1
|
Belfiori B, Riccioni C, Pietrella D, Rubini A, Caceres ME, Pupilli F, Bellucci M, De Marchis F. Vicia ervilia lectin (VEA) has an antibiofilm effect on both Gram-positive and Gram-negative pathogenic bacteria. Arch Microbiol 2024; 206:371. [PMID: 39122975 PMCID: PMC11315768 DOI: 10.1007/s00203-024-04100-6] [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: 06/06/2024] [Revised: 07/23/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
Bacterial growing resistance to antibiotics poses a critical threat to global health. This study investigates, for the first time, the antibiofilm properties of Vicia ervilia agglutinin (VEA) from six different V. ervilia accessions against pathogenic bacteria, and the yeast Candida albicans. In the absence of antimicrobial properties, purified VEA significantly inhibited biofilm formation, both in Gram-positive and Gram-negative bacteria, but not in C. albicans. With an inhibitory concentration ranging from 100 to 500 µg/ml, the VEA antibiofilm activity was more relevant against the Gram-positive bacteria Streptococcus aureus and Staphylococcus epidermidis, whose biofilm was reduced up to 50% by VEA purified from accessions #5 and #36. VEA antibiofilm variability between accessions was observed, likely due to co-purified small molecules rather than differences in VEA protein sequences. In conclusion, VEA seed extracts from the accessions with the highest antibiofilm activity could represent a valid approach for the development of an effective antibiofilm agent.
Collapse
Affiliation(s)
- Beatrice Belfiori
- Institute of Biosciences and Bioresources (IBBR), National Research Council (CNR), 06128, Perugia, Italy
| | - Claudia Riccioni
- Institute of Biosciences and Bioresources (IBBR), National Research Council (CNR), 06128, Perugia, Italy
| | - Donatella Pietrella
- Department of Medicine and Surgery, University of Perugia, Piazzale S. Gambuli 1, 06132, Perugia, Italy
| | - Andrea Rubini
- Institute of Biosciences and Bioresources (IBBR), National Research Council (CNR), 06128, Perugia, Italy
| | - Maria Eugenia Caceres
- Institute of Biosciences and Bioresources (IBBR), National Research Council (CNR), 06128, Perugia, Italy
| | - Fulvio Pupilli
- Institute of Biosciences and Bioresources (IBBR), National Research Council (CNR), 06128, Perugia, Italy
| | - Michele Bellucci
- Institute of Biosciences and Bioresources (IBBR), National Research Council (CNR), 06128, Perugia, Italy.
| | - Francesca De Marchis
- Institute of Biosciences and Bioresources (IBBR), National Research Council (CNR), 06128, Perugia, Italy.
| |
Collapse
|
2
|
Sokkar MF, Mosaad RM, Khalil M, Kamal L. MBL2 gene variants and susceptibility to meningitis in Egyptian patients. Gene 2023; 872:147442. [PMID: 37121343 DOI: 10.1016/j.gene.2023.147442] [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: 02/02/2023] [Revised: 04/12/2023] [Accepted: 04/18/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND Meningitis is inflammation of the membranes enclosing the brain and spinal cord. It is a fatal disease with severe morbidity and mortality. Mannose binding lectin (MBL) encoded by MBL2 gene activates complement system through lectin pathway in innate immunity to defense against the infections. OBJECTIVE the current study aimed to investigate the promoter and exon 1 variants of MBL2 gene among Egyptian patients having meningitis to explore their role in disease susceptibility. PATIENTS AND METHODS This case-control study, included 53 patients and 50 sex and age matched controls. MBL2 genotyping was done using Sanger sequencing. RESULTS The frequency of one promoter (c.-290C>G) and four in exon 1 (c.161G>A, c.170G>A, c.154C>T and c.132C>T) as well as another one located in its 5'utranslated part (c.-66C>T) variants were estimated. The incidence of the four individual exonic variants was not significantly different between cases and healthy individuals (all P> 0.05). The promoter variant, c.-290C>G was found in all examined patients (84.9% of the patients in homozygote state and 15.1% of patients in heterozygous state) with a highly significant variance in the prevalence of this variant between cases and control group (p=0.0001). Additionally, UTR variant (c.-66C>T) was also significantly higher in patients than controls (P=0.033).In comparison with clinical outcome, it was found that c.170G>A variant named C allele was associated with favorable outcome in the studied patients (P=0.025). CONCLUSION The results obtained showed that the Promoter (c.-290C>G) and UTR (c.-66 C>T) variants of MBL2 gene may be potential risk factors for disease susceptibility in Egyptian cases with meningitis. Our results also proposed that c.170G>A (C allele and CC genotype) could affect the severity and play a protective role in these patients. The other genetic variants of MBL2 gene, including c.132C>T, c.161G>A (A>B), and c.154C>T (A>D) that were investigated, did not show any association with susceptibility or severity of meningitis.
Collapse
Affiliation(s)
- Mona F Sokkar
- Molecular Genetics and Enzymology department, Human Genetics and Genome Research Institute (HGGR), National Research Centre (NRC), Cairo, Egypt
| | - Rehab M Mosaad
- Infection disease department, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt.
| | - Mahmoud Khalil
- Infection disease department, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Lamyaa Kamal
- Clinical and chemical pathology department, Elsahel Teaching Hospital, Cairo, Egypt
| |
Collapse
|
3
|
Bajgar A, Krejčová G. On the origin of the functional versatility of macrophages. Front Physiol 2023; 14:1128984. [PMID: 36909237 PMCID: PMC9998073 DOI: 10.3389/fphys.2023.1128984] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Macrophages represent the most functionally versatile cells in the animal body. In addition to recognizing and destroying pathogens, macrophages remove senescent and exhausted cells, promote wound healing, and govern tissue and metabolic homeostasis. In addition, many specialized populations of tissue-resident macrophages exhibit highly specialized functions essential for the function of specific organs. Sometimes, however, macrophages cease to perform their protective function and their seemingly incomprehensible response to certain stimuli leads to pathology. In this study, we address the question of the origin of the functional versatility of macrophages. To this end, we have searched for the evolutionary origin of macrophages themselves and for the emergence of their characteristic properties. We hypothesize that many of the characteristic features of proinflammatory macrophages evolved in the unicellular ancestors of animals, and that the functional repertoire of macrophage-like amoebocytes further expanded with the evolution of multicellularity and the increasing complexity of tissues and organ systems. We suggest that the entire repertoire of macrophage functions evolved by repurposing and diversification of basic functions that evolved early in the evolution of metazoans under conditions barely comparable to that in tissues of multicellular organisms. We believe that by applying this perspective, we may find an explanation for the otherwise counterintuitive behavior of macrophages in many human pathologies.
Collapse
Affiliation(s)
- Adam Bajgar
- Faculty of Science, Department of Molecular Biology and Genetics, University of South Bohemia, Ceske Budejovice, Czechia.,Biology Centre, Institute of Entomology, Academy of Sciences, Ceske Budejovice, Czechia
| | - Gabriela Krejčová
- Faculty of Science, Department of Molecular Biology and Genetics, University of South Bohemia, Ceske Budejovice, Czechia.,Biology Centre, Institute of Entomology, Academy of Sciences, Ceske Budejovice, Czechia
| |
Collapse
|
4
|
Vacariu CM, Tanner ME. Recent Advances in the Synthesis and Biological Applications of Peptidoglycan Fragments. Chemistry 2022; 28:e202200788. [PMID: 35560956 DOI: 10.1002/chem.202200788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Indexed: 11/09/2022]
Abstract
The biosynthesis, breakdown, and modification of peptidoglycan (PG) play vital roles in both bacterial viability and in the response of human physiology to bacterial infection. Studies on PG biochemistry are hampered by the fact that PG is an inhomogeneous insoluble macromolecule. Chemical synthesis is therefore an important means to obtain PG fragments that may serve as enzyme substrates and elicitors of the human immune response. This review outlines the recent advances in the synthesis and biochemical studies of PG fragments, PG biosynthetic intermediates (such as Park's nucleotides and PG lipids), and PG breakdown products (such as muramyl dipeptides and anhydro-muramic acid-containing fragments). A rich variety of synthetic approaches has been applied to preparing such compounds since carbohydrate, peptide, and phospholipid chemical methodologies must all be applied.
Collapse
Affiliation(s)
- Condurache M Vacariu
- Department of Chemistry, University of British Columbia, V6T 1Z1, Vancouver, British Columbia, Canada
| | - Martin E Tanner
- Department of Chemistry, University of British Columbia, V6T 1Z1, Vancouver, British Columbia, Canada
| |
Collapse
|
5
|
Naseri M, Maliha M, Dehghani M, Simon GP, Batchelor W. Rapid Detection of Gram-Positive and -Negative Bacteria in Water Samples Using Mannan-Binding Lectin-Based Visual Biosensor. ACS Sens 2022; 7:951-959. [PMID: 35290028 DOI: 10.1021/acssensors.1c01748] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Waterborne bacterial infection is a health threat worldwide, making accurate and timely bacteria detection crucial to prevent waterborne disease outbreaks. Inspired by the intrinsic capability of mannan-binding lectin (MBL) in recognizing the pathogen-associated molecular patterns (PAMPs), a visual biosensor is developed here for the on-site detection of both Gram-positive and -negative bacteria. The biosensor was synthesized by immobilization of the MBL protein onto the blue carboxyl-functionalized polystyrene microparticles (PSM), which is then used in a two-step assay to detect bacterial cells in water samples. The first step involved a 20 min incubation following the MBL-PSM and calcium chloride solution addition to the samples. The second step was to add ethanol to the resultant blue mixture and observe the color change with the naked eye after 15 min. The biosensor had a binary (all-or-none) response, which in the presence of bacterial cells kept its blue color, while in their absence the color changed from blue to colorless. Testing the water samples spiked with four Gram-negative bacteria including Acinetobacter baumannii, Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa and two Gram-positive bacteria of Enterococcus faecalis and Staphylococcus aureus showed that the biosensor could detect all tested bacteria with a concentration as low as 101.5 CFU/ml. The performance of biosensor using the water samples from a water treatment plant also confirmed its capability to detect the pathogens in real-life water samples without the need for instrumentation.
Collapse
Affiliation(s)
- Mahdi Naseri
- Bioresource Processing Research Institute of Australia (BioPRIA), Department of Chemical Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Maisha Maliha
- Bioresource Processing Research Institute of Australia (BioPRIA), Department of Chemical Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Mostafa Dehghani
- Bioresource Processing Research Institute of Australia (BioPRIA), Department of Chemical Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - George P Simon
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Warren Batchelor
- Bioresource Processing Research Institute of Australia (BioPRIA), Department of Chemical Engineering, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
6
|
Liu M, Wang Q, Liu H, Yin C, Mijiti X, Anwaierjiang A, Wan K, Xu M, Li M, Nong S, Li G, Xiao H. Association of Mannose-Binding Lectin 2 Gene Polymorphism with Tuberculosis Based on Mycobacterium tuberculosis Lineages. Infect Drug Resist 2022; 15:1225-1234. [PMID: 35355619 PMCID: PMC8959721 DOI: 10.2147/idr.s344935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/25/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Polymorphisms in MBL2 may contribute to the susceptibility to tuberculosis. The aim of the present study was to determine the associations of the polymorphisms of five loci (rs1800450, rs1800451, rs7096206, rs7095891, and rs11003125) in the MBL2 gene with susceptibility to tuberculosis and specific lineages of Mycobacterium tuberculosis causing tuberculosis in the Uyghur population of Xinjiang, China. Methods From January 2019 to January 2020, we enrolled 170 Uyghur tuberculosis patients as the case group and 147 Uyghur staff with no clinical symptoms as the control group from four designated tuberculosis hospitals in southern Xinjiang, China. The polymorphisms of five loci in MBL2 of human were detected by sequencing. Whole-genome sequencing was applied in 68 M. tuberculosis isolates from the case group and the data were used to perform genealogy analysis. Results The distributions of allele and genotype frequencies of five loci in MBL2 varied little between the case and control groups and varied little among the groups, including those infected with different lineages of M. tuberculosis and the control (except those of rs11003125), the P values were all >0.05. The distribution of alleles of rs11003125 was statistically different between patients infected with lineages 3 and 4 M. tuberculosis (χ2=7.037, P=0.008). The C allele and CC genotype of rs11003125 were found to be protective factors against lineage 4 infection when compared to lineage 3 (ORs were 0.190 and 0.158, respectively; 95% confidence intervals were 0.053~0.690 and 0.025~0.999, respectively). Conclusion Our results suggested that human’s susceptibility to tuberculosis is affected both by the host genetic polymorphisms and the lineage of the M. tuberculosis that people were exposed to. However, due to the limitation of the sample size in the present study, larger sample size and more rigorous design should be guaranteed in future studies.
Collapse
Affiliation(s)
- Mengwen Liu
- School of Public Health, Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
| | - Quan Wang
- The Eighth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830001, People’s Republic of China
| | - Haican Liu
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People’s Republic of China
| | - Chunjie Yin
- School of Public Health, Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
| | - Xiaokaiti Mijiti
- The Eighth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830001, People’s Republic of China
| | - Aiketaguli Anwaierjiang
- School of Public Health, Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
| | - Kanglin Wan
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People’s Republic of China
| | - Miao Xu
- The Eighth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830001, People’s Republic of China
| | - Machao Li
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People’s Republic of China
| | - Siqin Nong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People’s Republic of China
| | - Guilian Li
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, People’s Republic of China
- Correspondence: Guilian Li; Hui Xiao, Email ;
| | - Hui Xiao
- School of Public Health, Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
| |
Collapse
|
7
|
Kottom TJ, Carmona EM, Limper AH. Current State of Carbohydrate Recognition and C-Type Lectin Receptors in Pneumocystis Innate Immunity. Front Immunol 2021; 12:798214. [PMID: 34975910 PMCID: PMC8716372 DOI: 10.3389/fimmu.2021.798214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Pneumocystis jirovecii is one of the most common fungal pathogens in immunocompromised individuals. Pneumocystis jirovecii pneumonia (PJP) causes a significant host immune response that is driven greatly by the organism’s cell wall components including β-glucans and major surface glycoprotein (Msg). These ligands interact with a number of C-type lectin receptors (CLRs) leading to downstream activation of proinflammatory signaling pathways. This minireview provides a brief overview summarizing known CLR/Pneumocystis interactions.
Collapse
Affiliation(s)
- Theodore J. Kottom
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic, Rochester, MN, United States
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
- *Correspondence: Theodore J. Kottom,
| | - Eva M. Carmona
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic, Rochester, MN, United States
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Andrew H. Limper
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic, Rochester, MN, United States
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
8
|
Li Q, Yu C, Chen Y, Liu S, Azevedo P, Gong J, O K, Yang C. Citral alleviates peptidoglycan-induced inflammation and disruption of barrier functions in porcine intestinal epithelial cells. J Cell Physiol 2021; 237:1768-1779. [PMID: 34791644 DOI: 10.1002/jcp.30640] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 11/11/2022]
Abstract
Peptidoglycan (PGN) is a major polymer in bacterial cell walls and may constrain gut functionality and lower intestinal efficiencies in livestock. Citral has been reported to exhibit antibacterial and anti-inflammatory biological activities, improving the gastrointestinal function of swine. However, the protective effect of citral against PGN-elicited cellular responses and possible underlying mechanisms are unknown. In this study, the porcine jejunal epithelial cell line (IPEC-J2) was challenged with PGN from Staphylococcus aureus (S. aureus) or Bacillus subtilis (B. subtilis) to explore PGN-induced inflammatory responses. Our data showed that the inflammatory response stimulated by PGN from harmful bacteria (S. aureus) was more potent than that from commensal bacteria (B. subtilis) in IPEC-J2 cells. Based on the inflammatory model by PGN from S. aureus, it was demonstrated that PGN could significantly induce inflammatory cytokine production and influence nutrient absorption and barrier function in a dose-dependent manner. However, the PGN-mediated immune responses were remarkably suppressed by citral. In addition, citral significantly attenuated the effect of PGN on the intestine nutrient absorption and barrier function. The expression of TLR2 was strongly induced by PGN stimulation, which was suppressed by citral. All data nominated that citral downregulated PGN-induced inflammation via TLR2-mediated activation of the NF-κB signaling pathway in IPEC-J2 cells. Furthermore, the results also indicate that the PGN degradation through the inclusion of enzymes (e.g., muramidase) as well as the inclusion of citral for attenuating inflammation may improve pig gut health and functionality.
Collapse
Affiliation(s)
- Qiao Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Changning Yu
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yanhong Chen
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shangxi Liu
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Paula Azevedo
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Joshua Gong
- Guelph Research and Development Centre, Agriculture Agri-Food Canada, Guelph, Ontario, Canada
| | - Karmin O
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Chengbo Yang
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
9
|
Maulloo CD, Cao S, Watkins EA, Raczy MM, Solanki AS, Nguyen M, Reda JW, Shim HN, Wilson DS, Swartz MA, Hubbell JA. Lymph Node-Targeted Synthetically Glycosylated Antigen Leads to Antigen-Specific Immunological Tolerance. Front Immunol 2021; 12:714842. [PMID: 34630389 PMCID: PMC8498032 DOI: 10.3389/fimmu.2021.714842] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/07/2021] [Indexed: 12/19/2022] Open
Abstract
Inverse vaccines that tolerogenically target antigens to antigen-presenting cells (APCs) offer promise in prevention of immunity to allergens and protein drugs and treatment of autoimmunity. We have previously shown that targeting hepatic APCs through intravenous injection of synthetically glycosylated antigen leads to effective induction of antigen-specific immunological tolerance. Here, we demonstrate that targeting these glycoconjugates to lymph node (LN) APCs under homeostatic conditions leads to local and increased accumulation in the LNs compared to unmodified antigen and induces a tolerogenic state both locally and systemically. Subcutaneous administration directs the polymeric glycoconjugate to the draining LN, where the glycoconjugated antigen generates robust antigen-specific CD4+ and CD8+ T cell tolerance and hypo-responsiveness to antigenic challenge via a number of mechanisms, including clonal deletion, anergy of activated T cells, and expansion of regulatory T cells. Lag-3 up-regulation on CD4+ and CD8+ T cells represents an essential mechanism of suppression. Additionally, presentation of antigen released from the glycoconjugate to naïve T cells is mediated mainly by LN-resident CD8+ and CD11b+ dendritic cells. Thus, here we demonstrate that antigen targeting via synthetic glycosylation to impart affinity for APC scavenger receptors generates tolerance when LN dendritic cells are the cellular target.
Collapse
Affiliation(s)
- Chitavi D. Maulloo
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Shijie Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Elyse A. Watkins
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Michal M. Raczy
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Ani. S. Solanki
- Animal Resources Center, University of Chicago, Chicago, IL, United States
| | - Mindy Nguyen
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Joseph W. Reda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Ha-Na Shim
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - D. Scott Wilson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
- Biomedical Engineering Department, Johns Hopkins University, Baltimore, MD, United States
| | - Melody A. Swartz
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
- Committee on Immunology, University of Chicago, Chicago, IL, United States
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL, United States
- Committee on Cancer Biology, University of Chicago, Chicago, IL, United States
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
- Committee on Immunology, University of Chicago, Chicago, IL, United States
- Committee on Cancer Biology, University of Chicago, Chicago, IL, United States
| |
Collapse
|
10
|
Wasser B, Luchtman D, Löffel J, Robohm K, Birkner K, Stroh A, Vogelaar CF, Zipp F, Bittner S. CNS-localized myeloid cells capture living invading T cells during neuroinflammation. J Exp Med 2021; 217:151587. [PMID: 32219436 PMCID: PMC7971133 DOI: 10.1084/jem.20190812] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/20/2019] [Accepted: 01/17/2020] [Indexed: 11/28/2022] Open
Abstract
To study the role of myeloid cells in the central nervous system (CNS) in the pathogenesis of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), we used intravital microscopy, assessing local cellular interactions in vivo in EAE animals and ex vivo in organotypic hippocampal slice cultures. We discovered that myeloid cells actively engulf invading living Th17 lymphocytes, a process mediated by expression of activation-dependent lectin and its T cell–binding partner, N-acetyl-D-glucosamine (GlcNAc). Stable engulfment resulted in the death of the engulfed cells, and, remarkably, enhancement of GlcNAc exposure on T cells in the CNS ameliorated clinical EAE symptoms. These findings demonstrate the ability of myeloid cells to directly react to pathogenic T cell infiltration by engulfing living T cells. Amelioration of EAE via GlcNAc treatment suggests a novel first-defense pathway of myeloid cells as an initial response to CNS invasion and demonstrates that T cell engulfment by myeloid cells can be therapeutically exploited in vivo.
Collapse
Affiliation(s)
- Beatrice Wasser
- Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Dirk Luchtman
- Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Julian Löffel
- Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Kerstin Robohm
- Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Katharina Birkner
- Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Albrecht Stroh
- Focus Program Translational Neurosciences, Institute for Microscopic Anatomy and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Christina Francisca Vogelaar
- Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
11
|
Innate Immune Pattern Recognition Receptors of Mycobacterium tuberculosis: Nature and Consequences for Pathogenesis of Tuberculosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:179-215. [PMID: 34661896 DOI: 10.1007/978-3-030-67452-6_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Innate immunity against Mycobacterium tuberculosis is a critical early response to prevent the establishment of the infection. Despite recent advances in understanding the host-pathogen dialogue in the early stages of tuberculosis (TB), much has yet to be learnt. The nature and consequences of this dialogue ultimately determine the path of infection: namely, either early clearance of M. tuberculosis, or establishment of M. tuberculosis infection leading to active TB disease and/or latent TB infection. On the frontline in innate immunity are pattern recognition receptors (PRRs), with soluble factors (e.g. collectins and complement) and cell surface factors (e.g. Toll-like receptors and other C-type lectin receptors (Dectin 1/2, Nod-like receptors, DC-SIGN, Mincle, mannose receptor, and MCL) that play a central role in recognising M. tuberculosis and facilitating its clearance. However, in a 'double-edged sword' scenario, these factors can also be involved in enhancement of pathogenesis as well. Furthermore, innate immunity is also a critical bridge in establishing the subsequent adaptive immune response, which is also responsible for granuloma formation that cordons off M. tuberculosis infection, establishing latency and acting as a reservoir for bacterial persistence and dissemination of future disease. This chapter discusses the current understanding of pattern recognition of M. tuberculosis by innate immunity and the role this plays in the pathogenesis and protection against TB.
Collapse
|
12
|
Murugaiah V, Tsolaki AG, Kishore U. Collectins: Innate Immune Pattern Recognition Molecules. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1204:75-127. [PMID: 32152944 PMCID: PMC7120701 DOI: 10.1007/978-981-15-1580-4_4] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Collectins are collagen-containing C-type (calcium-dependent) lectins which are important pathogen pattern recognising innate immune molecules. Their primary structure is characterised by an N-terminal, triple-helical collagenous region made up of Gly-X-Y repeats, an a-helical coiled-coil trimerising neck region, and a C-terminal C-type lectin or carbohydrate recognition domain (CRD). Further oligomerisation of this primary structure can give rise to more complex and multimeric structures that can be seen under electron microscope. Collectins can be found in serum as well as in a range of tissues at the mucosal surfaces. Mannanbinding lectin can activate the complement system while other members of the collectin family are extremely versatile in recognising a diverse range of pathogens via their CRDs and bring about effector functions designed at the clearance of invading pathogens. These mechanisms include opsonisation, enhancement of phagocytosis, triggering superoxidative burst and nitric oxide production. Collectins can also potentiate the adaptive immune response via antigen presenting cells such as macrophages and dendritic cells through modulation of cytokines and chemokines, thus they can act as a link between innate and adaptive immunity. This chapter describes the structure-function relationships of collectins, their diverse functions, and their interaction with viruses, bacteria, fungi and parasites.
Collapse
Affiliation(s)
- Valarmathy Murugaiah
- College of Health and Life Sciences, Brunel University London, London, UB8 3PH, UK
| | - Anthony G Tsolaki
- College of Health and Life Sciences, Brunel University London, London, UB8 3PH, UK
| | - Uday Kishore
- College of Health and Life Sciences, Brunel University London, London, UB8 3PH, UK.
| |
Collapse
|
13
|
de Vor L, Rooijakkers SHM, van Strijp JAG. Staphylococci evade the innate immune response by disarming neutrophils and forming biofilms. FEBS Lett 2020; 594:2556-2569. [PMID: 32144756 DOI: 10.1002/1873-3468.13767] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/30/2020] [Accepted: 02/22/2020] [Indexed: 12/24/2022]
Abstract
Staphylococcus aureus and Staphylococcus epidermidis can cause many types of infections, ranging from skin infections to implant-associated infections. The primary innate immune response against bacterial infections involves complement activation, recruitment of phagocytes (most importantly neutrophils), and subsequent killing of the pathogen. However, staphylococci are not innocent bystanders; they actively obstruct this immune attack. To do that, S. aureus secretes several immune-evasion proteins to resist attack by the innate immune system. Furthermore, S. aureus and S. epidermidis are known for their ability to form biofilms on implanted medical devices and host tissues, which provides another important immune-evasion mechanism. Understanding these different strategies to resist immune attack will help to develop novel therapies against staphylococcal infections.
Collapse
Affiliation(s)
- Lisanne de Vor
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Suzan H M Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, The Netherlands
| |
Collapse
|
14
|
Dang Y, Nie L, Chen J. Molecular and functional characterisation of a mannose-binding lectin-like gene from Japanese sea bass (Lateolabrax japonicus). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 104:103558. [PMID: 31751630 DOI: 10.1016/j.dci.2019.103558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 11/17/2019] [Accepted: 11/17/2019] [Indexed: 06/10/2023]
Abstract
Mannose-binding lectin (MBL) plays an important role in host immune responses against pathogens. LjMBL-like-1 was identified from Japanese sea bass (Lateolabrax japonicas), which has selectivity for galactose. Herein, this lectin might be better designated as galactose-binding lectin (LjGalBL-1). LjGalBL-1 transcripts were detected in all tested tissues, with highest expression in liver. Upon Vibrio harveyi infection, LjGalBL-1 mRNA expression was increased in major immune-related tissues, and protein levels in serum were also upregulated. Recombinant LjGalBL-1 (rLjGalBL-1) bound to monosaccharides and polysaccharides, and both rLjGalBL-1 and native LjGalBL-1 (nLjGalBL-1) agglutinated three Gram-positive bacteria (Staphylococcus aureus, Streptococcus iniae and Micrococcus luteus) and four Gram-negative bacteria (Aeromonas hydrophila, Edwardsiella tarda, Vibrio anguillarum and V. harveyi) in a Ca2+-dependent manner in vitro. Moreover, rLjGalBL-1 increased the survival rate of V. harveyi-infected fish and decreased bacterial load in liver, spleen, kidney and blood. Thus, LjGalBL-1 protects L. japonicas against V. harveyi infection.
Collapse
Affiliation(s)
- Yunfei Dang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China
| | - Li Nie
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315832, China.
| |
Collapse
|
15
|
Laman JD, 't Hart BA, Power C, Dziarski R. Bacterial Peptidoglycan as a Driver of Chronic Brain Inflammation. Trends Mol Med 2020; 26:670-682. [PMID: 32589935 DOI: 10.1016/j.molmed.2019.11.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/08/2019] [Accepted: 11/15/2019] [Indexed: 12/12/2022]
Abstract
Peptidoglycan (PGN) is a cell wall component of both Gram-positive and Gram-negative bacteria. Signature fragments of PGN are proinflammatory through engagement of pattern recognition receptors (PRR) on resident tissue cells and circulating leukocytes. Despite its abundance in the gut microbiota, there is limited recognition that PGN could contribute to chronic neuroinflammation. This review highlights current insights into the roles of PGN as a determinant of brain inflammation, notably in multiple sclerosis (MS) and its experimental autoimmune encephalomyelitis (EAE) models. Recent studies demonstrate PGN in blood of healthy adult humans. PGN amplifies autoimmune pathology via activation of innate immune cells. Novel uptake routes through (altered) gut mucosa by myeloid leukocyte subsets promote PGN transport to the brain.
Collapse
Affiliation(s)
- Jon D Laman
- Department of Biomedical Sciences of Cells and Systems, Section of Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Bert A 't Hart
- Department of Biomedical Sciences of Cells and Systems, Section of Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Anatomy and Neuroscience, Free University Amsterdam, Amsterdam, The Netherlands
| | - Christopher Power
- Department of Medicine (Neurology), University of Alberta, Edmonton, AB, Canada
| | - Roman Dziarski
- Indiana University School of Medicine-Northwest, Gary, IN, USA
| |
Collapse
|
16
|
Lukácsi S, Mácsik-Valent B, Nagy-Baló Z, Kovács KG, Kliment K, Bajtay Z, Erdei A. Utilization of complement receptors in immune cell-microbe interaction. FEBS Lett 2020; 594:2695-2713. [PMID: 31989596 DOI: 10.1002/1873-3468.13743] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/19/2022]
Abstract
The complement system is a major humoral component of immunity and is essential for the fast elimination of pathogens invading the body. In addition to its indispensable role in innate immunity, the complement system is also involved in pathogen clearance during the effector phase of adaptive immunity. The fastest way of killing the invader is lysis by the membrane attack complex, which is formed by the terminal components of the complement cascade. Not all pathogens are lysed however and, if opsonized by a variety of molecules, they undergo phagocytosis and disposal inside immune cells. The most important complement-derived opsonins are C1q, the first component of the classical pathway, MBL, the initiator of the lectin pathway and C3-derived activation fragments, including C3b, iC3b and C3d, which all serve as ligands for their corresponding receptors. In this review, we discuss how complement receptors are utilized by various immune cells to tackle invading microbes, or by pathogens to evade host response.
Collapse
Affiliation(s)
- Szilvia Lukácsi
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary
| | | | - Zsuzsa Nagy-Baló
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Kristóf G Kovács
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | | | - Zsuzsa Bajtay
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary.,Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Anna Erdei
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary.,Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
17
|
Lan T, Li Z, Peng M, Niu D, Li Y, Li J. A four-CRD C-type lectin from razor clam Sinonovacula constricta mediates agglutination and phagocytosis. Gene 2020; 728:144287. [DOI: 10.1016/j.gene.2019.144287] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/04/2019] [Accepted: 12/11/2019] [Indexed: 11/26/2022]
|
18
|
Abstract
The evolutionary separated Gram-negative Chlamydiales show a biphasic life cycle and replicate exclusively within eukaryotic host cells. Members of the genus Chlamydia are responsible for many acute and chronic diseases in humans, and Chlamydia-related bacteria are emerging pathogens. We revisit past efforts to detect cell wall material in Chlamydia and Chlamydia-related bacteria in the context of recent breakthroughs in elucidating the underlying cellular and molecular mechanisms of the chlamydial cell wall biosynthesis. In this review, we also discuss the role of cell wall biosynthesis in chlamydial FtsZ-independent cell division and immune modulation. In the past, penicillin susceptibility of an invisible wall was referred to as the "chlamydial anomaly." In light of new mechanistic insights, chlamydiae may now emerge as model systems to understand how a minimal and modified cell wall biosynthetic machine supports bacterial cell division and how cell wall-targeting beta-lactam antibiotics can also act bacteriostatically rather than bactericidal. On the heels of these discussions, we also delve into the effects of other cell wall antibiotics in individual chlamydial lineages.
Collapse
|
19
|
Irazoki O, Hernandez SB, Cava F. Peptidoglycan Muropeptides: Release, Perception, and Functions as Signaling Molecules. Front Microbiol 2019; 10:500. [PMID: 30984120 PMCID: PMC6448482 DOI: 10.3389/fmicb.2019.00500] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/27/2019] [Indexed: 12/12/2022] Open
Abstract
Peptidoglycan (PG) is an essential molecule for the survival of bacteria, and thus, its biosynthesis and remodeling have always been in the spotlight when it comes to the development of antibiotics. The peptidoglycan polymer provides a protective function in bacteria, but at the same time is continuously subjected to editing activities that in some cases lead to the release of peptidoglycan fragments (i.e., muropeptides) to the environment. Several soluble muropeptides have been reported to work as signaling molecules. In this review, we summarize the mechanisms involved in muropeptide release (PG breakdown and PG recycling) and describe the known PG-receptor proteins responsible for PG sensing. Furthermore, we overview the role of muropeptides as signaling molecules, focusing on the microbial responses and their functions in the host beyond their immunostimulatory activity.
Collapse
Affiliation(s)
| | | | - Felipe Cava
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| |
Collapse
|
20
|
Mannan-Binding Lectin Suppresses Peptidoglycan-Induced TLR2 Activation and Inflammatory Responses. Mediators Inflamm 2019; 2019:1349784. [PMID: 30728747 PMCID: PMC6343158 DOI: 10.1155/2019/1349784] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/01/2018] [Indexed: 01/08/2023] Open
Abstract
Peptidoglycan (PGN), as the major components of the bacterial cell wall, is known to cause excessive proinflammatory cytokine production. Toll-like receptor 2 (TLR2) is abundantly expressed on immune cells and has been shown to be involved in PGN-induced signaling. Although more and more evidences have indicated that PGN is recognized by TLR2, the role of TLR2 PGN recognition is controversial. Mannan-binding lectin (MBL), a plasma C-type lectin, plays a key role in innate immunity. More and more evidences show that MBL could suppress the amplification of inflammatory signals. Whether MBL can alter PGN-elicited cellular responses through TLR2 in macrophages is still unknown, and possible mechanism underlying it should be investigated. In this study, we found that MBL significantly attenuated PGN-induced inflammatory cytokine production, including TNF-α and IL-6, in PMA-stimulated THP-1 cells at both mRNA and protein levels. The expression of TLR2 was strongly induced by PGN stimulation. Furthermore, the administration of TLR2-neutralized antibody effectively suppressed PGN-induced TNF-α and IL-6 expression. These results supplied the evidence that PGN from Saccharomyces cerevisiae could be recognized by TLR2. In addition, we also found that MBL decreased PGN-induced TLR2 expression and suppressed TLR2-mediated downstream signaling, including the phosphorylation of IκBα, nuclear translocation of NF-κBp65, and phosphorylation of MAPK p38 and ERK1/2. Administration of MBL alone did not have an effect on the expression of TLR2. Finally, our data showed that PGN-mediated immune responses were more severely suppressed by preincubation with MBL and indicated that MBL can combine with both TLR2 and PGN to block the inflammation cytokine expression induced by PGN. All these data suggest that MBL could downregulate inflammation by modulating PGN/TLR2 signaling pathways. This study supports an important role for MBL in immune regulation and signaling pathways involved in inflammatory responses.
Collapse
|
21
|
Nantachit N, Sunintaboon P, Ubol S. EDIII-DENV3 nanospheres drive immature dendritic cells into a mature phenotype in an in vitro model. Microbiol Immunol 2018; 61:305-317. [PMID: 28603864 DOI: 10.1111/1348-0421.12497] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/15/2017] [Accepted: 06/06/2017] [Indexed: 01/04/2023]
Abstract
Domain III of E protein of dengue virus (DENV) is a target for vaccine development. Unfortunately, this protein based platform has low general immunogenicity. To circumvent this problem, the use of an adjuvant-nanoparticle delivery system to facilitate immunogenicity of soluble DENV-EDIII protein was investigated. One of the key features of this delivery system is its ability to simultaneously deliver antigens and exert adjuvanticity on specialized immune cells. In this study, N-trimethyl chitosan (TMC) nanoparticles (NPs) were generated to be used as adjuvant and carrier for soluble E-domain III of dengue virus serotype 3 (sEDIII-D3). Using ionotropic gelation, purified sEDIII-D3 was encapsulated into TMC NPs to form EDIII-D3 TMC NPs. After optimization, EDIII-D3 TMC particles exhibited a loading efficiency of 81% and a loading capacity of 41%. The immunogenicity of EDIII-D3 TMC NPs was tested using monocyte-derived dendritic cells (MoDCs). It was found that EDIII-D3 TMC NPs were well taken up by MoDCs. In addition, EDIII-D3 TMC NP treated MoDCs significantly upregulated maturation markers (CD80, CD83, CD86 and HLA-DR) and induced secretion of various cytokines and chemokines (IFN-α, IL-1β, IL-6, IL-2, IL-12p70, IFN-γ, IL-4, IL-10, IL-8, MCP-1, macrophage inflammatory protein-1β, granulocyte-colony stimulating factor, granulocyte-macrophage colony-stimulating factor and IL-7). These results indicate that EDIII-D3 TMC NPs are potent immunogens, at least in vitro, with the ability to induce maturation of DCs and highlight the potential use of TMC NPs for enhancing immunogenicity of a non-replicating dengue vaccine.
Collapse
Affiliation(s)
- Nattika Nantachit
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Panya Sunintaboon
- Department of Chemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
22
|
Abstract
Only a few extracellular soluble proteins are known to modulate apoptosis. We considered that surfactant-associated protein D (SP-D), an innate immune collectin present on many mucosal surfaces, could regulate apoptosis. Although SP-D is known to be important for immune cell homeostasis, whether SP-D affects apoptosis is unknown. In this study we aimed to determine the effects of SP-D on Jurkat T cells and human T cells dying by apoptosis. Here we show that SP-D binds to Jurkat T cells and delays the progression of Fas (CD95)-Fas ligand and TRAIL-TRAIL receptor induced, but not TNF-TNF receptor-mediated apoptosis. SP-D exerts its effects by reducing the activation of initiator caspase-8 and executioner caspase-3. SP-D also delays the surface exposure of phosphatidylserine. The effect of SP-D was ablated by the presence of caspase-8 inhibitor, but not by intrinsic pathway inhibitors. The binding ability of SP-D to dying cells decreases during the early stages of apoptosis, suggesting the release of apoptotic cell surface targets during apoptosis. SP-D also delays FasL-induced death of primary human T cells. SP-D delaying the progression of the extrinsic pathway of apoptosis could have important implications in regulating immune cell homeostasis at mucosal surfaces.
Collapse
|
23
|
Zogheib E, Nyga R, Cornu M, Sendid B, Monconduit J, Jounieaux V, Maizel J, Segard C, Chouaki T, Dupont H. Prospective Observational Study on the Association Between Serum Mannose-Binding Lectin Levels and Severe Outcome in Critically Ill Patients with Pandemic Influenza Type A (H1N1) Infection. Lung 2017; 196:65-72. [PMID: 29273833 PMCID: PMC7101572 DOI: 10.1007/s00408-017-0067-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 11/01/2017] [Indexed: 12/18/2022]
Abstract
Background Mannose-binding lectin (MBL) plays an important role in the innate immune response. In addition to activating the complement, MBL can induce cytokine production and contribute to a deleterious inflammatory response with severe A(H1N1)pdm09 virus infection. Our aim was to determine if serum MBL levels correlate with the risk of mortality in intensive care units (ICU) patients with A(H1N1)pdm09 infection. Methods Prospective observational study was performed in ICU patients with acute respiratory distress syndrome due to influenza A(H1N1)pdm09 virus. Demographic characteristics and severity indices were recorded at ICU admission. MBL was assayed from blood drawn at influenza diagnosis within 24–48 h following the ICU admission. Outcomes were compared according to MBL levels. Results are expressed as median and interquartile range. Results Serum MBL levels were studied in 27 patients (age: 56 [IQR 29] years) with severe A(H1N1)pdm09 infection and in 70 healthy controls. Median admission SAPSII and SOFA scores were 49 [IQR 26] and 12 [IQR 5], respectively. Mortality rate after a 30-day was 37%. MBL was significantly higher in non-survivors (3741 [IQR 2336] ng/ml) vs survivors (215 [IQR 1307] ng/ml), p = 0.006, as well as control group (1814 [IQR 2250] ng/ml), p = 0.01. In contrast, MBL levels in survivors group were significantly lower than the controls group (215 [IQR 1307] ng/ml vs. 1814 [IQR 2250] ng/ml, p = 0.005). MBL cut-off > 1870 ng/ml had a sensitivity of 80% and a specificity of 88.2% for mortality [AUC = 0.82 (95% CI 0.63–0.94)]. Kaplan–Meier analysis demonstrated a strong association between MBL levels and mortality (log-rank 7.8, p = 0.005). MBL > 1870 ng/ml was independently associated with mortality (HR = 8.7, 95% CI 1.2–29.1, p = 0.007). Conclusions This study shows that baseline MBL > 1870 ng/ml is associated with higher mortality in ICU patients with severe A(H1N1)pdm09 infection.
Collapse
Affiliation(s)
- Elie Zogheib
- Cardiothoracic and Vascular Intensive Care Unit, Amiens University Hospital, Amiens, France. .,INSERM U1088, Jules Verne University of Picardy, Amiens, France.
| | - Remy Nyga
- Medical Parasitology and Mycology Department, Amiens University Hospital, Amiens, France
| | - Marjorie Cornu
- Medical Parasitology and Mycology Department, CHU, Lille, France.,INSERM U995, Team Fungal Associated Invasive & Inflammatory Diseases, Lille Inflammation Research International Center, Université de Lille, Lille, France
| | - Boualem Sendid
- Medical Parasitology and Mycology Department, CHU, Lille, France.,INSERM U995, Team Fungal Associated Invasive & Inflammatory Diseases, Lille Inflammation Research International Center, Université de Lille, Lille, France
| | - Julien Monconduit
- Respiratory Intensive Care Unit, Amiens University Hospital, Amiens, France
| | - Vincent Jounieaux
- Respiratory Intensive Care Unit, Amiens University Hospital, Amiens, France
| | - Julien Maizel
- INSERM U1088, Jules Verne University of Picardy, Amiens, France.,Medical Intensive Care Unit, Amiens University Hospital, Amiens, France
| | - Christine Segard
- Medical Virology Department, Amiens University Hospital, Amiens, France
| | - Taïeb Chouaki
- Medical Parasitology and Mycology Department, Amiens University Hospital, Amiens, France
| | - Hervé Dupont
- Cardiothoracic and Vascular Intensive Care Unit, Amiens University Hospital, Amiens, France.,INSERM U1088, Jules Verne University of Picardy, Amiens, France
| |
Collapse
|
24
|
Djiadeu P, Farmakovski N, Azzouz D, Kotra LP, Sweezey N, Palaniyar N. Surfactant protein D regulates caspase-8-mediated cascade of the intrinsic pathway of apoptosis while promoting bleb formation. Mol Immunol 2017; 92:190-198. [PMID: 29107869 DOI: 10.1016/j.molimm.2017.10.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/17/2017] [Accepted: 10/17/2017] [Indexed: 01/10/2023]
Abstract
Surfactant-associated protein D (SP-D) is a soluble innate immune collectin present on many mucosal surfaces. We recently showed that SP-D suppresses the extrinsic pathway of apoptosis by downregulating caspase-8 activation. However, the effects of SP-D on the intrinsic pathway of apoptosis are not clearly understood. In the intrinsic pathway, cytochrome c is released by mitochondria into the cytoplasm. Oxidation of cytochrome c by cytochrome c oxidase activates the apoptosome and caspase-9 cascade. Both caspase-8- and caspase-9-mediated branches are activated in the intrinsic pathway of apoptosis; however, little is known about the relevance of the caspase-8 pathway in this context. Here we studied the effects of SP-D on different branches of the intrinsic pathway of apoptosis using UV-irradiated Jurkat T-cells. We found that SP-D does not inhibit the caspase-9 branch of apoptosis and the relevance of the caspase-8-related branch became apparent when the caspase-9 pathway was inhibited by blocking cytochrome c oxidase. Under these conditions, SP-D reduces the activation of caspase-8, executioner caspase-3 and exposure of phosphatidylserine (PS) on the membranes of dying cells. By contrast, SP-D increases the formation of nuclear and membrane blebs. Inhibition of caspase-8 confirms the effect of SP-D is unique to the caspase-8 pathway. Overall, SP-D suppresses certain aspects of the intrinsic pathway of apoptosis via reduction of caspase-8 activation and PS flipping while at the same time increasing membrane and nuclear bleb formation. This novel regulatory aspect of SP-D could help to regulate intrinsic pathway of apoptosis to promote effective blebbing and breakdown of dying cells.
Collapse
Affiliation(s)
- Pascal Djiadeu
- Lung Innate Immunity Research Laboratory, Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada; Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada
| | - Nicole Farmakovski
- Lung Innate Immunity Research Laboratory, Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada
| | - Dhia Azzouz
- Lung Innate Immunity Research Laboratory, Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada
| | - Lakshmi P Kotra
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada; Center for Molecular Design and Preformulations, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario, M5G 1L7, Canada
| | - Neil Sweezey
- Lung Innate Immunity Research Laboratory, Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada; Departments of Paediatrics, Physiology and Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, M5G 1X8, Canada
| | - Nades Palaniyar
- Lung Innate Immunity Research Laboratory, Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada; Departments of Laboratory Medicine and Pathobiology and Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
25
|
Understanding the molecular differential recognition of muramyl peptide ligands by LRR domains of human NOD receptors. Biochem J 2017; 474:2691-2711. [DOI: 10.1042/bcj20170220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/27/2017] [Accepted: 07/03/2017] [Indexed: 12/28/2022]
Abstract
Human nucleotide-binding oligomerization domain proteins, hNOD1 and hNOD2, are host intracellular receptors with C-terminal leucine-rich repeat (LRR) domains, which recognize specific bacterial peptidoglycan (PG) fragments as their ligands. The specificity of this recognition is dependent on the third amino acid of the stem peptide of the PG ligand, which is usually meso-diaminopimelic acid (mesoDAP) or l-lysine (l-Lys). Since the LRR domains of hNOD receptors had been experimentally shown to confer the PG ligand-sensing specificity, we developed three-dimensional structures of hNOD1-LRR and the hNOD2-LRR to understand the mechanism of differential recognition of muramyl peptide ligands by hNOD receptors. The hNOD1-LRR and hNOD2-LRR receptor models exhibited right-handed curved solenoid shape. The hot-spot residues experimentally proved to be critical for ligand recognition were located in the concavity of the NOD-LRR and formed the recognition site. Our molecular docking analyses and molecular electrostatic potential mapping studies explain the activation of hNOD-LRRs, in response to effective molecular interactions of PG ligands at the recognition site; and conversely, the inability of certain PG ligands to activate hNOD-LRRs, by deviations from the recognition site. Based on molecular docking studies using PG ligands, we propose few residues — G825, D826 and N850 in hNOD1-LRR and L904, G905, W931, L932 and S933 in hNOD2-LRR, evolutionarily conserved across different host species, which may play a major role in ligand recognition. Thus, our integrated experimental and computational approach elucidates the molecular basis underlying the differential recognition of PG ligands by hNOD receptors.
Collapse
|
26
|
van Kempen G, Meijvis S, Endeman H, Vlaminckx B, Meek B, de Jong B, Rijkers G, Bos WJ. Mannose-binding lectin and l-ficolin polymorphisms in patients with community-acquired pneumonia caused by intracellular pathogens. Immunology 2017; 151:81-88. [PMID: 28032346 PMCID: PMC5382344 DOI: 10.1111/imm.12705] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/07/2016] [Accepted: 12/18/2016] [Indexed: 01/07/2023] Open
Abstract
Community-acquired pneumonia (CAP) is the leading infectious disease requiring hospitalization in the western world. Genetic variability affecting the host response to infection may play a role in susceptibility and outcome in patients with CAP. Mannose-binding lectin (MBL) and l-ficolin (l-FCN) are two important activators of the complement system and they can enhance phagocytosis by opsonization. In a prospective cohort of 505 Dutch patients with CAP and 227 control participants we studied whether polymorphisms in the MBL (MBL2) and FCN (FCN2) genes influenced susceptibility and outcome. No difference in frequency of these genotypes was found between patients with CAP in general and controls. However, the +6424G>T single nucleotide polymorphism (SNP) in FCN2 was more common in patients with a Coxiella burnetii pneumonia (P = 0·014). Moreover, the haplotypes coding for the highest MBL serum levels (YA/YA and YA/XA) predisposed to atypical pneumonia (C. burnetii, Legionella or Chlamydia species or Mycoplasma pneumoniae) compared with controls (P = 0·016). Furthermore, patients with these haplotypes were more often bacteraemic (P = 0·019). It can therefore be concluded that MBL2 and FCN2 polymorphisms are not major risk factors for CAP in general, but that the +6424G>T SNP in the FCN2 gene predisposes to C. burnetii pneumonia. In addition, patients with genotypes corresponding with high serum MBL levels are at risk for atypical pneumonia, possibly caused by enhanced phagocytosis, thereby promoting cell entry of these intracellular bacteria.
Collapse
Affiliation(s)
- Gijs van Kempen
- Department of Internal MedicineSt Antonius HospitalNieuwegeinThe Netherlands
- Department of Internal MedicineUniversity Medical CentreUtrechtThe Netherlands
| | - Sabine Meijvis
- Department of Internal MedicineSt Antonius HospitalNieuwegeinThe Netherlands
- Department of Internal MedicineUniversity Medical CentreUtrechtThe Netherlands
| | - Henrik Endeman
- Department of Intensive Care MedicineOnze Lieve Vrouwen GasthuisAmsterdamThe Netherlands
| | - Bart Vlaminckx
- Department of Medical Microbiology and ImmunologySt Antonius HospitalNieuwegeinThe Netherlands
| | - Bob Meek
- Department of Medical Microbiology and ImmunologySt Antonius HospitalNieuwegeinThe Netherlands
| | - Ben de Jong
- Department of Medical Microbiology and ImmunologySt Antonius HospitalNieuwegeinThe Netherlands
| | - Ger Rijkers
- Department of Medical Microbiology and ImmunologySt Antonius HospitalNieuwegeinThe Netherlands
- Department of SciencesRoosevelt AcademyMiddelburgThe Netherlands
| | - Willem Jan Bos
- Department of Internal MedicineSt Antonius HospitalNieuwegeinThe Netherlands
| |
Collapse
|
27
|
Mannose-Binding Lectin: Biologic Characteristics and Role in the Susceptibility to Infections and Ischemia-Reperfusion Related Injury in Critically Ill Neonates. J Immunol Res 2017; 2017:7045630. [PMID: 28246614 PMCID: PMC5299167 DOI: 10.1155/2017/7045630] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 11/08/2016] [Accepted: 12/28/2016] [Indexed: 01/14/2023] Open
Abstract
The mannose-binding lectin (MBL) is a member of the collectin family, belonging to the innate immunity system. Genetic, biologic, and clinical properties of MBL have been widely investigated throughout the last decades, although some interesting aspects of its potential clinical relevance are still poorly understood. Low circulating concentrations of MBL have been associated with increased risk of infection and poor neurologic outcome in neonates. On the other hand, an excessive and uncontrolled inflammatory response by the neonatal intestine after the exposure to luminal bacteria, leading to an increased production of MBL, may be involved in the onset of necrotizing enterocolitis. The purpose of the present review is to summarize the current knowledge about genetic and biologic characteristics of MBL and its role in the susceptibility to infections and to ischemia-reperfusion related tissue injuries to better explore its clinical relevance during the perinatal period and the possible future therapeutic applications.
Collapse
|
28
|
Bacterial peptidoglycan with amidated meso-diaminopimelic acid evades NOD1 recognition: an insight into NOD1 structure–recognition. Biochem J 2016; 473:4573-4592. [DOI: 10.1042/bcj20160817] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/04/2016] [Accepted: 10/14/2016] [Indexed: 12/16/2022]
Abstract
Nucleotide-binding oligomerization domain-containing protein 1 (NOD1) is an intracellular pattern recognition receptor that recognizes bacterial peptidoglycan (PG) containing meso-diaminopimelic acid (mesoDAP) and activates the innate immune system. Interestingly, a few pathogenic and commensal bacteria modify their PG stem peptide by amidation of mesoDAP (mesoDAPNH2). In the present study, NOD1 stimulation assays were performed using bacterial PG containing mesoDAP (PGDAP) and mesoDAPNH2 (PGDAPNH2) to understand the differences in their biomolecular recognition mechanism. PGDAP was effectively recognized, whereas PGDAPNH2 showed reduced recognition by the NOD1 receptor. Restimulation of the NOD1 receptor, which was initially stimulated with PGDAP using PGDAPNH2, did not show any further NOD1 activation levels than with PGDAP alone. But the NOD1 receptor initially stimulated with PGDAPNH2 responded effectively to restimulation with PGDAP. The biomolecular structure–recognition relationship of the ligand-sensing leucine-rich repeat (LRR) domain of human NOD1 (NOD1–LRR) with PGDAP and PGDAPNH2 was studied by different computational techniques to further understand the molecular basis of our experimental observations. The d-Glu–mesoDAP motif of GMTPDAP, which is the minimum essential motif for NOD1 activation, was found involved in specific interactions at the recognition site, but the interactions of the corresponding d-Glu–mesoDAP motif of PGDAPNH2 occur away from the recognition site of the NOD1 receptor. Hot-spot residues identified for effective PG recognition by NOD1–LRR include W820, G821, D826 and N850, which are evolutionarily conserved across different host species. These integrated results thus successfully provided the atomic level and biochemical insights on how PGs containing mesoDAPNH2 evade NOD1–LRR receptor recognition.
Collapse
|
29
|
Abstract
Atherosclerosis is a chronic inflammatory disorder characterized by immune cell activation, inflammation driven plaque formation and subsequent destabilization. In other disorders of an inflammatory nature, the chronic inflammatory state per se has been linked to acceleration of the atherosclerotic process which is underlined by an increased incidence of cardiovascular disease (CVD) in disorders such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA) and antiphopholipid (Hughes) syndrome (APS). In addition to systemic inflammation, additional mechanisms have been put forward that are more specific for the pathophysiology of the individual chronic inflammatory disorders.
Collapse
Affiliation(s)
- S I van Leuven
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
30
|
Tang Y, Ma D, Ming S, Zhang L, Zhou J, Shan G, Chen Z, Lu X, Zuo D. Mannan-binding lectin reduces CpG DNA-induced inflammatory cytokine production by human monocytes. Microbiol Immunol 2016; 59:231-7. [PMID: 25664598 DOI: 10.1111/1348-0421.12245] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/28/2015] [Accepted: 02/05/2015] [Indexed: 01/08/2023]
Abstract
Mannan-binding lectin (MBL) belongs to the collectin family and functions as an opsonin that can also initiate complement activation. Our previous study showed that MBL serves as a double-stranded RNA binding protein that attenuates polyriboinosinic-polyribocytidylic acid-induced TLR3 activation. Prompted by these findings, in the present study cross-talk between MBL and CpG-DNA-induced TLR9 activation was investigated. Here, it was found that MBL also interacts with the TLR9 agonist, CpG oligodeoxynucleotide (CpG-ODN), in a calcium-dependent manner. Purified MBL protein suppressed activation of nuclear factor-kappa B signaling and subsequent production of proinflammatory cytokines from human monocytes induced by CpG-ODN 2006. These observations indicate that MBL can down-regulate CpG DNA-induced TLR9 activation, emphasizing the importance of understanding the interaction of MBL with TLR agonist in host immune defense.
Collapse
Affiliation(s)
- Yuan Tang
- Department of Immunology, Southern Medical University, Guangzhou, 510515
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
A high throughput method for quantification of cell surface bound and internalized chitosan nanoparticles. Int J Biol Macromol 2015; 81:858-66. [DOI: 10.1016/j.ijbiomac.2015.09.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 08/15/2015] [Accepted: 09/13/2015] [Indexed: 01/09/2023]
|
32
|
Choi KM, Shim SH, An CM, Nam BH, Jeong JM, Kim JW, Park CI. Functional characterisation and expression analysis of recombinant serum amyloid P isoform 1 (RbSAP1) from rock bream (Oplegnathus fasciatus). FISH & SHELLFISH IMMUNOLOGY 2015; 45:277-285. [PMID: 25917975 DOI: 10.1016/j.fsi.2015.04.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 04/15/2015] [Accepted: 04/18/2015] [Indexed: 06/04/2023]
Abstract
Lectins are carbohydrate-binding proteins that play important roles in the recognition and elimination of pathogens via the innate immune system. Pentraxins (PTX) are humoral lectins, which are multifunctional proteins in vertebrates. Pentraxins can be divided into two groups based on their primary structure: short (C-reactive protein and serum amyloid P [SAP]) and long pentraxins (PTX3 and neuronal pentraxins). Previously, SAP was shown to have Ca(2+)-dependent binding specificity for various ligands and to be a major acute phase protein. In this study, we identified and characterised the gene encoding SAP isoform 1 in rock bream (Oplegnathus fasciatus) (RbSAP1) and analysed its expression in various tissues after a pathogen challenge. An alignment analysis conducted based on the deduced amino acid sequence of RbSAP1 (1918 bp full-length cDNA with a 699 bp open reading frame encoding 232 amino acids) and SAPs and PTXs isolated from other organisms, revealed that the pentraxin domain and cysteine residues of the deduced protein are conserved. RbSAP1, which was ubiquitously expressed in all tissues examined, was predominantly detected in head kidney, trunk kidney, peripheral blood leukocytes, and gills. RbSAP1 expression was dramatically up-regulated in the kidney and liver after infection with Edwardsiella tarda, Streptococcus iniae, or red seabream iridovirus. Purified rRbSAP1 was able to bind Gram-negative bacteria, Gram-positive bacteria, and pathogen-associated molecular patterns. Interestingly, rRbSAP1 aggregated Gram-negative bacteria in the presence of Ca(2+). The anti-pathogen activity of rRbSAP1 suggests that SAP functions in innate immunity in the rock bream.
Collapse
Affiliation(s)
- Kwang-Min Choi
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea
| | - Sang Hee Shim
- College of Pharmacy, Duksung Women's University, 33, Samyang-ro 144-gil, Dobong Gu, Seoul 132-714, Republic of Korea
| | - Cheul Min An
- Biotechnology Research Division, National Fisheries Research and Development Institute, 216 Gijanghaean-ro, Gijang-eup, Gijang-gun, Busan, 619-705, Republic of Korea
| | - Bo-Hye Nam
- Biotechnology Research Division, National Fisheries Research and Development Institute, 216 Gijanghaean-ro, Gijang-eup, Gijang-gun, Busan, 619-705, Republic of Korea
| | - Ji-Min Jeong
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea
| | - Ju-Won Kim
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea
| | - Chan-Il Park
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea.
| |
Collapse
|
33
|
Zhang XH, Shi YH, Chen J. Molecular characterization of a transmembrane C-type lectin receptor gene from ayu (Plecoglossus altivelis) and its effect on the recognition of different bacteria by monocytes/macrophages. Mol Immunol 2015; 66:439-50. [PMID: 26010409 DOI: 10.1016/j.molimm.2015.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 05/06/2015] [Accepted: 05/08/2015] [Indexed: 01/14/2023]
Abstract
C-type lectin receptors (CTLRs) play vital roles in immune responses as pattern-recognition receptors (PRRs). In this study, we identified a novel C-type lectin receptor (PaCTLRC) gene from ayu, Plecoglossus altivelis. Predicted PaCTLRC is a single transmembrane receptor with a typical carbohydrate recognition domain (CRD) at its C-terminus. Sequence comparison and phylogenetic tree analysis showed that PaCTLRC was most closely related to Atlantic salmon (Salmo salar) CLRC, but was significantly different from two other ayu CTLRs, aCLR and PaCD209L. PaCTLRC transcript was detected in all tested tissues and cells, with high levels in the liver; and its expression was significantly altered upon Vibrio anguillarum infection. Refolded recombinant PaCTLRC (rPaCTLRC) agglutinated three types of Gram-positive bacteria (Listeria monocytogenes, Staphylococcus aureus and Streptococcus iniae) and four types of Gram-negative bacteria (Aeromonas hydrophila, Escherichia coli, V. anguillarum and Vibrio parahaemolyticus) in a Ca(2+)-dependent manner in vitro, and Gram-positive bacteria were shown to be biologically relevant ligands for PaCTLRC. rPaCTLRC bound to d-mannose, d-galactose, l-fucose, N-acetyl-d-glucosamine (GlcNAc), lipopolysaccharide (LPS) and peptidoglycan (PGN), exhibiting a relative binding strength to d-mannose and PGN. d-Mannose, l-fucose, GlcNAc, LPS and PGN could inhibit the agglutinating activity of rPaCTLRC, while d-galactose did not functioned. PaCTLRC neutralization using anti-PaCTLRC IgG resulted in the inhibition of phagocytosis by ayu monocytes/macrophages (MO/MΦ) of S. aureus but not of E. coli, and produced a consistently higher survival rate of S. aureus than that of E. coli. d-Mannose, LPS and PGN treatment had no significant influence on the phagocytosis of ayu MO/MΦ. These results suggest that PaCTLRC may serve as a Gram-positive bacteria-preferred PRR which is involved in pathogen recognition and signal transduction in ayu MO/MΦ.
Collapse
Affiliation(s)
- Xue-Heng Zhang
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Yu-Hong Shi
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Jiong Chen
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
34
|
Li M, Li C, Ma C, Li H, Zuo H, Weng S, Chen X, Zeng D, He J, Xu X. Identification of a C-type lectin with antiviral and antibacterial activity from pacific white shrimp Litopenaeus vannamei. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 46:231-240. [PMID: 24792214 DOI: 10.1016/j.dci.2014.04.014] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 04/20/2014] [Accepted: 04/22/2014] [Indexed: 06/03/2023]
Abstract
C-type lectins (CTLs) play crucial roles in innate immune responses in invertebrates by recognizing and eliminating microinvaders. In this study, a CTL from pacific white shrimp Litopenaeus vannamei (LvCTL3) was identified. LvCTL3 contains a single C-type lectin-like domain (CTLD), which shows similarities to those of other shrimp CTLs and has a mutated 'EPD' motif in Ca(2+)-binding site 2. LvCTL3 mRNA can be detected in all tested tissues and expression of LvCTL3 in gills was up-regulated after Lipopolysaccharides, poly (I:C), Vibrio parahaemolyticus and white spot syndrome virus (WSSV) challenges, suggesting activation responses of LvCTL3 to bacterial, virus and immune stimulant challenges. The 5'flanking regulatory region of LvCTL3 was cloned and we identified a NF-κB binding motif in the LvCTL3 promoter region. Dual-luciferase reporter assays indicated that over-expression of L. vannamei dorsal can dramatically up regulate the promoter activity of LvCTL3, suggesting that LvCTL3 expression could be regulated through NF-κB signaling pathway. As far as we know, this is the first report on signaling pathway involve in shrimp CTLs expression. The recombinant LvCTL3 protein was expressed in Escherichia coli and purified by Ni-affinity chromatography. The purified LvCTL3 can agglutinate Gram-negative microbe Vibrio alginolyticus and V. parahaemolyticus and Gram-positive bacteria Bacillus subtilis in the presence of calcium ions, but cannot agglutinate Gram-positive bacteria Streptococcus agalactiae. The agglutination activity of LvCTL3 was abolished when Ca(2+) was chelated with EDTA, suggesting the function of LvCTL3 is Ca(2+)-dependent. In vivo challenge experiments showed that the recombinant LvCTL3 protein can significantly reduce the mortalities of V. parahemolyticus and WSSV infection, indicating LvCTL3 might play significant roles in shrimp innate immunity defense against bacterial and viral infection.
Collapse
Affiliation(s)
- Ming Li
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Institute of Fisheries, Nanning, PR China
| | - Chaozheng Li
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Chunxia Ma
- Guangxi Veterinary Research Institute, Guangxi University, Nanning, PR China
| | - Haoyang Li
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Hongliang Zuo
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Shaoping Weng
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Xiaohan Chen
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Institute of Fisheries, Nanning, PR China
| | - Digang Zeng
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Institute of Fisheries, Nanning, PR China
| | - Jianguo He
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China.
| | - Xiaopeng Xu
- MOE Key Laboratory of Aquatic Product Safety/State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China.
| |
Collapse
|
35
|
van Kessel KPM, Bestebroer J, van Strijp JAG. Neutrophil-Mediated Phagocytosis of Staphylococcus aureus. Front Immunol 2014; 5:467. [PMID: 25309547 PMCID: PMC4176147 DOI: 10.3389/fimmu.2014.00467] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 09/12/2014] [Indexed: 01/13/2023] Open
Abstract
Initial elimination of invading Staphylococcus aureus from the body is mediated by professional phagocytes. The neutrophil is the major phagocyte of the innate immunity and plays a key role in the host defense against staphylococcal infections. Opsonization of the bacteria with immunoglobulins and complement factors enables efficient recognition by the neutrophil that subsequently leads to intracellular compartmentalization and killing. Here, we provide a review of the key processes evolved in neutrophil-mediated phagocytosis of S. aureus and briefly describe killing. As S. aureus is not helpless against the professional phagocytes, we will also highlight its immune evasion arsenal related to phagocytosis.
Collapse
Affiliation(s)
- Kok P M van Kessel
- Medical Microbiology, University Medical Center Utrecht , Utrecht , Netherlands
| | - Jovanka Bestebroer
- Medical Microbiology, University Medical Center Utrecht , Utrecht , Netherlands
| | - Jos A G van Strijp
- Medical Microbiology, University Medical Center Utrecht , Utrecht , Netherlands
| |
Collapse
|
36
|
Xiao W, Meng G, Zhao Y, Yuan H, Li T, Peng Y, Zhao Y, Luo M, Zhao W, Li Z, Zheng X. Human secreted stabilin-1-interacting chitinase-like protein aggravates the inflammation associated with rheumatoid arthritis and is a potential macrophage inflammatory regulator in rodents. Arthritis Rheumatol 2014; 66:1141-52. [PMID: 24470346 DOI: 10.1002/art.38356] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 01/07/2014] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To study the relationship between the human secreted protein stabilin-1-interacting chitinase-like protein (SI-CLP) and rheumatoid arthritis (RA). METHODS The expression of SI-CLP in peripheral blood mononuclear cells (PBMCs) and synovial fluid from patients with RA and the effects of cytokines on SI-CLP expression were examined by Western blotting. Fluorescence-activated cell sorting analysis was performed to investigate the binding between SI-CLP and cells. Bone marrow-derived macrophages were isolated from wild-type and SI-CLP(-/-) mice, and real-time quantitative polymerase chain reaction was performed to detect the levels of messenger RNA for cytokines or SI-CLP in SI-CLP- or cytokine-treated macrophages. Histologic studies were conducted to evaluate inflammation and the expression of interleukin-12 (IL-12), IL-13, and SI-CLP in lesions. Enzyme-linked immunosorbent assays were used to detect the cytokine levels in bone marrow-derived macrophages. Rats or mice with collagen-induced arthritis (CIA) and SI-CLP(-/-) mice were used to study the function of SI-CLP in RA. RESULTS SI-CLP expression was increased in PBMCs and detectable in synovial fluid from patients with RA. Administration of SI-CLP to rats with CIA aggravated arthritis-associated inflammation. SI-CLP was specifically attached to the surface protein of macrophages, which elevated the expression of IL-1β, IL-6, IL-12, and IL-13 in macrophages and mouse bone marrow-derived macrophages, up-regulating ERK phosphorylation. Moreover, SI-CLP was up-regulated by both IL-12 and IL-13 through JNK and JAK/STAT signaling, respectively. Knockout of SI-CLP resulted in a decrease in the expression of IL-1β, IL-6, IL-12, and IL-13 and lower susceptibility to CIA compared with wild-type mice. SI-CLP treatment also aggravated arthritis-related inflammation in wild-type and SI-CLP(-/-) mice. CONCLUSION SI-CLP functions as a regulator of the inflammatory response by macrophages. The decrease in inflammation-associated cytokine levels resulting from SI-CLP knockout may explain the lower susceptibility to CIA in SI-CLP(-/-) mice.
Collapse
Affiliation(s)
- Weichun Xiao
- State Key Lab of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hijikata M, Matsushita I, Hang NTL, Maeda S, Thuong PH, Tam DB, Shimbo T, Sakurada S, Cuong VC, Lien LT, Keicho N. Age-dependent association of mannose-binding lectin polymorphisms with the development of pulmonary tuberculosis in Viet Nam. Hum Immunol 2014; 75:840-6. [PMID: 24952212 DOI: 10.1016/j.humimm.2014.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 06/05/2014] [Accepted: 06/07/2014] [Indexed: 11/16/2022]
Abstract
Mannose-binding lectin (MBL) binds to pathogens and induces complement-mediated opsonophagocytosis. Although the association between MBL2 polymorphisms and tuberculosis (TB) has been studied in various populations, the results are controversial. We explored the stages of TB associated with MBL2 polymorphisms. X/Y (rs7096206) and A/B (rs1800450) were genotyped in 765 new patients with active pulmonary TB without HIV infection and 556 controls in Hanoi, Viet Nam. The MBL2 nucleotide sequences were further analyzed, and plasma MBL levels were measured in 109 apparently healthy healthcare workers and 65 patients with TB. Latent TB infection (LTBI) was detected by interferon-gamma release assay (IGRA). The YA/YA diplotype, which exhibited high plasma MBL levels, was associated with protection against active TB in younger patients (mean age = 32)≦ 45 years old (odds ratio, 0.61; 95% confidence interval, 0.46-0.80). The resistant diplotype was less frequently found in the younger patients at diagnosis (P = 0.0021). MBL2 diplotype frequencies and plasma MBL levels were not significantly different between the IGRA-positive and -negative groups. MBL2 YA/YA exhibited a protective role against the development of TB in younger patients, whereas the MBL2 genotype and MBL levels were not associated with LTBI. High MBL levels may protect against the early development of pulmonary TB after infection.
Collapse
Affiliation(s)
- Minako Hijikata
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo 204-8533, Japan; National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Ikumi Matsushita
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo 204-8533, Japan
| | | | - Shinji Maeda
- Department of Mycobacterium Reference and Research, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo 204-8533, Japan
| | | | - Do Bang Tam
- Department of Biochemistry, Hematology and Blood Transfusion, Hanoi Lung Hospital, Hanoi, Viet Nam
| | - Takuro Shimbo
- Department of Clinical Study and Informatics, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Shinsaku Sakurada
- National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | | | | | - Naoto Keicho
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo 204-8533, Japan; National Center for Global Health and Medicine, Tokyo 162-8655, Japan.
| |
Collapse
|
38
|
Sivakamavalli J, Vaseeharan B. Purification, characterization and functional role of lectin from green tiger shrimp Penaeus semisulcatus. Int J Biol Macromol 2014; 67:64-70. [DOI: 10.1016/j.ijbiomac.2014.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 03/05/2014] [Accepted: 03/07/2014] [Indexed: 10/25/2022]
|
39
|
Prognostic value of mannose-binding lectin: 90-day outcome in patients with acute ischemic stroke. Mol Neurobiol 2014; 51:230-9. [PMID: 24691546 DOI: 10.1007/s12035-014-8682-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 03/12/2014] [Indexed: 01/04/2023]
Abstract
Complement activation and inflammation have been suggested in the pathogenesis of stroke; mannose-binding lectin (MBL) was found to have roles during the process. We therefore evaluated the short-term prognostic value of serum MBL in Chinese patients with an acute ischemic stroke (AIS). Consecutive AIS patients admitted to the emergency department were identified. Clinical information was collected. Serum concentration of MBL and NIH Stroke Scale (NIHSS) was measured at the time of admission. Short-term functional outcome was measured by modified Rankin scale (mRS) 90 days after admission. Multivariate analyses were performed using logistic regression models. During the inclusion period, 231 patients were diagnosed with AIS, and 220 completed follow-up. The results indicated that the serum MBL levels were significantly (P = 0.000) higher in acutely ischemic stroke patients as compared with normal controls. MBL was an independent prognostic marker of short-term functional outcome and death (odds ratio (OR) 5.28 (2.88-10.67) and 6.99 (3.55-13.97), respectively, P = 0.000 for both, adjusted for NIHSS, other predictors, and vascular risk factors) in patients with AIS. MBL improved the area under the receiver operating characteristic curve of the NIHSS score for functional outcome from 0.826 (95 % CI 0.773-0.879) to 0.857 (95 % CI 0.808-0.905, P = 0.000) and for mortality from 0.768 (95 % CI 0.682-0.853) to 0.822 (95 % CI 0.747-0.896, P = 0.000). Serum MBL levels are a useful, complementary tool to predict functional outcome and mortality 90 days after stroke.
Collapse
|
40
|
Liu H, Zhou J, Ma D, Lu X, Ming S, Shan G, Zhang X, Hou J, Chen Z, Zuo D. Mannan binding lectin attenuates double-stranded RNA-mediated TLR3 activation and innate immunity. FEBS Lett 2014; 588:866-72. [PMID: 24530528 DOI: 10.1016/j.febslet.2014.01.064] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 01/21/2014] [Indexed: 01/04/2023]
Abstract
Mannan binding lectin (MBL) functions as a pattern recognition molecule (PRM) which is able to initiate complement activation. Here, we characterize a previously unrecognized attribute of MBL as a double-stranded RNA (dsRNA) binding protein capable of modifying Toll like receptor 3 (TLR3) activation. MBL interacts with poly(I:C) and suppresses poly(I:C)-induced activation of TLR3 pathways and subsequent cytokine production. In addition, MBL binds to TLR3 directly. Surprisingly, disrupting the interaction between MBL and complement receptor 1 (CR1) or restraining the traffic of MBL to phagosome reversed the MBL limited TLR3 activation. We demonstrate the importance of MBL guided ligands intracellular localization, emphasizing the significance of understanding the dynamics of TLR agonists complexed with MBL or other PRMs inside the cell in immune defense.
Collapse
Affiliation(s)
- Hongzhi Liu
- Department of Immunology, Southern Medical University, Guangzhou 510515, China
| | - Jia Zhou
- Department of Immunology, Southern Medical University, Guangzhou 510515, China
| | - Di Ma
- Department of Immunology, Southern Medical University, Guangzhou 510515, China
| | - Xiao Lu
- Department of Immunology, Southern Medical University, Guangzhou 510515, China
| | - Siqi Ming
- Department of Immunology, Southern Medical University, Guangzhou 510515, China
| | - Guiqiu Shan
- Department of Immunology, Southern Medical University, Guangzhou 510515, China; Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, China
| | - Xiaoyong Zhang
- Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China
| | - Jinlin Hou
- Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China
| | - Zhengliang Chen
- Department of Immunology, Southern Medical University, Guangzhou 510515, China.
| | - Daming Zuo
- Department of Immunology, Southern Medical University, Guangzhou 510515, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
41
|
An JH, Kurokawa K, Jung DJ, Kim MJ, Kim CH, Fujimoto Y, Fukase K, Coggeshall KM, Lee BL. Human SAP is a novel peptidoglycan recognition protein that induces complement-independent phagocytosis of Staphylococcus aureus. THE JOURNAL OF IMMUNOLOGY 2013; 191:3319-27. [PMID: 23966633 DOI: 10.4049/jimmunol.1300940] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The human pathogen Staphylococcus aureus is responsible for many community-acquired and hospital-associated infections and is associated with high mortality. Concern over the emergence of multidrug-resistant strains has renewed interest in the elucidation of host mechanisms that defend against S. aureus infection. We recently demonstrated that human serum mannose-binding lectin binds to S. aureus wall teichoic acid (WTA), a cell wall glycopolymer--a discovery that prompted further screening to identify additional serum proteins that recognize S. aureus cell wall components. In this report, we incubated human serum with 10 different S. aureus mutants and determined that serum amyloid P component (SAP) bound specifically to a WTA-deficient S. aureus ΔtagO mutant, but not to tagO-complemented, WTA-expressing cells. Biochemical characterization revealed that SAP recognizes bacterial peptidoglycan as a ligand and that WTA inhibits this interaction. Although SAP binding to peptidoglycan was not observed to induce complement activation, SAP-bound ΔtagO cells were phagocytosed by human polymorphonuclear leukocytes in an FcγR-dependent manner. These results indicate that SAP functions as a host defense factor, similar to other peptidoglycan recognition proteins and nucleotide-binding oligomerization domain-like receptors.
Collapse
Affiliation(s)
- Jang-Hyun An
- Host Defense Protein Laboratory, College of Pharmacy, Pusan National University, Busan 609-735, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Sukhithasri V, Nisha N, Biswas L, Anil Kumar V, Biswas R. Innate immune recognition of microbial cell wall components and microbial strategies to evade such recognitions. Microbiol Res 2013; 168:396-406. [DOI: 10.1016/j.micres.2013.02.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 02/08/2013] [Accepted: 02/09/2013] [Indexed: 01/19/2023]
|
43
|
Ma YJ, Kang HJ, Kim JY, Garred P, Lee MS, Lee BL. Mouse mannose-binding lectin-A and ficolin-A inhibit lipopolysaccharide-mediated pro-inflammatory responses on mast cells. BMB Rep 2013; 46:376-81. [PMID: 23884105 PMCID: PMC4133918 DOI: 10.5483/bmbrep.2013.46.7.055] [Citation(s) in RCA: 14] [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: 03/11/2013] [Revised: 03/12/2013] [Accepted: 03/12/2013] [Indexed: 01/08/2023] Open
Abstract
It is unknown how soluble pattern-recognition receptors in blood, such as mannose-binding lectin (MBL) and ficolins, modulate mast cell-mediated inflammatory responses. We investigate how mouse MBL-A or ficolin-A regulate mouse bone marrow-derived mast cells (mBMMCs)-derived inflammatory response against bacterial lipopolysaccharide (LPS) stimulation. LPS-mediated pro-inflammatory cytokine productions on mBMMCs obtained from Toll-like receptor4 (TLR4)-deficient mice, TLR2-defficient mice, and their wildtype, were specifically attenuated by the addition of either mouse MBL-A or ficolin-A in a dose-dependent manner. However, the inhibitory effects by mouse MBL-A or ficolin-A were restored by the addition of mannose or N-acetylglucosamine, respectively. These results suggest that mouse MBL-A and ficolin-A bind to LPS via its carbohydrate-recognition domain and fibrinogen-like domain, respectively, whereby cytokine production by LPS-mediated TLR4 in mBMMCs appears to be down-regulated, indicating that mouse MBL and ficolin may have an inhibitory function toward mouse TLR4-mediated excessive inflammation on the mast cells.
Collapse
Affiliation(s)
- Ying Jie Ma
- The Global Research Laboratory of Insect Symbiosis, College of Pharmacy, Pusan National University, Busan 609-735, Korea
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Sect 7631, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hee Jung Kang
- Department of Laboratory Medicine, Hallym University College of Medicine, Anyang 431-070, Korea
| | - Ji Yeon Kim
- Department of Laboratory Medicine, Hallym University College of Medicine, Anyang 431-070, Korea
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Sect 7631, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Myung-Shik Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | - Bok Luel Lee
- The Global Research Laboratory of Insect Symbiosis, College of Pharmacy, Pusan National University, Busan 609-735, Korea
| |
Collapse
|
44
|
Cai Y, Zhang W, Xiong S. Mannose-binding lectin blunts macrophage polarization and ameliorates lupus nephritis. PLoS One 2013; 8:e62465. [PMID: 23626823 PMCID: PMC3633861 DOI: 10.1371/journal.pone.0062465] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 03/20/2013] [Indexed: 12/31/2022] Open
Abstract
Background Deficiency in clearance of self nuclear antigens, including DNA, is the hallmark of systemic lupus erythematosus (SLE), a chronic autoimmnue disease characterized by the production of various autoantibodies, immune complex deposition and severe organ damage. Our previous studies revealed that administration of syngeneic BALB/c mice with activated lymphocyte-derived DNA (ALD-DNA) could induce SLE disease. Mannose-binding lectin (MBL), a secreted pattern recognition receptor with binding activity to DNA, has been proved to be a modulator of inflammation, but whether MBL takes responsibility for DNA clearance, modulates the DNA-mediated immune responses, and is involved in the development of DNA-induced SLE disease remain poorly understood. Methodology/Principal Findings The levels of serum MBL significantly decreased in lupus mice induced by ALD-DNA and were negatively correlated with SLE disease. MBL blunted macrophage M2b polarization by inhibiting the MAPK and NF-κB signaling while enhancing the activation of CREB. Furthermore, MBL suppressed the ability of ALD-DNA–stimulated macrophages to polarize T cells toward Th1 cells and Th17 cells. Importantly, MBL supplement in vivo could ameliorate lupus nephritis. Conclusion/Significance These results suggest MBL supplement could alleviate SLE disease and might imply a potential therapeutic strategy for DNA-induced SLE, which would further our understanding of the protective role of MBL in SLE disease.
Collapse
Affiliation(s)
- Yanxing Cai
- Department of Immunology and Institute for Immunobiology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Weijuan Zhang
- Department of Immunology and Institute for Immunobiology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Sidong Xiong
- Department of Immunology and Institute for Immunobiology, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People’s Republic of China
- * E-mail:
| |
Collapse
|
45
|
Low mannose-binding lectin serum levels are associated with reduced kidney graft survival. Kidney Int 2013; 83:264-71. [DOI: 10.1038/ki.2012.373] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
46
|
Geiss-Liebisch S, Rooijakkers SHM, Beczala A, Sanchez-Carballo P, Kruszynska K, Repp C, Sakinc T, Vinogradov E, Holst O, Huebner J, Theilacker C. Secondary cell wall polymers of Enterococcus faecalis are critical for resistance to complement activation via mannose-binding lectin. J Biol Chem 2012; 287:37769-77. [PMID: 22908219 DOI: 10.1074/jbc.m112.358283] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The complement system is part of our first line of defense against invading pathogens. The strategies used by Enterococcus faecalis to evade recognition by human complement are incompletely understood. In this study, we identified an insertional mutant of the wall teichoic acid (WTA) synthesis gene tagB in E. faecalis V583 that exhibited an increased susceptibility to complement-mediated killing by neutrophils. Further analysis revealed that increased killing of the mutant was due to a higher rate of phagocytosis by neutrophils, which correlated with higher C3b deposition on the bacterial surface. Our studies indicated that complement activation via the lectin pathway was much stronger on the tagB mutant compared with wild type. In concordance, we found an increased binding of the key lectin pathway components mannose-binding lectin and mannose-binding lectin-associated serine protease-2 (MASP-2) on the mutant. To understand the mechanism of lectin pathway inhibition by E. faecalis, we purified and characterized cell wall carbohydrates of E. faecalis wild type and V583ΔtagB. NMR analysis revealed that the mutant strain lacked two WTAs with a repeating unit of →6)[α-l-Rhap-(1→3)]β-D-GalpNAc-(1→5)-Rbo-1-P and →6) β-D-Glcp-(1→3) [α-D-Glcp-(1→4)]-β-D-GalpNAc-(1→5)-Rbo-1-P→, respectively (Rbo, ribitol). In addition, compositional changes in the enterococcal rhamnopolysaccharide were noticed. Our study indicates that in E. faecalis, modification of peptidoglycan by secondary cell wall polymers is critical to evade recognition by the complement system.
Collapse
Affiliation(s)
- Stefan Geiss-Liebisch
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Breisacher Strasse 117, 79106 Freiburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
A step-by-step approach to study the influence of N-acetylation on the adjuvanticity of N,N,N-trimethyl chitosan (TMC) in an intranasal nanoparticulate influenza virus vaccine. Eur J Pharm Sci 2012; 45:467-74. [DOI: 10.1016/j.ejps.2011.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 09/20/2011] [Accepted: 10/04/2011] [Indexed: 11/15/2022]
|
48
|
Ling MT, Tu W, Han Y, Mao H, Chong WP, Guan J, Liu M, Lam KT, Law HKW, Peiris JSM, Takahashi K, Lau YL. Mannose-binding lectin contributes to deleterious inflammatory response in pandemic H1N1 and avian H9N2 infection. J Infect Dis 2011; 205:44-53. [PMID: 22080095 PMCID: PMC3242741 DOI: 10.1093/infdis/jir691] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Mannose-binding lectin (MBL) is a pattern-recognition molecule, which functions as a first line of host defense. Pandemic H1N1 (pdmH1N1) influenza A virus caused massive infection in 2009 and currently circulates worldwide. Avian influenza A H9N2 (H9N2/G1) virus has infected humans and has the potential to be the next pandemic virus. Antiviral function and immunomodulatory role of MBL in pdmH1N1 and H9N2/G1 virus infection have not been investigated. METHODS In this study, MBL wild-type (WT) and MBL knockout (KO) murine models were used to examine the role of MBL in pdmH1N1 and H9N2/G1 virus infection. RESULTS Our study demonstrated that in vitro, MBL binds to pdmH1N1 and H9N2/G1 viruses, likely via the carbohydrate recognition domain of MBL. Wild-type mice developed more severe disease, as evidenced by a greater weight loss than MBL KO mice during influenza virus infection. Furthermore, MBL WT mice had enhanced production of proinflammatory cytokines and chemokines compared with MBL KO mice, suggesting that MBL could upregulate inflammatory responses that may potentially worsen pdmH1N1 and H9N2/G1 virus infections. CONCLUSIONS Our study provided the first in vivo evidence that MBL may be a risk factor during pdmH1N1 and H9N2/G1 infection by upregulating proinflammatory response.
Collapse
Affiliation(s)
- Man To Ling
- Department of Paediatrics and Adolescent Medicine, University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Wang TH, Kung YL, Lee MH, Su NW. N-acetyl-D-galactosamine-specific lectin isolated from the seeds of Carica papaya. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:4217-4224. [PMID: 21405109 DOI: 10.1021/jf104962g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
N-Acetyl-D-galactosamine (GalNAc)-specific lectins are of great interest because they have been reported to detect tumor-associated antigens of malignant cells. We isolated a novel lectin from Carica papaya seeds, named C. papaya lectin (CPL). Purification of the lectin involved ammonium sulfate fractionation and DEAE anion exchange and repeated gel filtration chromatography. Inhibition of CPL causing hemagglutination on human erythrocytes showed that the lectin shows specificity to GalNAc and lactose. Surface plasmon resonance further revealed that the lectin possesses high specificity toward GalNAc with a dissociation constant of 5.5 × 10(-9) M. The lectin is composed of 38- and 40-kDa subunits with a molecular mass of ∼804 kDa estimated by size-exclusion high-performance liquid chromatography. Incubation of CPL with Jurkat T cells showed significant induction of IL-2 cytokine, which suggests that CPL has potent immunomodulatory effects on immune cells.
Collapse
Affiliation(s)
- Teng-Hsu Wang
- Department of Agricultural Chemistry, National Taiwan University, Taipei, Taiwan
| | | | | | | |
Collapse
|
50
|
Tukhvatulin AI, Logunov DY, Shcherbinin DN, Shmarov MM, Naroditsky BS, Gudkov AV, Gintsburg AL. Toll-like receptors and their adapter molecules. BIOCHEMISTRY (MOSCOW) 2011; 75:1098-114. [PMID: 21077829 DOI: 10.1134/s0006297910090038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Toll-like receptors (TLR) are among key receptors of the innate mammalian immune system. Receptors of this family are able to recognize specific highly conserved molecular regions (patterns) in pathogen structures, thus initiating reactions of both innate and acquired immune response finally resulting in the elimination of the pathogen. In this case every individual TLR type is able to bind a broad spectrum of molecules of microbial origin characterized by different chemical properties and structures. Recent data demonstrate the existence of a multistep mechanism of the TLR recognition of the pathogen in which, in addition to receptors proper, the involvement of different adapter molecules is necessary. However, functions of separate adapter molecules as well as the principles of formation of a multicomponent system of ligand-specific recognition are still not quite understandable. We describe all identified as well as possible (candidate) adapter TLR molecules by giving their brief characteristics, and we also propose generalized possible variants of the TLR ligand-specific recognition with involvement of adapter molecules.
Collapse
Affiliation(s)
- A I Tukhvatulin
- Gamaleya Institute of Epidemiology and Microbiology, Russian Academy of Medical Sciences, Moscow, 123098, Russia.
| | | | | | | | | | | | | |
Collapse
|