disorders such as cerebral palsy and autism spectrum disorders.
Collapse
|
27
|
Parida R. Human MOSPD2: A bacterial Lmb mimicked auto-antigen is involved in immune infertility. J Transl Autoimmun 2019; 1:100002. [PMID: 32743492 PMCID: PMC7388392 DOI: 10.1016/j.jtauto.2019.100002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/16/2019] [Accepted: 05/19/2019] [Indexed: 12/12/2022] Open
Abstract
Autoantibody production is one of the leading factors of immune infertility, an autoimmune disease of the male reproductive system. The potential involvement of MHC-class II derived self-peptides against bacterial proteins in the antisperm antibody (ASA) production has been reported previously. Apparently, Streptococcus agalactiae has been considered as an important pathogen to impart infection-induced infertility in a bacteriospermia associated leukocytospermia (LCS/BS) state. Hence, the present study attempts to confirm S. agalactiae specific Laminin binding protein (Lmb) derived self-peptide ('KDSYTKKAKAFKKEA') namely human Motile Sperm domain-containing protein 2 (MOSPD2) as an auto-antigen in LCS/BS condition. Semen samples were collected from infertile men with LCS/BS (n = 17) and their fertile counterparts (n = 10). Gram-positive bacteria were predominantly identified in the entire 17 LCS samples using culture method followed by 16S rDNA sequencing technique. TLRs 2 and 4 expression used as markers of immune response in spermatozoa and sperm dysfunction were elevated in the LCS/BS spermatozoa as compared to their fertile counterparts. A significant increase in oxidative stress indices i.e., protein carbonylation, lipid peroxidation and acridine orange test (AOT), was also observed in the LCS/BS spermatozoa. Spermatozoa lysate (both auto and heterologous), bacterial lysate (control) and synthesized MOSPD2 self-peptide were used to test their antigenicity against the autoantibodies by rocket immunoelectrophoresis (RIEP) assay. Seminal plasma from LCS/BS patients with S. agalactiae was used as the source of autoantibodies. Spermatozoa and bacteria lysate; and MOSPD2 self-peptide were able to bind autoantibodies in the seminal plasma. Besides, the self-peptide showed a dose dependent increase in the precipitation of antibody. T-cell epitope mapping of 48 Enterococcus faecalis and 91Staphylococcus aureus surface proteins confirmed MOSPD2 as a global auto-antigen. Thus, augmentation of TLR expression in LCS/BS spermatozoa inferred MOSPD2 to be a putative immunogen. Altogether, these findings will delineate the significance of MOSPD2 auto-antigen in a bacteria derived immune infertility condition.
Collapse
Affiliation(s)
- Rajeshwari Parida
- Department of Zoology, Ravenshaw University, Cuttack, 753003, Odisha, India
| |
Collapse
|
28
|
Zhang Q, Chao TC, Patil VS, Qin Y, Tiwari SK, Chiou J, Dobin A, Tsai CM, Li Z, Dang J, Gupta S, Urdahl K, Nizet V, Gingeras TR, Gaulton KJ, Rana TM. The long noncoding RNA ROCKI regulates inflammatory gene expression. EMBO J 2019; 38:e100041. [PMID: 30918008 PMCID: PMC6463213 DOI: 10.15252/embj.2018100041] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 12/15/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) can regulate target gene expression by acting in cis (locally) or in trans (non-locally). Here, we performed genome-wide expression analysis of Toll-like receptor (TLR)-stimulated human macrophages to identify pairs of cis-acting lncRNAs and protein-coding genes involved in innate immunity. A total of 229 gene pairs were identified, many of which were commonly regulated by signaling through multiple TLRs and were involved in the cytokine responses to infection by group B Streptococcus We focused on elucidating the function of one lncRNA, named lnc-MARCKS or ROCKI (Regulator of Cytokines and Inflammation), which was induced by multiple TLR stimuli and acted as a master regulator of inflammatory responses. ROCKI interacted with APEX1 (apurinic/apyrimidinic endodeoxyribonuclease 1) to form a ribonucleoprotein complex at the MARCKS promoter. In turn, ROCKI-APEX1 recruited the histone deacetylase HDAC1, which removed the H3K27ac modification from the promoter, thus reducing MARCKS transcription and subsequent Ca2+ signaling and inflammatory gene expression. Finally, genetic variants affecting ROCKI expression were linked to a reduced risk of certain inflammatory and infectious disease in humans, including inflammatory bowel disease and tuberculosis. Collectively, these data highlight the importance of cis-acting lncRNAs in TLR signaling, innate immunity, and pathophysiological inflammation.
Collapse
Affiliation(s)
- Qiong Zhang
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Ti-Chun Chao
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Veena S Patil
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Yue Qin
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Shashi Kant Tiwari
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Joshua Chiou
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| | | | - Chih-Ming Tsai
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Zhonghan Li
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Jason Dang
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Shagun Gupta
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Kevin Urdahl
- Center for Infectious Disease Research (CIDR), Seattle, WA, USA
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Victor Nizet
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego School of Medicine, La Jolla, CA, USA
| | | | - Kyle J Gaulton
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Tariq M Rana
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
29
|
Wietzorrek G, Drexel M, Trieb M, Santos-Sierra S. Anti-inflammatory activity of small-molecule antagonists of Toll-like receptor 2 (TLR2) in mice. Immunobiology 2019; 224:1-9. [PMID: 30509503 DOI: 10.1016/j.imbio.2018.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/07/2018] [Accepted: 11/12/2018] [Indexed: 12/17/2022]
Abstract
Toll-like receptor 2 (TLR2) is currently investigated as a potential therapeutic target in diseases with underlying inflammation like sepsis and arthritis. We reported the discovery, by virtual screening and biological testing, of eight TLR2 antagonists (AT1-AT8) which showed TLR2-inhibitory activity in human cells (Murgueitio et al., 2014). In this study, we have deepened in the mechanism of action and selectivity (TLR2/1 or TLR2/6) of those compounds in mouse primary cells and in vivo. The antagonists reduced, in a dose-dependent way the TNFα production (e.g. AT5 IC50 7.4 μM) and also reduced the nitric oxide (NO) formation in mouse bone marrow-derived macrophages (BMDM). Treatment of BMDM with the antagonists showed that downstream of TLR2, MAPKs phosphorylation and IkBα degradation was reduced. Notably, in a mouse model of tri-acylated lipopeptide (Pam3CSK4)-induced inflammation, AT5 attenuated the TNFα and IL-6 inflammatory response. Further, the effect of AT5 in the stimulation of BMDM by the endogenous alarmin HMGB1 was investigated. Our results indicate that AT4-AT7 and, particularly AT5 appear as good starting points for the development of inhibitors targeting TLR2 in inflammatory disorders.
Collapse
Affiliation(s)
- G Wietzorrek
- Section of Molecular and Cellular Pharmacology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - M Drexel
- Department of Pharmacology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - M Trieb
- Section of Biochemical Pharmacology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - S Santos-Sierra
- Section of Biochemical Pharmacology, Medical University of Innsbruck, A-6020, Innsbruck, Austria.
| |
Collapse
|
30
|
SslE (YghJ), a Cell-Associated and Secreted Lipoprotein of Neonatal Septicemic Escherichia coli, Induces Toll-Like Receptor 2-Dependent Macrophage Activation and Proinflammation through NF-κB and MAP Kinase Signaling. Infect Immun 2018; 86:IAI.00399-18. [PMID: 29891541 DOI: 10.1128/iai.00399-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
SslE (YghJ), a cell surface-associated and secreted lipoprotein, was identified as a potential vaccine candidate for extraintestinal pathogenic Escherichia coli, providing nearly complete protection from sepsis in a mouse model. We earlier found that SslE from neonatal septicemic E. coli could trigger the secretion of various proinflammatory cytokines in murine macrophages, the signaling pathway of which is still obscure. In this study, we showed that SslE specifically binds to Toll-like receptor 2 (TLR2)/TLR1 heterodimers and recruits downstream adaptors MyD88, TIRAP, and TRAF6. In addition, SslE stimulates nuclear translocation of NF-κB and activates different mitogen-activated protein (MAP) kinase signaling cascades specific to the secretion of each cytokine in murine macrophages, which becomes impaired in TLR2 small interfering RNA (siRNA)-transfected cells and in cells blocked with a monoclonal antibody (MAb) against TLR2, suggesting the involvement of TLR2 in NF-κB and MAP kinase activation and subsequent cytokine secretion. Furthermore, our study is the first to show that SslE can stimulate TLR2-dependent production of other proinflammatory hallmarks, such as reactive nitrogen and oxygen species as well as type 1 chemokines, which contribute to the anti-infection immune response of the host. Also, the overexpression of major histocompatibility complex class II (MHC II) and other costimulatory molecules (CD80 and CD86) in macrophages essentially indicates that SslE promotes macrophage activation and M1 polarization, which are crucial in framing the host's innate immune response to this protein, and hence, SslE could be a potent immunotherapeutic target against E. coli sepsis.
Collapse
|
31
|
Szylberg Ł, Bodnar M, Lebioda A, Krepska P, Kowalewski A, Bręborowicz G, Marszałek A. Differences in the Expression of TLR-2, NOD2, and NF-κB in Placenta Between Twins. Arch Immunol Ther Exp (Warsz) 2018; 66:463-470. [PMID: 29796818 PMCID: PMC6245244 DOI: 10.1007/s00005-018-0514-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 05/08/2018] [Indexed: 12/12/2022]
Abstract
Dizygotic twins share the same type of genetic relationship as non-twin siblings. Whereas monozygotic (MZ) twins are considered to have identical genetic material, they still differ. There is a number of reasons for early MZ twin discordance, including differences in the in utero environment, stochasticity, genetic mosaicism, and epigenetic factors. During gestation, the efficient innate immune system is of utmost importance. Our study was based on immunohistochemical evaluation of the differences in innate immune protein expression (TLR-2, NOD2, and NF-κB) in the 95 placentas between twins. Our study revealed statistical significant differences between diamniotic–dichorionic and monoamniotic–dichorionic twins. Monoamniotic–monochorionic twins exhibited no significant differences in protein expressions. To identify epigenetic factors causing the differences between twins, we made a series of comparisons with clinical data. The study revealed more cases with infections, miscarriages, in vitro fertilization, and premature rupture of membranes within the group with higher differences level of NF-κB, NOD2 and TLR-2 between twins. In case of twin-to-twin transfusion syndrome, there were no significant differences in innate immune protein expressions between twins. These results show that dissimilar genetic material and separate in utero environment promote discordance in innate immune protein expressions between twins. Moreover, additional blood flow between twins may be favorable in life-threatening conditions ensuring similar microenvironment.
Collapse
Affiliation(s)
- Łukasz Szylberg
- Chair and Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland. .,Chair and Department of Oncologic Pathology and Prophylactics, Greater Poland Cancer Center, Poznań University of Medical Sciences and Department of Oncologic Pathology, Poznan, Poland. .,Department of Pathomorphology, Military Clinical Hospital, Bydgoszcz, Poland.
| | - Magdalena Bodnar
- Chair and Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Anna Lebioda
- Department and Clinic of Perinatology and Gynecology, Poznań University of Medical Sciences, Poznan, Poland
| | - Patrycja Krepska
- Department of Obstetrics and Gynecology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Adam Kowalewski
- Chair and Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Grzegorz Bręborowicz
- Department and Clinic of Perinatology and Gynecology, Poznań University of Medical Sciences, Poznan, Poland
| | - Andrzej Marszałek
- Chair and Department of Oncologic Pathology and Prophylactics, Greater Poland Cancer Center, Poznań University of Medical Sciences and Department of Oncologic Pathology, Poznan, Poland
| |
Collapse
|
32
|
Sobocińska J, Roszczenko-Jasińska P, Ciesielska A, Kwiatkowska K. Protein Palmitoylation and Its Role in Bacterial and Viral Infections. Front Immunol 2018; 8:2003. [PMID: 29403483 PMCID: PMC5780409 DOI: 10.3389/fimmu.2017.02003] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/26/2017] [Indexed: 12/11/2022] Open
Abstract
S-palmitoylation is a reversible, enzymatic posttranslational modification of proteins in which palmitoyl chain is attached to a cysteine residue via a thioester linkage. S-palmitoylation determines the functioning of proteins by affecting their association with membranes, compartmentalization in membrane domains, trafficking, and stability. In this review, we focus on S-palmitoylation of proteins, which are crucial for the interactions of pathogenic bacteria and viruses with the host. We discuss the role of palmitoylated proteins in the invasion of host cells by bacteria and viruses, and those involved in the host responses to the infection. We highlight recent data on protein S-palmitoylation in pathogens and their hosts obtained owing to the development of methods based on click chemistry and acyl-biotin exchange allowing proteomic analysis of protein lipidation. The role of the palmitoyl moiety present in bacterial lipopolysaccharide and lipoproteins, contributing to infectivity and affecting recognition of bacteria by innate immune receptors, is also discussed.
Collapse
Affiliation(s)
- Justyna Sobocińska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Paula Roszczenko-Jasińska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Anna Ciesielska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
33
|
Kolter J, Henneke P. Codevelopment of Microbiota and Innate Immunity and the Risk for Group B Streptococcal Disease. Front Immunol 2017; 8:1497. [PMID: 29209311 PMCID: PMC5701622 DOI: 10.3389/fimmu.2017.01497] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/24/2017] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of neonatal late-onset sepsis (LOD), which manifests between the third day and the third month of life, remains poorly understood. Group B Streptococcus (GBS) is the most important cause of LOD in infants without underlying diseases or prematurity and the third most frequent cause of meningitis in the Western world. On the other hand, GBS is a common intestinal colonizer in infants. Accordingly, despite its adaption to the human lower gastrointestinal tract, GBS has retained its potential virulence and its transition from a commensal to a dangerous pathogen is unpredictable in the individual. Several cellular innate immune mechanisms, in particular Toll-like receptors, the inflammasome and the cGAS pathway, are engaged by GBS effectors like nucleic acids. These are likely to impact on the GBS-specific host resistance. Given the long evolution of streptococci as a normal constituent of the human microbiota, the emergence of GBS as the dominant neonatal sepsis cause just about 50 years ago is remarkable. It appears that intensive usage of tetracycline starting in the 1940s has been a selection advantage for the currently dominant GBS clones with superior adhesive and invasive properties. The historical replacement of Group A by Group B streptococci as a leading neonatal pathogen and the higher frequency of other β-hemolytic streptococci in areas with low GBS prevalence suggests the existence of a confined streptococcal niche, where locally competing streptococcal species are subject to environmental and immunological selection pressure. Thus, it seems pivotal to resolve neonatal innate immunity at mucous surfaces and its impact on microbiome composition and quality, i.e., genetic heterogeneity and metabolism, at the microanatomical level. Then, designer pro- and prebiotics, such as attenuated strains of GBS, and oligonucleotide priming of mucosal immunity may unfold their potential and facilitate adaptation of potentially hazardous streptococci as part of a beneficial local microbiome, which is stabilized by mucocutaneous innate immunity.
Collapse
Affiliation(s)
- Julia Kolter
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| |
Collapse
|
34
|
Kim AR, Ahn KB, Kim HY, Seo HS, Kum KY, Yun C, Han SH. Streptococcus gordonii lipoproteins induce IL-8 in human periodontal ligament cells. Mol Immunol 2017; 91:218-224. [DOI: 10.1016/j.molimm.2017.09.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/06/2017] [Accepted: 09/18/2017] [Indexed: 12/21/2022]
|
35
|
Ehrnström B, Beckwith KS, Yurchenko M, Moen SH, Kojen JF, Lentini G, Teti G, Damås JK, Espevik T, Stenvik J. Toll-Like Receptor 8 Is a Major Sensor of Group B Streptococcus But Not Escherichia coli in Human Primary Monocytes and Macrophages. Front Immunol 2017; 8:1243. [PMID: 29042860 PMCID: PMC5632357 DOI: 10.3389/fimmu.2017.01243] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/19/2017] [Indexed: 02/05/2023] Open
Abstract
TLR8 is the major endosomal sensor of degraded RNA in human monocytes and macrophages. It has been implicated in the sensing of viruses and more recently also bacteria. We previously identified a TLR8-IFN regulatory factor 5 (IRF5) signaling pathway that mediates IFNβ and interleukin-12 (IL-12) induction by Staphylococcus aureus and is antagonized by TLR2. The relative importance of TLR8 for the sensing of various bacterial species is however still unclear. We here compared the role of TLR8 and IRF5 for the sensing of Group B Streptococcus (GBS), S. aureus, and Escherichia coli in human primary monocytes and monocyte-derived macrophages (MDM). GBS induced stronger IFNβ and TNF production as well as IRF5 nuclear translocation compared to S. aureus grown to the stationary phase, while S. aureus in exponential growth appeared similarly potent to GBS. Cytokine induction in primary human monocytes by GBS was not dependent on hemolysins, and induction of IFNβ and IL-12 as well as IRF5 activation were reduced with TLR2 ligand costimulation. Heat inactivation of GBS reduced IRF5 and NF-kB translocation, while only the viable E. coli activated IRF5. The attenuated stimulation correlated with loss of bacterial RNA integrity. The E. coli-induced IRF5 translocation was not inhibited by TLR2 costimulation, suggesting that IRF5 was activated via a TLR8-independent mechanism. Gene silencing of MDM using siRNA revealed that GBS-induced IFNβ, IL-12-p35, and TNF production was dependent on TLR8 and IRF5. In contrast, cytokine induction by E. coli was TLR8 independent but still partly dependent on IRF5. We conclude that TLR8-IRF5 signaling is more important for the sensing of GBS than for stationary grown S. aureus in human primary monocytes and MDM, likely due to reduced resistance of GBS to phagosomal degradation and to a lower production of TLR2 activating lipoproteins. TLR8 does not sense viable E. coli, while IRF5 still contributes to E. coli-induced cytokine production, possibly via a cytosolic nucleic acid sensing mechanism.
Collapse
Affiliation(s)
- Birgitta Ehrnström
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Infectious Diseases, Clinic of Medicine, St. Olavs Hospital HF, Trondheim University Hospital, Trondheim, Norway
| | - Kai Sandvold Beckwith
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mariia Yurchenko
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Siv Helen Moen
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - June Frengen Kojen
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Germana Lentini
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Giuseppe Teti
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Jan Kristian Damås
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Infectious Diseases, Clinic of Medicine, St. Olavs Hospital HF, Trondheim University Hospital, Trondheim, Norway
| | - Terje Espevik
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jørgen Stenvik
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Infectious Diseases, Clinic of Medicine, St. Olavs Hospital HF, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
36
|
Pombinho R, Camejo A, Vieira A, Reis O, Carvalho F, Almeida MT, Pinheiro JC, Sousa S, Cabanes D. Listeria monocytogenes CadC Regulates Cadmium Efflux and Fine-tunes Lipoprotein Localization to Escape the Host Immune Response and Promote Infection. J Infect Dis 2017; 215:1468-1479. [PMID: 28368435 DOI: 10.1093/infdis/jix118] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 03/06/2017] [Indexed: 11/12/2022] Open
Abstract
Listeria monocytogenes is a major intracellular human foodborne bacterial pathogen. We previously revealed L. monocytogenes cadC as highly expressed during mouse infection. Here we show that L. monocytogenes CadC is a sequence-specific, DNA-binding and cadmium-dependent regulator of CadA, an efflux pump conferring cadmium resistance. CadC but not CadA is required for L. monocytogenes infection in vivo. Interestingly, CadC also directly represses lspB, a gene encoding a lipoprotein signal peptidase whose expression appears detrimental for infection. lspB overexpression promotes the release of the LpeA lipoprotein to the extracellular medium, inducing tumor necrosis factor α and interleukin 6 expression, thus impairing L. monocytogenes survival in macrophages. We propose that L. monocytogenes uses CadC to repress lspB expression during infection to avoid LpeA exposure to the host immune system, diminishing inflammatory cytokine expression and promoting intramacrophagic survival and virulence. CadC appears as the first metal efflux pump regulator repurposed during infection to fine-tune lipoprotein processing and host responses.
Collapse
Affiliation(s)
- Rita Pombinho
- Instituto de Investigação e Inovação em Saúde and.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, and.,Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Ana Camejo
- Instituto de Investigação e Inovação em Saúde and.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, and.,Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Ana Vieira
- Instituto de Investigação e Inovação em Saúde and.,Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Olga Reis
- Instituto de Investigação e Inovação em Saúde and.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, and.,Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Filipe Carvalho
- Instituto de Investigação e Inovação em Saúde and.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, and.,Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Maria Teresa Almeida
- Instituto de Investigação e Inovação em Saúde and.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, and.,Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Jorge Campos Pinheiro
- Instituto de Investigação e Inovação em Saúde and.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, and.,Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Sandra Sousa
- Instituto de Investigação e Inovação em Saúde and.,Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Didier Cabanes
- Instituto de Investigação e Inovação em Saúde and.,Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Porto, Portugal
| |
Collapse
|
37
|
Sachdev U, Lotze MT. Perpetual change: autophagy, the endothelium, and response to vascular injury. J Leukoc Biol 2017; 102:221-235. [PMID: 28626046 PMCID: PMC6608075 DOI: 10.1189/jlb.3ru1116-484rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/15/2022] Open
Abstract
Current studies of vascular health, aging, and autophagy emphasize how the endothelium adapts to stress and contributes to disease. The endothelium is far from an inert barrier to blood-borne cells, pathogens, and chemical signals; rather, it actively translates circulating mediators into tissue responses, changing rapidly in response to physiologic stressors. Macroautophagy-the cellular ingestion of effete organelles and protein aggregates to provide anabolic substrates to fuel bioenergetics in times of stress-plays an important role in endothelial cell homeostasis, vascular remodeling, and disease. These roles include regulating vascular tone, sustaining or limiting cell survival, and contributing to the development of atherosclerosis secondary to infection, inflammation, and angiogenesis. Autophagy modulates these critical functions of the endothelium in a dynamic and perpetual response to tissue and intravascular cues.
Collapse
Affiliation(s)
- Ulka Sachdev
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
38
|
Upadhyay K, Park JE, Yoon TW, Halder P, Kim YI, Metcalfe V, Talati AJ, English BK, Yi AK. Group B Streptococci Induce Proinflammatory Responses via a Protein Kinase D1-Dependent Pathway. THE JOURNAL OF IMMUNOLOGY 2017; 198:4448-4457. [PMID: 28461572 DOI: 10.4049/jimmunol.1601089] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 04/02/2017] [Indexed: 12/31/2022]
Abstract
Group B streptococci (GBS) are one of the leading causes of life-threatening illness in neonates. Proinflammatory responses to GBS mediated through host innate immune receptors play a critical role in the disease manifestation. However, the mechanisms involved in proinflammatory responses against GBS, as well as the contribution of signaling modulators involved in host immune defense, have not been fully elucidated. In the present study, we investigated the role of protein kinase D (PKD)1 in the proinflammatory responses to GBS. We found that both live and antibiotic-killed GBS induce activation of PKD1 through a pathway that is dependent on the TLR signaling adaptor MyD88 and its downstream kinase IL-1R-associated kinase 1, but independent of TNFR-associated factor 6. Our studies using pharmacological PKD inhibitors and PKD1-knockdown macrophages revealed that PKD1 is indispensable for GBS-mediated activation of MAPKs and NF-κB and subsequent expression of proinflammatory mediators. Furthermore, systemic administration of a PKD inhibitor protects d-galactosamine-sensitized mice from shock-mediated death caused by antibiotic-killed GBS. These findings imply that PKD1 plays a critical regulatory role in GBS-induced proinflammatory reactions and sepsis, and inhibition of PKD1 activation together with antibiotic treatment in GBS-infected neonates could be an effective way to control GBS diseases.
Collapse
Affiliation(s)
- Kirtikumar Upadhyay
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN 38163.,Department of Obstetrics and Gynecology, The University of Tennessee Health Science Center, Memphis, TN 38163.,Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103
| | - Jeoung-Eun Park
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN 38163
| | - Tae Won Yoon
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN 38163; and
| | - Priyanka Halder
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN 38163; and
| | - Young-In Kim
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN 38163.,Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103
| | - Victoria Metcalfe
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN 38163; and
| | - Ajay J Talati
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN 38163.,Department of Obstetrics and Gynecology, The University of Tennessee Health Science Center, Memphis, TN 38163.,Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103
| | - B Keith English
- Department of Pediatrics and Human Development, Michigan State University, Lansing, MI 48912
| | - Ae-Kyung Yi
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN 38163; and
| |
Collapse
|
39
|
Yu X, Åvall-Jääskeläinen S, Koort J, Lindholm A, Rintahaka J, von Ossowski I, Palva A, Hynönen U. A Comparative Characterization of Different Host-sourced Lactobacillus ruminis Strains and Their Adhesive, Inhibitory, and Immunomodulating Functions. Front Microbiol 2017; 8:657. [PMID: 28450859 PMCID: PMC5390032 DOI: 10.3389/fmicb.2017.00657] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 03/30/2017] [Indexed: 12/19/2022] Open
Abstract
Lactobacillus ruminis, an autochthonous member of the gastrointestinal microbiota of humans and many animals, is a less characterized but interesting species for many reasons, including its intestinal prevalence and possible positive roles in host–microbe crosstalk. In this study, we isolated a novel L. ruminis strain (GRL 1172) from porcine feces and analyzed its functional characteristics and niche adaptation factors in parallel with those of three other L. ruminis strains (a human isolate, ATCC 25644, and two bovine isolates, ATCC 27780 and ATCC 27781). All the strains adhered to fibronectin, type I collagen, and human colorectal adenocarcinoma cells (HT-29), but poorly to type IV collagen, porcine intestinal epithelial cells (IPEC-1), and human colon adenocarcinoma cells (Caco-2). In competition assays, all the strains were able to inhibit the adhesion of Yersinia enterocolitica and enterotoxigenic Escherichia coli (ETEC, F4+) to fibronectin, type I; collagen, IPEC-1, and Caco-2 cells, and the inhibition rates tended to be higher than in exclusion assays. The culture supernatants of the tested strains inhibited the growth of six selected pathogens to varying extents. The inhibition was solely based on the low pH resulting from acid production during growth. All four L. ruminis strains supported the barrier function maintenance of Caco-2 cells, as shown by the modest increase in trans-epithelial electrical resistance and the prevention of dextran diffusion during co-incubation. However, the strains could not prevent the barrier damage caused by ETEC in the Caco-2 cell model. All the tested strains and their culture supernatants were able to provoke Toll-like receptor (TLR) 2-mediated NF-κB activation and IL-8 production in vitro to varying degrees. The induction of TLR5 signaling revealed that flagella were expressed by all the tested strains, but to different extents. Flagella and pili were observed by electron microscopy on the newly isolated strain GRL 1172.
Collapse
Affiliation(s)
- Xia Yu
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of HelsinkiHelsinki, Finland
| | - Silja Åvall-Jääskeläinen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of HelsinkiHelsinki, Finland
| | - Joanna Koort
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of HelsinkiHelsinki, Finland
| | - Agneta Lindholm
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of HelsinkiHelsinki, Finland
| | - Johanna Rintahaka
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of HelsinkiHelsinki, Finland
| | - Ingemar von Ossowski
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of HelsinkiHelsinki, Finland
| | - Airi Palva
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of HelsinkiHelsinki, Finland
| | - Ulla Hynönen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of HelsinkiHelsinki, Finland
| |
Collapse
|
40
|
Bastos PAD, da Costa JP, Vitorino R. A glimpse into the modulation of post-translational modifications of human-colonizing bacteria. J Proteomics 2016; 152:254-275. [PMID: 27888141 DOI: 10.1016/j.jprot.2016.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/22/2016] [Accepted: 11/07/2016] [Indexed: 12/19/2022]
Abstract
Protein post-translational modifications (PTMs) are a key bacterial feature that holds the capability to modulate protein function and responses to environmental cues. Until recently, their role in the regulation of prokaryotic systems has been largely neglected. However, the latest developments in mass spectrometry-based proteomics have allowed an unparalleled identification and quantification of proteins and peptides that undergo PTMs in bacteria, including in species which directly or indirectly affect human health. Herein, we address this issue by carrying out the largest and most comprehensive global pooling and comparison of PTM peptides and proteins from bacterial species performed to date. Data was collected from 91 studies relating to PTM bacterial peptides or proteins identified by mass spectrometry-based methods. The present analysis revealed that there was a considerable overlap between PTMs across species, especially between acetylation and other PTMs, particularly succinylation. Phylogenetically closer species may present more overlapping phosphoproteomes, but environmental triggers also contribute to this proximity. PTMs among bacteria were found to be extremely versatile and diverse, meaning that the same protein may undergo a wide variety of different modifications across several species, but it could also suffer different modifications within the same species.
Collapse
Affiliation(s)
- Paulo André Dias Bastos
- Department of Medical Sciences, Institute for Biomedicine-iBiMED, University of Aveiro, Aveiro, Portugal; Department of Chemistry, University of Aveiro, Portugal
| | | | - Rui Vitorino
- Department of Medical Sciences, Institute for Biomedicine-iBiMED, University of Aveiro, Aveiro, Portugal; Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
41
|
Shahmirzadi SV, Nguyen MT, Götz F. Evaluation of Staphylococcus aureus Lipoproteins: Role in Nutritional Acquisition and Pathogenicity. Front Microbiol 2016; 7:1404. [PMID: 27679612 PMCID: PMC5020093 DOI: 10.3389/fmicb.2016.01404] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/24/2016] [Indexed: 01/22/2023] Open
Abstract
Bacterial lipoproteins (Lpp) represent a major class of membrane proteins. They are distinguished by a lipid moiety at the N-terminus by which they are anchored either in the outer leaflet of the cytoplasmic membrane or, in Gram-negative bacteria, also in the inner leaflet of the outer membrane. In Gram-positive bacteria Lpp significantly contribute to nutrient transport, Toll-like receptor 2 activation and pathogenicity. Here we examine the Lpp of Staphylococcus aureus USA300, as a prototype for a multiple antibiotic resistant and community-acquired pathogen that is rapidly spreading worldwide. The compiled Lpp were grouped according to the postulated function and dissemination of homologs in the genus Staphylococcus and beyond. Based on this evaluation we also point out Lpp as promising vaccine candidates.
Collapse
Affiliation(s)
- Shideh V Shahmirzadi
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen Tübingen, Germany
| | - Minh-Thu Nguyen
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen Tübingen, Germany
| | - Friedrich Götz
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen Tübingen, Germany
| |
Collapse
|
42
|
Mohammadi N, Midiri A, Mancuso G, Patanè F, Venza M, Venza I, Passantino A, Galbo R, Teti G, Beninati C, Biondo C. Neutrophils Directly Recognize Group B Streptococci and Contribute to Interleukin-1β Production during Infection. PLoS One 2016; 11:e0160249. [PMID: 27509078 PMCID: PMC4980021 DOI: 10.1371/journal.pone.0160249] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/15/2016] [Indexed: 11/29/2022] Open
Abstract
Previous studies have shown that the pro-inflammatory cytokine IL-1β has a crucial role in host defenses against group B streptococcus (GBS), a frequent human pathogen, by recruiting neutrophils to infection sites. We examined here the cell types and mechanisms involved in IL-1β production during infection. Using a GBS-induced peritonitis model in mice, we first found that a large proportion of exudate cells contain intracellular IL-1β by immunofluorescence. Of the IL-1β positive cells, 82 and 7% were neutrophils and macrophages, respectively, suggesting that the former cell type might significantly contribute to IL-1β production. Accordingly, depletion of neutrophils with anti-Ly6G antibodies resulted in a significant reduction in the levels of IL-1β, but not of TNF-α or IL-6. We next found that neutrophils are capable of releasing mature IL-1β and TNF-α directly in response to in vitro stimulation with GBS. The production of pro-IL-1β and TNF-α in these cells required the Toll-like receptor (TLR) adaptor MyD88 and the chaperone protein UNC93B1, which is involved in mobilization of a subfamily of TLRs to the endosomes. Moreover, pro-IL-1β processing and IL-1β release was triggered by GBS hemolysin and required components of the canonical inflammasome, including caspase-1, ASC and NLRP3. Collectively our findings indicate that neutrophils make a significant contribution to IL-1β production during GBS infection, thereby amplifying their own recruitment. These cells directly recognize GBS by means of endosomal TLRs and cytosolic sensors, leading to activation of the caspase-1 inflammasome.
Collapse
Affiliation(s)
- Nastaran Mohammadi
- Department of Clinical and Experimental Medicine, University of Messina, 98125, Messina, Italy
| | - Angelina Midiri
- Department of Human Pathology, University of Messina, 98125, Messina, Italy
| | - Giuseppe Mancuso
- Department of Human Pathology, University of Messina, 98125, Messina, Italy
| | - Francesco Patanè
- Department of Human Pathology, University of Messina, 98125, Messina, Italy
| | - Mario Venza
- Department of Clinical and Experimental Medicine, University of Messina, 98125, Messina, Italy
| | - Isabella Venza
- Department of Clinical and Experimental Medicine, University of Messina, 98125, Messina, Italy
| | | | - Roberta Galbo
- Department of Biological, Chemical and Environmental Sciences, University of Messina, 98125, Messina, Italy
| | - Giuseppe Teti
- Department of Clinical and Experimental Medicine, University of Messina, 98125, Messina, Italy
- Charybdis Vaccines Srl, 98125, Messina, Italy
- * E-mail:
| | - Concetta Beninati
- Department of Human Pathology, University of Messina, 98125, Messina, Italy
- Scylla Biotech Srl, 98125, Messina, Italy
| | - Carmelo Biondo
- Department of Human Pathology, University of Messina, 98125, Messina, Italy
| |
Collapse
|
43
|
Lipoproteins of Gram-Positive Bacteria: Key Players in the Immune Response and Virulence. Microbiol Mol Biol Rev 2016; 80:891-903. [PMID: 27512100 DOI: 10.1128/mmbr.00028-16] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Since the discovery in 1973 of the first of the bacterial lipoproteins (Lpp) in Escherichia coli, Braun's lipoprotein, the ever-increasing number of publications indicates the importance of these proteins. Bacterial Lpp belong to the class of lipid-anchored proteins that in Gram-negative bacteria are anchored in both the cytoplasmic and outer membranes and in Gram-positive bacteria are anchored only in the cytoplasmic membrane. In contrast to the case for Gram-negative bacteria, in Gram-positive bacteria lipoprotein maturation and processing are not vital. Physiologically, Lpp play an important role in nutrient and ion acquisition, allowing particularly pathogenic species to better survive in the host. Bacterial Lpp are recognized by Toll-like receptor 2 (TLR2) of the innate immune system. The important role of Lpp in Gram-positive bacteria, particularly in the phylum Firmicutes, as key players in the immune response and pathogenicity has emerged only in recent years. In this review, we address the role of Lpp in signaling and modulating the immune response, in inflammation, and in pathogenicity. We also address the potential of Lpp as promising vaccine candidates.
Collapse
|
44
|
Yanai S, Tokuhara D, Tachibana D, Saito M, Sakashita Y, Shintaku H, Koyama M. Diabetic pregnancy activates the innate immune response through TLR5 or TLR1/2 on neonatal monocyte. J Reprod Immunol 2016; 117:17-23. [PMID: 27351455 DOI: 10.1016/j.jri.2016.06.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 05/09/2016] [Accepted: 06/20/2016] [Indexed: 11/28/2022]
Abstract
Diabetes mellitus (DM) during pregnancy causes congenital malformation, macrosomia, respiratory distress syndrome, and other abnormalities in neonates, but whether maternal DM affects the neonatal innate immune system is unknown. Therefore we aimed to reveal the influence of DM in pregnancy on the toll-like receptor (TLR)-mediated innate immune response in neonates. Cord blood was collected after full-term vaginal or cesarean delivery and classified into a DM group (n=8) and non-DM (control) group (n=7). Mononuclear cells were harvested from cord blood by using density gradient centrifugation, after which anti-CD14 magnetic beads were used to isolate monocytes from the mononuclear population. After monocytes were cultured with lipopolysaccharide (TLR4 ligand), flagellin (TLR5 ligand), Pam3CSK4 (TLR1/TLR2 ligand), zymosan (TLR2/TLR6 ligand), or macrophage-activating lipopeptide (TLR2/TLR6 ligand) for 12h, the cytokine levels (interleukin [IL]-8, IL-6, IL-1β, IL-10, tumor necrosis factor alpha and IL-12) in the culture supernatants were measured. Compared with the control group, the DM group had higher concentrations of IL-8 (P=0.01) and tumor necrosis factor alpha (P=0.02) after monocyte cultures were stimulated with Pam3CSK4 and higher concentrations of IL-8 (P=0.01) after flagellin treatment. In contrast, stimulation with lipopolysaccharide, zymosan, or macrophage-activating lipopeptide did not lead to any difference in cytokine profiles between the two groups. These data indicate that maternal DM induces excessive inflammatory activation in neonates via a TLR5- or TLR1/2-mediated innate immune response.
Collapse
Affiliation(s)
- Sakika Yanai
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Daisuke Tokuhara
- Department of Pediatrics, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan.
| | - Daisuke Tachibana
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Mika Saito
- Department of Pediatrics, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Yuko Sakashita
- Department of Pediatrics, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Haruo Shintaku
- Department of Pediatrics, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Masayasu Koyama
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| |
Collapse
|
45
|
Allard MJ, Bergeron JD, Baharnoori M, Srivastava LK, Fortier LC, Poyart C, Sébire G. A sexually dichotomous, autistic-like phenotype is induced by Group B Streptococcus maternofetal immune activation. Autism Res 2016; 10:233-245. [PMID: 27220806 DOI: 10.1002/aur.1647] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 03/30/2016] [Accepted: 04/27/2016] [Indexed: 11/09/2022]
Abstract
Group B Streptococcus (GBS) is a commensal bacterium present in the lower genital tract of 15-30% of healthy pregnant women. GBS is the leading cause of chorioamnionitis and cerebral injuries in newborns, occurring most often in the absence of maternofetal pathogen translocation. Despite GBS being the most frequent bacterium colonizing pregnant women, no preclinical studies have investigated the impact of end-gestational maternal GBS exposure on the offspring's brain development and its behavioral correlates. Our hypothesis is that GBS-induced gestational infection/inflammation has a deleterious neurodevelopmental impact on uninfected offspring. Our goal was to study the impact of maternal GBS infection on the placental and neurodevelopmental features in the offspring using a new preclinical rat model. GBS-exposed placentas exhibited chorioamnionitis characterized by the presence of Gram-positive cocci and polymorphonuclear cells, with the latter being significantly more prominent in the labyrinth of male offspring. GBS-exposed male offspring had reduced thickness of periventricular white matter. In addition, they exhibited autistic-like behaviors, such as abnormal social interaction and communication, impaired processing of sensory information and hyperactivity. Overall, these data show for the first time that gestational exposure to GBS plays an important role in the generation of neurodevelopmental abnormalities reminiscent of human autism spectrum disorders (ASD). These results provide new evidence in favor of the role of a common and modifiable infectious/inflammatory environmental factor in human ASD pathophysiology. Autism Res 2017, 10: 233-245. © 2016 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Marie-Julie Allard
- Department of Pediatrics, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Julie D Bergeron
- Département de Pédiatrie, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Moogeh Baharnoori
- Division of Neurology, University of Toronto, Toronto, Ontario, Canada
| | - Lalit K Srivastava
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | - Louis-Charles Fortier
- Département de Microbiologie et d'Infectiologie, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Claire Poyart
- INSERM U 1016, Institut Cochin, Team Barriers and Pathogens; DHU Risques et grossesse, Hôpitaux Universitaire Paris Centre, Université Paris Descartes, Paris, France
| | - Guillaume Sébire
- Department of Pediatrics, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.,Département de Pédiatrie, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
46
|
Kolter J, Feuerstein R, Spoeri E, Gharun K, Elling R, Trieu-Cuot P, Goldmann T, Waskow C, Chen ZJ, Kirschning CJ, Deshmukh SD, Henneke P. Streptococci Engage TLR13 on Myeloid Cells in a Site-Specific Fashion. THE JOURNAL OF IMMUNOLOGY 2016; 196:2733-41. [PMID: 26873993 DOI: 10.4049/jimmunol.1501014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 01/14/2016] [Indexed: 12/11/2022]
Abstract
Streptococci are common human colonizers with a species-specific mucocutaneous distribution. At the same time, they are among the most important and most virulent invasive bacterial pathogens. Thus, site-specific cellular innate immunity, which is predominantly executed by resident and invading myeloid cells, has to be adapted with respect to streptococcal sensing, handling, and response. In this article, we show that TLR13 is the critical mouse macrophage (MΦ) receptor in the response to group B Streptococcus, both in bone marrow-derived MΦs and in mature tissue MΦs, such as those residing in the lamina propria of the colon and the dermis, as well as in microglia. In contrast, TLR13 and its chaperone UNC-93B are dispensable for a potent cytokine response of blood monocytes to group B Streptococcus, although monocytes serve as the key progenitors of intestinal and dermal MΦs. Furthermore, a specific role for TLR13 with respect to MΦ function is supported by the response to staphylococci, where TLR13 and UNC-93B limit the cytokine response in bone marrow-derived MΦs and microglia, but not in dermal MΦs. In summary, TLR13 is a critical and site-specific receptor in the single MΦ response to β-hemolytic streptococci.
Collapse
Affiliation(s)
- Julia Kolter
- Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Reinhild Feuerstein
- Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Evelyne Spoeri
- Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Kourosh Gharun
- Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Roland Elling
- Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Patrick Trieu-Cuot
- Institute Pasteur, Unité de Biologie des Bactéries Pathogènes à Gram-Positif, CNRS ERL3526, 75724 Paris Cedex 15, France
| | - Tobias Goldmann
- Institute of Neuropathology, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Claudia Waskow
- Regeneration in Hematopoiesis and Animal Models of Hematopoiesis, Faculty of Medicine, Technical University, 01307 Dresden, Germany
| | - Zhijian J Chen
- Southwestern Medical School, University of Texas, Dallas, TX 75390
| | - Carsten J Kirschning
- Institute of Medical Microbiology, Medical Center, University of Essen, 45147 Essen, Germany
| | - Sachin D Deshmukh
- Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Center for Sepsis Control and Care, Medical Center, University of Jena, 07747 Jena, Germany; and
| | - Philipp Henneke
- Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Center for Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
47
|
LaRock CN, Nizet V. Inflammasome/IL-1β Responses to Streptococcal Pathogens. Front Immunol 2015; 6:518. [PMID: 26500655 PMCID: PMC4597127 DOI: 10.3389/fimmu.2015.00518] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/24/2015] [Indexed: 02/06/2023] Open
Abstract
Inflammation mediated by the inflammasome and the cytokine IL-1β are some of the earliest and most important alarms to infection. These pathways are responsive to the virulence factors that pathogens use to subvert immune processes, and thus are typically activated only by microbes with potential to cause severe disease. Among the most serious human infections are those caused by the pathogenic streptococci, in part because these species numerous strategies for immune evasion. Since the virulence factor armament of each pathogen is unique, the role of IL-1β and the pathways leading to its activation varies for each infection. This review summarizes the role of IL-1β during infections caused by streptococcal pathogens, with emphasis on emergent mechanisms and concepts countering paradigms determined for other organisms.
Collapse
Affiliation(s)
- Christopher N LaRock
- Department of Pediatrics, University of California San Diego , La Jolla, CA , USA
| | - Victor Nizet
- Department of Pediatrics, University of California San Diego , La Jolla, CA , USA ; Skaggs School of Medicine and Pharmaceutical Sciences, University of California San Diego , La Jolla, CA , USA
| |
Collapse
|
48
|
Theilacker C, Diederich AK, Otto A, Sava IG, Wobser D, Bao Y, Hese K, Broszat M, Henneke P, Becher D, Huebner J. Enterococcus faecalis Glycolipids Modulate Lipoprotein-Content of the Bacterial Cell Membrane and Host Immune Response. PLoS One 2015; 10:e0132949. [PMID: 26172831 PMCID: PMC4501811 DOI: 10.1371/journal.pone.0132949] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 06/21/2015] [Indexed: 12/25/2022] Open
Abstract
In this study, we investigated the impact of the cell membrane composition of E. faecalis on its recognition by the host immune system. To this end, we employed an E. faecalis deletion mutant (ΔbgsA) that does not synthesize the major cell membrane glycolipid diglycosyl-diacylglycerol (DGlcDAG). Proteomic analysis revealed that 13 of a total of 21 upregulated surface-associated proteins of E. faecalis ΔbgsA were lipoproteins. This led to a total lipoprotein content in the cell membrane of 35.8% in ΔbgsA compared to only 9.4% in wild-type bacteria. Increased lipoprotein content strongly affected the recognition of ΔbgsA by mouse macrophages in vitro with an increased stimulation of TNF-α production by heat-fixed bacteria and secreted antigens. Inactivation of the prolipoprotein diacylglycerol transferase (lgt) in ΔbgsA abrogated TNF-α induction by a ΔbgsA_lgt double mutant indicating that lipoproteins mediate increased activation of mouse macrophages by ΔbgsA. Heat-fixed ΔbgsA bacteria, culture supernatant, or cell membrane lipid extract activated transfected HEK cells in a TLR2-dependent fashion; the same was not true of wild-type bacteria. In mice infected intraperitoneally with a sublethal dose of E. faecalis we observed a 70% greater mortality in mice infected with ΔbgsA compared with wild-type-infected mice. Increased mortality due to ΔbgsA infection was associated with elevated plasma levels of the inflammatory cytokines TNF-α, IL-6 and MIP-2. In summary, our results provide evidence that an E. faecalis mutant lacking its major bilayer forming glycolipid DGlcDAG upregulates lipoprotein expression leading to increased activation of the host innate immune system and virulence in vivo.
Collapse
Affiliation(s)
- Christian Theilacker
- Center for Chronic Immunodeficiency (CCI), University Medical Center Freiburg, Freiburg, Germany
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
- * E-mail:
| | - Ann-Kristin Diederich
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
- Division of Pediatric Infectious Diseases, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
- Department of Microbiology, Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Andreas Otto
- Institute for Microbiology, Department of Microbial Physiology, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Irina G. Sava
- Research Center for Nutrition and Food Science, Technical University Munich, Freising, Germany
| | - Dominique Wobser
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Yinyin Bao
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Katrin Hese
- Center for Chronic Immunodeficiency (CCI), University Medical Center Freiburg, Freiburg, Germany
| | - Melanie Broszat
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Center for Chronic Immunodeficiency (CCI), University Medical Center Freiburg, Freiburg, Germany
- Center for Paediatrics and Adolescent Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Dörte Becher
- Institute for Microbiology, Department of Microbial Physiology, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Johannes Huebner
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
- Division of Pediatric Infectious Diseases, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
49
|
Ocvirk S, Sava IG, Lengfelder I, Lagkouvardos I, Steck N, Roh JH, Tchaptchet S, Bao Y, Hansen JJ, Huebner J, Carroll IM, Murray BE, Sartor RB, Haller D. Surface-Associated Lipoproteins Link Enterococcus faecalis Virulence to Colitogenic Activity in IL-10-Deficient Mice Independent of Their Expression Levels. PLoS Pathog 2015; 11:e1004911. [PMID: 26067254 PMCID: PMC4466351 DOI: 10.1371/journal.ppat.1004911] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 04/24/2015] [Indexed: 12/22/2022] Open
Abstract
The commensal Enterococcus faecalis is among the most common causes of nosocomial infections. Recent findings regarding increased abundance of enterococci in the intestinal microbiota of patients with inflammatory bowel diseases and induction of colitis in IL-10-deficient (IL-10-/-) mice put a new perspective on the contribution of E. faecalis to chronic intestinal inflammation. Based on the expression of virulence-related genes in the inflammatory milieu of IL-10-/- mice using RNA-sequencing analysis, we characterized the colitogenic role of two bacterial structures that substantially impact on E. faecalis virulence by different mechanisms: the enterococcal polysaccharide antigen and cell surface-associated lipoproteins. Germ-free wild type and IL-10-/- mice were monoassociated with E. faecalis wild type OG1RF or the respective isogenic mutants for 16 weeks. Intestinal tissue and mesenteric lymph nodes (MLN) were collected to characterize tissue pathology, loss of intestinal barrier function, bacterial adhesion to intestinal epithelium and immune cell activation. Bone marrow-derived dendritic cells (BMDC) were stimulated with bacterial lysates and E. faecalis virulence was additionally investigated in three invertebrate models. Colitogenic activity of wild type E. faecalis (OG1RF score: 7.2±1.2) in monoassociated IL-10-/- mice was partially impaired in E. faecalis lacking enterococcal polysaccharide antigen (ΔepaB score: 4.7±2.3; p<0.05) and was almost completely abrogated in E. faecalis deficient for lipoproteins (Δlgt score: 2.3±2.3; p<0.0001). Consistently both E. faecalis mutants showed significantly impaired virulence in Galleria mellonella and Caenorhabditis elegans. Loss of E-cadherin in the epithelium was shown for all bacterial strains in inflamed IL-10-/- but not wild type mice. Inactivation of epaB in E. faecalis reduced microcolony and biofilm formation in vitro, altered bacterial adhesion to intestinal epithelium of germ-free Manduca sexta larvae and impaired penetration into the colonic mucus layer of IL-10-/- mice. Lipoprotein-deficient E. faecalis exhibited an impaired TLR2-mediated activation of BMDCs in vitro despite their ability to fully reactivate MLN cells as well as MLN-derived colitogenic T cells ex vivo. E. faecalis virulence factors accounting for bacterial adhesion to mucosal surfaces as well as intestinal barrier disruption partially contribute to colitogenic activity of E. faecalis. Beyond their well-known role in infections, cell surface-associated lipoproteins are essential structures for colitogenic activity of E. faecalis by mediating innate immune cell activation. Enterococcus faecalis is a commensal of the human intestinal core microbiota harboring several putative virulence factors, which highlight its role as opportunistic pathogen. This dualistic character is supported by recent evidence linking Enterococcus spp. to the pathogenesis of inflammatory bowel diseases (IBD). Although several studies suggest a crucial role for opportunistic pathogens in IBD pathogenesis targeting genetically susceptible individuals, the dynamic relationship between disease-relevant host compartments and specific bacterial structures able to trigger intestinal inflammation remain unclear. Here, we report that cell surface-associated lipoproteins and the enterococcal polysaccharide antigen, which are relevant for E. faecalis virulence in invertebrate infection models, but whose expression is minimally affected by the intestinal inflammatory milieu, exhibit colitogenic activity in a mouse model susceptible for chronic colitis. Bacterial lipoproteins trigger innate immune cell activation and are a critical prerequisite for E. faecalis-induced colitis. The enterococcal polysaccharide antigen mediates bacterial mucus penetration and adhesion to mucosal surfaces, promotes the formation of biofilm and contributes to E. faecalis colitogenic activity. Using E. faecalis as a model organism, we demonstrate that colitogenic activity of opportunistic pathogens can be assigned to specific bacterial structures, a finding that may help to identify the most essential steps in IBD-related microbe-host interactions.
Collapse
Affiliation(s)
- Soeren Ocvirk
- Technische Universität München, Chair of Nutrition and Immunology, ZIEL–Research Center for Nutrition and Food Sciences, Freising-Weihenstephan, Germany
| | - Irina G. Sava
- Technische Universität München, Chair of Nutrition and Immunology, ZIEL–Research Center for Nutrition and Food Sciences, Freising-Weihenstephan, Germany
| | - Isabella Lengfelder
- Technische Universität München, Chair of Nutrition and Immunology, ZIEL–Research Center for Nutrition and Food Sciences, Freising-Weihenstephan, Germany
| | - Ilias Lagkouvardos
- Technische Universität München, Chair of Nutrition and Immunology, ZIEL–Research Center for Nutrition and Food Sciences, Freising-Weihenstephan, Germany
| | - Natalie Steck
- Technische Universität München, Chair of Nutrition and Immunology, ZIEL–Research Center for Nutrition and Food Sciences, Freising-Weihenstephan, Germany
| | - Jung H. Roh
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Medical School, Houston, Texas, United States of America
| | - Sandrine Tchaptchet
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Yinyin Bao
- Division of Infectious Diseases, Department of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Jonathan J. Hansen
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Johannes Huebner
- Division of Infectious Diseases, Department of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Ian M. Carroll
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Barbara E. Murray
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Medical School, Houston, Texas, United States of America
| | - R. Balfour Sartor
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Dirk Haller
- Technische Universität München, Chair of Nutrition and Immunology, ZIEL–Research Center for Nutrition and Food Sciences, Freising-Weihenstephan, Germany
- * E-mail:
| |
Collapse
|
50
|
Biagini M, Garibaldi M, Aprea S, Pezzicoli A, Doro F, Becherelli M, Taddei AR, Tani C, Tavarini S, Mora M, Teti G, D'Oro U, Nuti S, Soriani M, Margarit I, Rappuoli R, Grandi G, Norais N. The Human Pathogen Streptococcus pyogenes Releases Lipoproteins as Lipoprotein-rich Membrane Vesicles. Mol Cell Proteomics 2015; 14:2138-49. [PMID: 26018414 DOI: 10.1074/mcp.m114.045880] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Indexed: 01/24/2023] Open
Abstract
Bacterial lipoproteins are attractive vaccine candidates because they represent a major class of cell surface-exposed proteins in many bacteria and are considered as potential pathogen-associated molecular patterns sensed by Toll-like receptors with built-in adjuvanticity. Although Gram-negative lipoproteins have been extensively characterized, little is known about Gram-positive lipoproteins. We isolated from Streptococcus pyogenes a large amount of lipoproteins organized in vesicles. These vesicles were obtained by weakening the bacterial cell wall with a sublethal concentration of penicillin. Lipid and proteomic analysis of the vesicles revealed that they were enriched in phosphatidylglycerol and almost exclusively composed of lipoproteins. In association with lipoproteins, a few hypothetical proteins, penicillin-binding proteins, and several members of the ExPortal, a membrane microdomain responsible for the maturation of secreted proteins, were identified. The typical lipidic moiety was apparently not necessary for lipoprotein insertion in the vesicle bilayer because they were also recovered from the isogenic diacylglyceryl transferase deletion mutant. The vesicles were not able to activate specific Toll-like receptor 2, indicating that lipoproteins organized in these vesicular structures do not act as pathogen-associated molecular patterns. In light of these findings, we propose to name these new structures Lipoprotein-rich Membrane Vesicles.
Collapse
Affiliation(s)
- Massimiliano Biagini
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Manuela Garibaldi
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Susanna Aprea
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Alfredo Pezzicoli
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Francesco Doro
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Marco Becherelli
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Anna Rita Taddei
- §Centro Interdipartimentale di Microscopia Elettronica, Università della Tuscia, Viterbo, Italy
| | - Chiara Tani
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Simona Tavarini
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Marirosa Mora
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Giuseppe Teti
- ¶Dipartimento di Scienze Pediatriche, Ginecologiche, Microbiologiche e Biomediche, Università degli Studi di Messina, Messina, Italy
| | - Ugo D'Oro
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Sandra Nuti
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Marco Soriani
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Immaculada Margarit
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Rino Rappuoli
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Guido Grandi
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay
| | - Nathalie Norais
- From the ‡Novartis Vaccines and Diagnostics (a GSK company), Via Fiorentiina 1, 53100 Siena, Itlay;
| |
Collapse
|