1
|
Gao C, Sang L, Sun S, Chen D, Wang J, Xie X. Effects of kudzu vine (Pueraria lobata) on the intestinal microflora and volatile fatty acids in meat rabbits. Trop Anim Health Prod 2025; 57:70. [PMID: 39964604 PMCID: PMC11836158 DOI: 10.1007/s11250-025-04313-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 01/31/2025] [Indexed: 02/21/2025]
Abstract
Adding flavonoid-rich kudzu vine (Pueraria lobata) meal to animal feed can improve intestinal microflora. Here, we investigated the effects of kudzu vine meal on the growth performance, caecal microbial diversity, caecal microbial number, intestinal pH, and volatile fatty acids (VFAs) of meat rabbits. Two hundred weaned Ira rabbits were randomly divided into four groups and fifty rabbits in each group. Meat rabbits in the control group (group A) were fed a basal diet without kudzu vine meal; those in the three test groups were fed experimental diets comprising 15% (group B), 25% (group C), and 35% (group D) kudzu vine meal. The results showed that the daily feed intake and mortality of meat rabbits decreased significantly after adding kudzu vine meal to their diet. The feed-to-gain ratio and mortality rate in 35% kudzu vine dietary were 3.5 and 7.5%, respectively, significantly lower than those in the control group. The Shannon and Chao 1 index of the bacterial diversity decreased significantly after adding kudzu vine meal to their diet. With the increasing amount of kudzu vine meal, the variety of dominant phyla did not change, but the relative abundance increased with the increase of kudzu vine addition. The intestinal total VFAs, acetic acid and propionic acid were the highest in 15% kudzu vine dietary, and significantly higher than those of 25% kudzu vine dietary. The content of total bacteria in 35% kudzu vine dietary was the lowest and the Escherichia coli and Salmonella showed a decreasing trend. In conclusion, our study confirms the adding kudzu vine meal significantly decreased the mortality rates and feed-to-gain ratio of meat rabbits, and fed diets with 25%-35% kudzu vine meal are recommended to achieve optimal results in meat rabbits.
Collapse
Affiliation(s)
- Chengfang Gao
- Fujian Key Laboratory of Animal Genetics and Breeding, Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China
| | - Lei Sang
- Fujian Key Laboratory of Animal Genetics and Breeding, Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China
| | - Shikun Sun
- Fujian Key Laboratory of Animal Genetics and Breeding, Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China
| | - Dongjin Chen
- Fujian Key Laboratory of Animal Genetics and Breeding, Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China
| | - Jinxiang Wang
- Fujian Key Laboratory of Animal Genetics and Breeding, Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China
| | - Xiping Xie
- Fujian Key Laboratory of Animal Genetics and Breeding, Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, 350013, China.
| |
Collapse
|
2
|
Russell MW, Kilian M, Mestecky J. Role of IgA1 protease-producing bacteria in SARS-CoV-2 infection and transmission: a hypothesis. mBio 2024; 15:e0083324. [PMID: 39207101 PMCID: PMC11492985 DOI: 10.1128/mbio.00833-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Secretory (S) IgA antibodies against severe acute respiratory syndrome (SARS)-CoV-2 are induced in saliva and upper respiratory tract (URT) secretions by natural infection and may be critical in determining the outcome of initial infection. Secretory IgA1 (SIgA1) is the predominant isotype of antibodies in these secretions. Neutralization of SARS-CoV-2 is most effectively accomplished by polymeric antibodies such as SIgA. We hypothesize that cleavage of SIgA1 antibodies against SARS-CoV-2 by unique bacterial IgA1 proteases to univalent Fabα antibody fragments with diminished virus neutralizing activity would facilitate the descent of the virus into the lungs to cause serious disease and also enhance its airborne transmission to others. Recent studies of the nasopharyngeal microbiota of patients with SARS-CoV-2 infection have revealed significant increases in the proportions of IgA1 protease-producing bacteria in comparison with healthy subjects. Similar considerations might apply also to other respiratory viral infections including influenza, possibly explaining the original attribution of influenza to Haemophilus influenzae, which produces IgA1 protease.
Collapse
Affiliation(s)
- Michael W. Russell
- Department of
Microbiology and Immunology, Jacobs School of Medicine and Biomedical
Sciences, University at Buffalo,
Buffalo, New York, USA
| | - Mogens Kilian
- Department of
Biomedicine, Aarhus University,
Aarhus, Denmark
| | - Jiri Mestecky
- Department of
Microbiology, Heersink School of Medicine, University of Alabama at
Birmingham, Birmingham,
Alabama, USA
- />Institute of
Microbiology, laboratory of Cellular and Molecular Immunology, Czech
Academy of Sciences,
Prague, Czechia
| |
Collapse
|
3
|
Yokota C, Fujimoto K, Yamakawa N, Kono M, Miyaoka D, Shimohigoshi M, Uematsu M, Watanabe M, Kamei Y, Sugimoto A, Kawasaki N, Yabuno T, Okamura T, Kuroda E, Hamaguchi S, Sato S, Hotomi M, Akeda Y, Ishii KJ, Yasutomi Y, Sunami K, Uematsu S. Prime-boost-type PspA3 + 2 mucosal vaccine protects cynomolgus macaques from intratracheal challenge with pneumococci. Inflamm Regen 2023; 43:55. [PMID: 37964391 PMCID: PMC10647109 DOI: 10.1186/s41232-023-00305-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/19/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Although vaccination is recommended for protection against invasive pneumococcal disease, the frequency of pneumococcal pneumonia is still high worldwide. In fact, no vaccines are effective for all pneumococcal serotypes. Fusion pneumococcal surface protein A (PspA) has been shown to induce a broad range of cross-reactivity with clinical isolates and afford cross-protection against pneumococcal challenge in mice. Furthermore, we developed prime-boost-type mucosal vaccines that induce both antigen-specific IgG in serum and antigen-specific IgA in targeted mucosal organs in previous studies. We investigated whether our prime-boost-type immunization with a fusion PspA was effective against pneumococcal infection in mice and cynomolgus macaques. METHODS C57BL/6 mice were intramuscularly injected with fusion PspA combined with CpG oligodeoxynucleotides and/or curdlan. Six weeks later, PspA was administered intranasally. Blood and bronchoalveolar lavage fluid were collected and antigen-specific IgG and IgA titers were measured. Some mice were given intranasal Streptococcus pneumoniae and the severity of infection was analyzed. Macaques were intramuscularly injected with fusion PspA combined with CpG oligodeoxynucleotides and/or curdlan at week 0 and week 4. Then, 13 or 41 weeks later, PspA was administered intratracheally. Blood and bronchoalveolar lavage fluid were collected and antigen-specific IgG and IgA titers were measured. Some macaques were intranasally administered S. pneumoniae and analyzed for the severity of pneumonia. RESULTS Serum samples from mice and macaques injected with antigens in combination with CpG oligodeoxynucleotides and/or curdlan contained antigen-specific IgG. Bronchial samples contained antigen-specific IgA after the fusion PspA boosting. This immunization regimen effectively prevented S. pneumoniae infection. CONCLUSIONS Prime-boost-type immunization with a fusion PspA prevented S. pneumoniae infection in mice and macaques.
Collapse
Affiliation(s)
- Chieko Yokota
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Kosuke Fujimoto
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Division of Metagenome Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Natsuko Yamakawa
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan
| | - Masamitsu Kono
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Daichi Miyaoka
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Masaki Shimohigoshi
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Miho Uematsu
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Miki Watanabe
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yukari Kamei
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Akira Sugimoto
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Natsuko Kawasaki
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Takato Yabuno
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Tomotaka Okamura
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan
| | - Eisuke Kuroda
- Department of Infection Control and Prevention, Graduate School of Medicine, Osaka University, Osaka, Japan
- Division of Fostering Required Medica Human Resources, Center for Infectious Diseases Education and Research (CiDER), Osaka University, Osaka, Japan
| | - Shigeto Hamaguchi
- Department of Infection Control and Prevention, Graduate School of Medicine, Osaka University, Osaka, Japan
- Division of Fostering Required Medica Human Resources, Center for Infectious Diseases Education and Research (CiDER), Osaka University, Osaka, Japan
| | - Shintaro Sato
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Microbiology and Immunology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| | - Muneki Hotomi
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Yukihiro Akeda
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ken J Ishii
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yasuhiro Yasutomi
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan
| | - Kishiko Sunami
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Satoshi Uematsu
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan.
- Division of Metagenome Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Research Institute for Drug Discovery Science, Osaka Metropolitan University, Osaka, Japan.
- International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan.
| |
Collapse
|
4
|
Redzic JS, Rahkola J, Tran N, Holyoak T, Lee E, Martín-Galiano AJ, Meyer N, Zheng H, Eisenmesser E. A substrate-induced gating mechanism is conserved among Gram-positive IgA1 metalloproteases. Commun Biol 2022; 5:1190. [PMID: 36336763 PMCID: PMC9637739 DOI: 10.1038/s42003-022-04173-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022] Open
Abstract
The mucosal adaptive immune response is dependent on the production of IgA antibodies and particularly IgA1, yet opportunistic bacteria have evolved mechanisms to specifically block this response by producing IgA1 proteases (IgA1Ps). Our lab was the first to describe the structures of a metal-dependent IgA1P (metallo-IgA1P) produced from Gram-positive Streptococcus pneumoniae both in the absence and presence of its IgA1 substrate through cryo-EM single particle reconstructions. This prior study revealed an active-site gating mechanism reliant on substrate-induced conformational changes to the enzyme that begged the question of whether such a mechanism is conserved among the wider Gram-positive metallo-IgA1P subfamily of virulence factors. Here, we used cryo-EM to characterize the metallo-IgA1P of a more distantly related family member from Gemella haemolysans, an emerging opportunistic pathogen implicated in meningitis, endocarditis, and more recently bacteremia in the elderly. While the substrate-free structures of these two metallo-IgA1Ps exhibit differences in the relative starting positions of the domain responsible for gating substrate, the enzymes have similar domain orientations when bound to IgA1. Together with biochemical studies that indicate these metallo-IgA1Ps have similar binding affinities and activities, these data indicate that metallo-IgA1P binding requires the specific IgA1 substrate to open the enzymes for access to their active site and thus, largely conform to an "induced fit" model.
Collapse
Affiliation(s)
- Jasmina S Redzic
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, School of Medicine, Aurora, CO, 80045, USA
| | - Jeremy Rahkola
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Disease, University of Colorado Denver School of Medicine and Denver Veterans Affairs Medical Center, Aurora, CO, 80045, USA
| | - Norman Tran
- Department of Biology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Todd Holyoak
- Department of Biology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Eunjeong Lee
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, School of Medicine, Aurora, CO, 80045, USA
| | | | - Nancy Meyer
- Pacific Northwest Cryo-EM Center, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Hongjin Zheng
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, School of Medicine, Aurora, CO, 80045, USA
| | - Elan Eisenmesser
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
5
|
Russell MW, Mestecky J. Mucosal immunity: The missing link in comprehending SARS-CoV-2 infection and transmission. Front Immunol 2022; 13:957107. [PMID: 36059541 PMCID: PMC9428579 DOI: 10.3389/fimmu.2022.957107] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/27/2022] [Indexed: 12/21/2022] Open
Abstract
SARS-CoV-2 is primarily an airborne infection of the upper respiratory tract, which on reaching the lungs causes the severe acute respiratory disease, COVID-19. Its first contact with the immune system, likely through the nasal passages and Waldeyer's ring of tonsils and adenoids, induces mucosal immune responses revealed by the production of secretory IgA (SIgA) antibodies in saliva, nasal fluid, tears, and other secretions within 4 days of infection. Evidence is accumulating that these responses might limit the virus to the upper respiratory tract resulting in asymptomatic infection or only mild disease. The injectable systemic vaccines that have been successfully developed to prevent serious disease and its consequences do not induce antibodies in mucosal secretions of naïve subjects, but they may recall SIgA antibody responses in secretions of previously infected subjects, thereby helping to explain enhanced resistance to repeated (breakthrough) infection. While many intranasally administered COVID vaccines have been found to induce potentially protective immune responses in experimental animals such as mice, few have demonstrated similar success in humans. Intranasal vaccines should have advantage over injectable vaccines in inducing SIgA antibodies in upper respiratory and oral secretions that would not only prevent initial acquisition of the virus, but also suppress community spread via aerosols and droplets generated from these secretions.
Collapse
Affiliation(s)
- Michael W. Russell
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Jiri Mestecky
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
6
|
Induction of Susceptibility to Disseminated Infection with IgA1 Protease-Producing Encapsulated Pathogens Streptococcus pneumoniae, Haemophilus influenzae Type b, and Neisseria meningitidis. mBio 2022; 13:e0055022. [PMID: 35420467 PMCID: PMC9239265 DOI: 10.1128/mbio.00550-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Streptococcus pneumoniae, Neisseria meningitidis, and Haemophilus influenzae are the principal causes of bacterial meningitis. It is unexplained why only occasional individuals develop invasive infection, while the vast majority remain healthy and develop immunity when encountering these pathogens. A capsular polysaccharide and an IgA1 protease are common to these pathogens. We tested the hypothesis that patients are primed to susceptibility to invasive infection by other bacteria that express the same capsular polysaccharide but no IgA1 protease. Thereby, the subsequently colonizing pathogen may protect its surface with IgA1 protease-generated Fab fragments of IgA1 devoid of Fc-mediated effector functions. Military recruits who remained healthy when acquiring meningococci showed a significant response of inhibitory antibodies against the IgA1 protease of the colonizing clone concurrent with serum antibodies against its capsular polysaccharide. At hospitalization, 70.8% of meningitis patients carried fecal bacteria cross-reactive with the capsule of the actual pathogen, in contrast to 6% of controls (P < 0.0001). These were Escherichia coli K100, K1, and K92 in patients with infection caused by H. influenzae type b and N. meningitidis groups B and C, respectively. This concurred with a significant IgA1 response to the capsule but not to the IgA1 protease of the pathogen. The demonstrated multitude of relationships between capsular types and distinct IgA1 proteases in pneumococci suggests an alternative route of immunological priming associated with recombining bacteria. The findings support the model and offer an explanation for the rare occurrence of invasive diseases in spite of the comprehensive occurrence of the pathogens.
Collapse
|
7
|
Wang Z, Rahkola J, Redzic JS, Chi YC, Tran N, Holyoak T, Zheng H, Janoff E, Eisenmesser E. Mechanism and inhibition of Streptococcus pneumoniae IgA1 protease. Nat Commun 2020; 11:6063. [PMID: 33247098 PMCID: PMC7695701 DOI: 10.1038/s41467-020-19887-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/30/2020] [Indexed: 01/27/2023] Open
Abstract
Opportunistic pathogens such as Streptococcus pneumoniae secrete a giant metalloprotease virulence factor responsible for cleaving host IgA1, yet the molecular mechanism has remained unknown since their discovery nearly 30 years ago despite the potential for developing vaccines that target these enzymes to block infection. Here we show through a series of cryo-electron microscopy single particle reconstructions how the Streptococcus pneumoniae IgA1 protease facilitates IgA1 substrate recognition and how this can be inhibited. Specifically, the Streptococcus pneumoniae IgA1 protease subscribes to an active-site-gated mechanism where a domain undergoes a 10.0 Å movement to facilitate cleavage. Monoclonal antibody binding inhibits this conformational change, providing a direct means to block infection at the host interface. These structural studies explain decades of biological and biochemical studies and provides a general strategy to block Streptococcus pneumoniae IgA1 protease activity to potentially prevent infection.
Collapse
Affiliation(s)
- Zhiming Wang
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, School of Medicine, Aurora, CO, 80045, USA
| | - Jeremy Rahkola
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Disease, University of Colorado Denver School of Medicine and Denver Veterans Affairs Medical Center, Aurora, CO, 80045, USA
| | - Jasmina S Redzic
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, School of Medicine, Aurora, CO, 80045, USA
| | - Ying-Chih Chi
- Cryo-EM Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Norman Tran
- Department of Biology, University of Waterloo, Waterloo, ON, Canada, N2L 3G1
| | - Todd Holyoak
- Department of Biology, University of Waterloo, Waterloo, ON, Canada, N2L 3G1
| | - Hongjin Zheng
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, School of Medicine, Aurora, CO, 80045, USA.
| | - Edward Janoff
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Disease, University of Colorado Denver School of Medicine and Denver Veterans Affairs Medical Center, Aurora, CO, 80045, USA.
| | - Elan Eisenmesser
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
8
|
Formation and Maturation of the Phagosome: A Key Mechanism in Innate Immunity against Intracellular Bacterial Infection. Microorganisms 2020; 8:microorganisms8091298. [PMID: 32854338 PMCID: PMC7564318 DOI: 10.3390/microorganisms8091298] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023] Open
Abstract
Phagocytosis is an essential mechanism in innate immune defense, and in maintaining homeostasis to eliminate apoptotic cells or microbes, such as Mycobacterium tuberculosis, Salmonella enterica, Streptococcus pyogenes and Legionella pneumophila. After internalizing microbial pathogens via phagocytosis, phagosomes undergo a series of ‘maturation’ steps, to form an increasingly acidified compartment and subsequently fuse with the lysosome to develop into phagolysosomes and effectively eliminate the invading pathogens. Through this mechanism, phagocytes, including macrophages, neutrophils and dendritic cells, are involved in the processing of microbial pathogens and antigen presentation to T cells to initiate adaptive immune responses. Therefore, phagocytosis plays a role in the bridge between innate and adaptive immunity. However, intracellular bacteria have evolved diverse strategies to survive and replicate within hosts. In this review, we describe the sequential stages in the phagocytosis process. We also discuss the immune evasion strategies used by pathogens to regulate phagosome maturation during intracellular bacterial infection, and indicate that these might be used for the development of potential therapeutic strategies for infectious diseases.
Collapse
|
9
|
Yamaguchi M, Hirose Y, Takemura M, Ono M, Sumitomo T, Nakata M, Terao Y, Kawabata S. Streptococcus pneumoniae Evades Host Cell Phagocytosis and Limits Host Mortality Through Its Cell Wall Anchoring Protein PfbA. Front Cell Infect Microbiol 2019; 9:301. [PMID: 31482074 PMCID: PMC6710382 DOI: 10.3389/fcimb.2019.00301] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/05/2019] [Indexed: 12/20/2022] Open
Abstract
Streptococcus pneumoniae is a Gram-positive bacterium belonging to the oral streptococcus species, mitis group. This pathogen is a leading cause of community-acquired pneumonia, which often evades host immunity and causes systemic diseases, such as sepsis and meningitis. Previously, we reported that PfbA is a β-helical cell surface protein contributing to pneumococcal adhesion to and invasion of human epithelial cells in addition to its survival in blood. In the present study, we investigated the role of PfbA in pneumococcal pathogenesis. Phylogenetic analysis indicated that the pfbA gene is highly conserved in S. pneumoniae and Streptococcus pseudopneumoniae within the mitis group. Our in vitro assays showed that PfbA inhibits neutrophil phagocytosis, leading to pneumococcal survival. We found that PfbA activates NF-κB through TLR2, but not TLR4. In addition, TLR2/4 inhibitor peptide treatment of neutrophils enhanced the survival of the S. pneumoniae ΔpfbA strain as compared to a control peptide treatment, whereas the treatment did not affect survival of a wild-type strain. In a mouse pneumonia model, the host mortality and level of TNF-α in bronchoalveolar lavage fluid were comparable between wild-type and ΔpfbA-infected mice, while deletion of pfbA decreased the bacterial burden in bronchoalveolar lavage fluid. In a mouse sepsis model, the ΔpfbA strain demonstrated significantly increased host mortality and TNF-α levels in plasma, but showed reduced bacterial burden in lung and liver. These results indicate that PfbA may contribute to the success of S. pneumoniae species by inhibiting host cell phagocytosis, excess inflammation, and mortality by interacting with TLR2.
Collapse
Affiliation(s)
- Masaya Yamaguchi
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Yujiro Hirose
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Moe Takemura
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan.,Department of Oral and Maxillofacial Surgery II, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masayuki Ono
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan.,Department of Fixed Prosthodontics, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Tomoko Sumitomo
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masanobu Nakata
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shigetada Kawabata
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
10
|
Loughran AJ, Orihuela CJ, Tuomanen EI. Streptococcus pneumoniae: Invasion and Inflammation. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0004-2018. [PMID: 30873934 PMCID: PMC6422050 DOI: 10.1128/microbiolspec.gpp3-0004-2018] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Indexed: 12/22/2022] Open
Abstract
Streptococcus pneumoniae (the pneumoccus) is the leading cause of otitis media, community-acquired pneumonia, and bacterial meningitis. The success of the pneumococcus stems from its ability to persist in the population as a commensal and avoid killing by immune system. This chapter first reviews the molecular mechanisms that allow the pneumococcus to colonize and spread from one anatomical site to the next. Then, it discusses the mechanisms of inflammation and cytotoxicity during emerging and classical pneumococcal infections.
Collapse
Affiliation(s)
- Allister J Loughran
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Carlos J Orihuela
- Department of Microbiology, The University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294
| | - Elaine I Tuomanen
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105
| |
Collapse
|
11
|
Cross BW, Ruhl S. Glycan recognition at the saliva - oral microbiome interface. Cell Immunol 2018; 333:19-33. [PMID: 30274839 DOI: 10.1016/j.cellimm.2018.08.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 01/25/2023]
Abstract
The mouth is a first critical interface where most potentially harmful substances or pathogens contact the host environment. Adaptive and innate immune defense mechanisms are established there to inactivate or eliminate pathogenic microbes that traverse the oral environment on the way to their target organs and tissues. Protein and glycoprotein components of saliva play a particularly important role in modulating the oral microbiota and helping with the clearance of pathogens. It has long been acknowledged that glycobiological and glycoimmunological aspects play a pivotal role in oral host-microbe, microbe-host, and microbe-microbe interactions in the mouth. In this review, we aim to delineate how glycan-mediated host defense mechanisms in the oral cavity support human health. We will describe the role of glycans attached to large molecular size salivary glycoproteins which act as a first line of primordial host defense in the human mouth. We will further discuss how glycan recognition contributes to both colonization and clearance of oral microbes.
Collapse
Affiliation(s)
- Benjamin W Cross
- Department of Oral Biology, University at Buffalo, Buffalo, NY, United States
| | - Stefan Ruhl
- Department of Oral Biology, University at Buffalo, Buffalo, NY, United States.
| |
Collapse
|
12
|
Klimesova K, Jiraskova Zakostelska Z, Tlaskalova-Hogenova H. Oral Bacterial and Fungal Microbiome Impacts Colorectal Carcinogenesis. Front Microbiol 2018; 9:774. [PMID: 29731748 PMCID: PMC5920026 DOI: 10.3389/fmicb.2018.00774] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 04/05/2018] [Indexed: 12/31/2022] Open
Abstract
Host's physiology is significantly influenced by microbiota colonizing the epithelial surfaces. Complex microbial communities contribute to proper mucosal barrier function, immune response, and prevention of pathogen invasion and have many other crucial functions. The oral cavity and large intestine are distant parts of the digestive tract, both heavily colonized by commensal microbiota. Nevertheless, they feature different proportions of major bacterial and fungal phyla, mostly due to distinct epithelial layers organization and different oxygen levels. A few obligate anaerobic strains inhabiting the oral cavity are involved in the pathogenesis of oral diseases. Interestingly, these microbiota components are also enriched in gut inflammatory and tumor tissue. An altered microbiota composition - dysbiosis - and formation of polymicrobial biofilms seem to play important roles in the development of oral diseases and colorectal cancer. In this review, we describe the differences in composition of commensal microbiota in the oral cavity and large intestine and the mechanisms by which microbiota affect the inflammatory and carcinogenic response of the host.
Collapse
Affiliation(s)
- Klara Klimesova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the CAS, Prague, Czechia
| | | | | |
Collapse
|
13
|
Novel Two-Component System of Streptococcus sanguinis Affecting Functions Associated with Viability in Saliva and Biofilm Formation. Infect Immun 2018; 86:IAI.00942-17. [PMID: 29339459 DOI: 10.1128/iai.00942-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 01/08/2018] [Indexed: 02/07/2023] Open
Abstract
Streptococcus sanguinis is a pioneer species of teeth and a common opportunistic pathogen of infective endocarditis. In this study, we identified a two-component system, S. sanguinis SptRS (SptRS Ss ), affecting S. sanguinis survival in saliva and biofilm formation. Isogenic mutants of sptRSs (SKsptR) and sptSSs (SKsptS) showed reduced cell counts in ex vivo assays of viability in saliva compared to those of parent strain SK36 and complemented mutants. Reduced counts of the mutants in saliva were associated with reduced growth rates in nutrient-poor medium (RPMI) and increased susceptibility to the deposition of C3b and the membrane attach complex (MAC) of the complement system, a defense component of saliva and serum. Conversely, sptRSs and sptSSs mutants showed increased biofilm formation associated with higher levels of production of H2O2 and extracellular DNA. Reverse transcription-quantitative PCR (RT-qPCR) comparisons of strains indicated a global role of SptRS Ss in repressing genes for H2O2 production (2.5- to 15-fold upregulation of spxB, spxR, vicR, tpk, and ackA in sptRSs and sptSSs mutants), biofilm formation, and/or evasion of host immunity (2.1- to 11.4-fold upregulation of srtA, pcsB, cwdP, iga, and nt5e). Compatible with the homology of SptR Ss with AraC-type regulators, duplicate to multiple conserved repeats were identified in 1,000-bp regulatory regions of downstream genes, suggesting that SptR Ss regulates transcription by DNA looping. Significant transcriptional changes in the regulatory genes vicR, spxR, comE, comX, and mecA in the sptRSs and sptSSs mutants further indicated that SptRS Ss is part of a regulatory network that coordinates cell wall homeostasis, H2O2 production, and competence. This study reveals that SptRS Ss is involved in the regulation of crucial functions for S. sanguinis persistence in the oral cavity.
Collapse
|
14
|
Ahearn CP, Gallo MC, Murphy TF. Insights on persistent airway infection by non-typeable Haemophilus influenzae in chronic obstructive pulmonary disease. Pathog Dis 2017; 75:3753446. [PMID: 28449098 PMCID: PMC5437125 DOI: 10.1093/femspd/ftx042] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/28/2017] [Indexed: 12/21/2022] Open
Abstract
Non-typeable Haemophilus influenzae (NTHi) is the most common bacterial cause of infection of the lower airways in adults with chronic obstructive pulmonary disease (COPD). Infection of the COPD airways causes acute exacerbations, resulting in substantial morbidity and mortality. NTHi has evolved multiple mechanisms to establish infection in the hostile environment of the COPD airways, allowing the pathogen to persist in the airways for months to years. Persistent infection of the COPD airways contributes to chronic airway inflammation that increases symptoms and accelerates the progressive loss of pulmonary function, which is a hallmark of the disease. Persistence mechanisms of NTHi include the expression of multiple redundant adhesins that mediate binding to host cellular and extracellular matrix components. NTHi evades host immune recognition and clearance by invading host epithelial cells, forming biofilms, altering gene expression and displaying surface antigenic variation. NTHi also binds host serum factors that confer serum resistance. Here we discuss the burden of COPD and the role of NTHi infections in the course of the disease. We provide an overview of NTHi mechanisms of persistence that allow the pathogen to establish a niche in the hostile COPD airways.
Collapse
Affiliation(s)
- Christian P. Ahearn
- Department of Microbiology and Immunology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
- Clinical and Translational Research Center, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Mary C. Gallo
- Department of Microbiology and Immunology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
- Clinical and Translational Research Center, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Timothy F. Murphy
- Department of Microbiology and Immunology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
- Clinical and Translational Research Center, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
- Division of Infectious Disease, Department of Medicine, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| |
Collapse
|
15
|
Santín GRG, Salgado AV, Bastida NMM, Gómez IDLR, Benítez JGS, Zerón HM. Salivary Immunoglobulin Gene Expression in Patients with Caries. Open Access Maced J Med Sci 2017; 5:236-243. [PMID: 28507635 PMCID: PMC5420781 DOI: 10.3889/oamjms.2017.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 01/14/2017] [Accepted: 02/07/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND: Immunoglobulins mediate the host’s humoral immune response are expressed in saliva. AIM: To quantify the FcαR, FcγRIIB, and FcαμR gene expression in the saliva of Mexican patients with caries in mixed and permanent dentition. SUBJECTS AND METHODS: This was a comparative cross-sectional study. mRNA was isolated from 200 μL of saliva following the RNA III Tissue Fresh-frozen protocol of the MagNA Pure LC Instrument 2.0 (Roche Diagnostics GmbH, Nederland BV) and the FcαR, FcαμR and FcγRIIB were quantified through TaqMan Assays. RESULTS: One hundred individuals, 50 with mixed dentition and 50 with permanent dentition, were included in the study. Statistically, it was found a significant difference (p = 0.025) in the IgG (FcγRIIB) expression between the studied groups. CONCLUSION: Although we confirmed the existence of FcαR, FcγRIIB and FcαμR gene expression in saliva, only a significant difference in the expression of FcγRIIB between the mixed dentition and permanent dentition was found.
Collapse
Affiliation(s)
- Gema Regina Guadarrama Santín
- Laboratory of Molecular Biology, Medical Sciences Research Center (CICMED), Autonomous University of the State of Mexico (UAEMex), Jesús Carranza 205, Col. Universidad, C.P. 50130, Toluca, México, Mexico
| | - Angel Visoso Salgado
- Laboratory of Molecular Biology, Medical Sciences Research Center (CICMED), Autonomous University of the State of Mexico (UAEMex), Jesús Carranza 205, Col. Universidad, C.P. 50130, Toluca, México, Mexico
| | - Norma Margarita Montiel Bastida
- Laboratory of Molecular Biology, Medical Sciences Research Center (CICMED), Autonomous University of the State of Mexico (UAEMex), Jesús Carranza 205, Col. Universidad, C.P. 50130, Toluca, México, Mexico
| | - Isaías de la Rosa Gómez
- Laboratory of Molecular Biology, Medical Sciences Research Center (CICMED), Autonomous University of the State of Mexico (UAEMex), Jesús Carranza 205, Col. Universidad, C.P. 50130, Toluca, México, Mexico
| | - Jonnathan Guadalupe Santillán Benítez
- Laboratory of Molecular Biology, Medical Sciences Research Center (CICMED), Autonomous University of the State of Mexico (UAEMex), Jesús Carranza 205, Col. Universidad, C.P. 50130, Toluca, México, Mexico
| | - Hugo Mendieta Zerón
- Laboratory of Molecular Biology, Medical Sciences Research Center (CICMED), Autonomous University of the State of Mexico (UAEMex), Jesús Carranza 205, Col. Universidad, C.P. 50130, Toluca, México, Mexico
| |
Collapse
|
16
|
Role of Serum Mycoplasma pneumoniae IgA, IgM, and IgG in the Diagnosis of Mycoplasma pneumoniae-Related Pneumonia in School-Age Children and Adolescents. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00471-16. [PMID: 27760779 DOI: 10.1128/cvi.00471-16] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 10/07/2016] [Indexed: 01/10/2023]
Abstract
Mycoplasma pneumoniae is an important causative pathogen of community-acquired pneumonia in children. Rapid and reliable laboratory diagnosis of M. pneumoniae infection is important so that appropriate antibiotic treatment can be initiated to reduce the misuse of drugs and resistance rates. Anti-M. pneumoniae immunoglobulin M (IgM) is an indicator of recent primary infection but can persist for several months after initial infection. It has been suggested that anti-M. pneumoniae immunoglobulin A (IgA) can be a reliable indicator for recent M. pneumoniae infection in adults. We investigated the clinical diagnostic value of M. pneumoniae IgA in school-age children and adolescents with M. pneumoniae-related pneumonia. Eighty children with pneumonia and seropositive for M. pneumoniae IgM or with a 4-fold increase of anti-M. pneumoniae immunoglobulin G (IgG) were enrolled from May 2015 to March 2016. The titers of M. pneumoniae IgA, IgM, and IgG, the clinical features, and laboratory examinations of blood, C-reactive protein, and liver enzymes were analyzed. The initial positivity rates for M. pneumoniae IgM and IgA upon admission to the hospital were 63.6 and 33.8%, respectively. One week after admission, the cumulative positivity rates for M. pneumoniae IgM and IgA increased to 97.5 and 56.3%, respectively. Detection of M. pneumoniae IgM was more sensitive than detection of M. pneumoniae IgA for the diagnosis of M. pneumoniae-related pneumonia in school-age children and adolescents; however, paired sera are necessary for a more accurate diagnosis.
Collapse
|
17
|
Hatzios SK, Abel S, Martell J, Hubbard T, Sasabe J, Munera D, Clark L, Bachovchin DA, Qadri F, Ryan ET, Davis BM, Weerapana E, Waldor MK. Chemoproteomic profiling of host and pathogen enzymes active in cholera. Nat Chem Biol 2016; 12:268-274. [PMID: 26900865 PMCID: PMC4765928 DOI: 10.1038/nchembio.2025] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 12/31/2015] [Indexed: 12/15/2022]
Abstract
Activity-based protein profiling (ABPP) is a chemoproteomic tool for detecting active enzymes in complex biological systems. We used ABPP to identify secreted bacterial and host serine hydrolases that are active in animals infected with the cholera pathogen Vibrio cholerae. Four V. cholerae proteases were consistently active in infected rabbits, and one, VC0157 (renamed IvaP), was also active in human choleric stool. Inactivation of IvaP influenced the activity of other secreted V. cholerae and rabbit enzymes in vivo, and genetic disruption of all four proteases increased the abundance of intelectin, an intestinal lectin, and its binding to V. cholerae in infected rabbits. Intelectin also bound to other enteric bacterial pathogens, suggesting that it may constitute a previously unrecognized mechanism of bacterial surveillance in the intestine that is inhibited by pathogen-secreted proteases. Our work demonstrates the power of activity-based proteomics to reveal host-pathogen enzymatic dialog in an animal model of infection.
Collapse
Affiliation(s)
- Stavroula K. Hatzios
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Sören Abel
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Department of Pharmacy, University of Tromsø (UiT), The Arctic University of Norway, Tromsø, Norway
| | | | - Troy Hubbard
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Jumpei Sasabe
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Diana Munera
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Lars Clark
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | | | - Firdausi Qadri
- International Centre for Diarrhoeal Disease Research, Bangladesh (ICDDR,B), Dhaka, Bangladesh
| | - Edward T. Ryan
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Brigid M. Davis
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | | | - Matthew K. Waldor
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| |
Collapse
|
18
|
Lanka GKK, Yu JJ, Gong S, Gupta R, Mustafa SB, Murthy AK, Zhong G, Chambers JP, Guentzel MN, Arulanandam BP. IgA modulates respiratory dysfunction as a sequela to pulmonary chlamydial infection as neonates. Pathog Dis 2016; 74:ftv121. [PMID: 26755533 DOI: 10.1093/femspd/ftv121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2015] [Indexed: 11/12/2022] Open
Abstract
Neonatal Chlamydia lung infections are associated with serious sequelae such as asthma and airway hyper-reactivity in children and adults. Our previous studies demonstrated the importance of Th-1 type cytokines, IL-12 and IFN-γ in protection against neonatal pulmonary chlamydial challenge; however, the role of the humoral arm of defense has not been elucidated. We hypothesized that B-cells and IgA, the major mucosal antibody, play a protective role in newborns against development of later life respiratory sequelae to Chlamydia infection. Our studies using neonatal mice revealed that all WT and IgA-deficient (IgA(-/-)) animals survived a sublethal pulmonary Chlamydia muridarum challenge at one day after birth with similar reduction in bacterial burdens over time. In contrast, all B-cell-deficient (μMT) mice succumbed to infection at the same challenge dose correlating to failure to control bacterial burdens in the lungs. Although IgA may not be important for bacterial clearance, we observed IgA(-/-) mice displayed greater respiratory dysfunction 5 weeks post challenge. Specifically, comparative respiratory functional analyses revealed a significant shift upward in P-V loops, and higher dynamic resistance in IgA(-/-) animals. This study provides insight(s) into the protective role of IgA in neonates against pulmonary chlamydial infection induced respiratory pathological sequelae observed later in life.
Collapse
Affiliation(s)
- Gopala Krishna Koundinya Lanka
- Department of Biology, The South Texas Center for Emerging Infectious Diseases, and the Center for Excellence in Infection Genomics, University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| | - Jieh-Juen Yu
- Department of Biology, The South Texas Center for Emerging Infectious Diseases, and the Center for Excellence in Infection Genomics, University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| | - Siqi Gong
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Rishein Gupta
- Department of Biology, The South Texas Center for Emerging Infectious Diseases, and the Center for Excellence in Infection Genomics, University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| | - Shamimunisa B Mustafa
- Department of Pediatrics, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Ashlesh K Murthy
- Department of Pathology, Midwestern University, 555 31st Street, Downers Grove, IL 60515, USA
| | - Guangming Zhong
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - James P Chambers
- Department of Biology, The South Texas Center for Emerging Infectious Diseases, and the Center for Excellence in Infection Genomics, University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| | - M Neal Guentzel
- Department of Biology, The South Texas Center for Emerging Infectious Diseases, and the Center for Excellence in Infection Genomics, University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| | - Bernard P Arulanandam
- Department of Biology, The South Texas Center for Emerging Infectious Diseases, and the Center for Excellence in Infection Genomics, University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| |
Collapse
|
19
|
Murphy TF, Kirkham C, Jones MM, Sethi S, Kong Y, Pettigrew MM. Expression of IgA Proteases by Haemophilus influenzae in the Respiratory Tract of Adults With Chronic Obstructive Pulmonary Disease. J Infect Dis 2015; 212:1798-805. [PMID: 25995193 DOI: 10.1093/infdis/jiv299] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 05/13/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Immunoglobulin (Ig)A proteases of Haemophilus influenzae are highly specific endopeptidases that cleave the hinge region of human IgA1 and also mediate invasion and trafficking in human respiratory epithelial cells, facilitating persistence of H. influenzae. Little is known about the expression of IgA proteases in clinical settings of H. influenzae infection. METHODS We identified and characterized IgA protease genes in H. influenzae and studied their expression and proteolytic specificity, in vitro and in vivo in 169 independent strains of H. influenzae collected longitudinally over 10 years from adults with chronic obstructive pulmonary disease. RESULTS The H. influenzae pangenome has 2 alleles of IgA protease genes; all strains have igaA, and 40% of strains have igaB. Each allele has 2 variants with differing proteolytic specificities for human IgA1. A total of 88% of 169 strains express IgA protease activity. Expression of the 4 forms of IgA protease varies among strains. Based on the presence of IgA1 fragments in sputum samples, each of the different forms of IgA protease is selectively expressed in the human airways during infection. CONCLUSIONS Four variants of IgA proteases are variably expressed by H. influenzae during infection of the human airways.
Collapse
Affiliation(s)
- Timothy F Murphy
- Division of Infectious Diseases Department of Microbiology and Immunology Clinical and Translational Research Center, University at Buffalo, State University of New York
| | - Charmaine Kirkham
- Division of Infectious Diseases Clinical and Translational Research Center, University at Buffalo, State University of New York
| | - Megan M Jones
- Department of Microbiology and Immunology Clinical and Translational Research Center, University at Buffalo, State University of New York
| | - Sanjay Sethi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine Veterans Affairs Western New York Healthcare System, Buffalo, New York
| | - Yong Kong
- Department of Molecular Biophysics and Biochemistry, W.M. Keck Biotechnology Resource Laboratory
| | - Melinda M Pettigrew
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut
| |
Collapse
|
20
|
Finney LJ, Ritchie A, Pollard E, Johnston SL, Mallia P. Lower airway colonization and inflammatory response in COPD: a focus on Haemophilus influenzae. Int J Chron Obstruct Pulmon Dis 2014; 9:1119-32. [PMID: 25342897 DOI: 10.2147/copd.s54477] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Bacterial infection of the lower respiratory tract in chronic obstructive pulmonary disease (COPD) patients is common both in stable patients and during acute exacerbations. The most frequent bacteria detected in COPD patients is Haemophilus influenzae, and it appears this organism is uniquely adapted to exploit immune deficiencies associated with COPD and to establish persistent infection in the lower respiratory tract. The presence of bacteria in the lower respiratory tract in stable COPD is termed colonization; however, there is increasing evidence that this is not an innocuous phenomenon but is associated with airway inflammation, increased symptoms, and increased risk for exacerbations. In this review, we discuss host immunity that offers protection against H. influenzae and how disturbance of these mechanisms, combined with pathogen mechanisms of immune evasion, promote persistence of H. influenzae in the lower airways in COPD. In addition, we examine the role of H. influenzae in COPD exacerbations, as well as interactions between H. influenzae and respiratory virus infections, and review the role of treatments and their effect on COPD outcomes. This review focuses predominantly on data derived from human studies but will refer to animal studies where they contribute to understanding the disease in humans.
Collapse
Affiliation(s)
- Lydia J Finney
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Andrew Ritchie
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | | | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Patrick Mallia
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| |
Collapse
|
21
|
Janoff EN, Rubins JB, Fasching C, Charboneau D, Rahkola JT, Plaut AG, Weiser JN. Pneumococcal IgA1 protease subverts specific protection by human IgA1. Mucosal Immunol 2014; 7:249-56. [PMID: 23820749 PMCID: PMC4456019 DOI: 10.1038/mi.2013.41] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 05/17/2013] [Indexed: 02/04/2023]
Abstract
Bacterial immunoglobulin A1 (IgA1) proteases may sabotage the protective effects of IgA. In vitro, both exogenous and endogenously produced IgA1 protease inhibited phagocytic killing of Streptococcus pneumoniae by capsule-specific IgA1 human monoclonal antibodies (hMAbs) but not IgA2. These IgA1 proteases cleaved and reduced binding of the the effector Fcα1 heavy chain but not the antigen-binding F(ab)/light chain to pneumococcal surfaces. In vivo, IgA1 protease-resistant IgA2, but not IgA1 protease-sensitive IgA1, supported 60% survival in mice infected with wild-type S. pneumoniae. IgA1 hMAbs protected mice against IgA1 protease-deficient but not -producing pneumococci. Parallel mouse sera with human IgA2 showed more efficient complement-mediated reductions in pneumococci with neutrophils than did IgA1, particularly with protease-producing organisms. After natural human pneumococcal bacteremia, purified serum IgG inhibited IgA1 protease activity in 7 of 11 patients (64%). These observations provide the first evidence in vivo that IgA1 protease can circumvent killing of S. pneumoniae by human IgA. Acquisition of IgA1 protease-neutralizing IgG after infection directs attention to IgA1 protease both as a determinant of successful colonization and infection and as a potential vaccine candidate.
Collapse
Affiliation(s)
- Edward N. Janoff
- Mucosal and Vaccine Research Colorado (MAVRC), University of Colorado Denver, Infectious Diseases, Aurora, CO 80045
| | - Jeffrey B. Rubins
- University of Minnesota School of Medicine, Minneapolis, Minnesota Denver Veterans Affairs Medical Center, Denver, CO 80220
| | - Claudine Fasching
- Veterans Affairs Medical Center Denver Veterans Affairs Medical Center, Denver, CO 80220
| | - Darlene Charboneau
- University of Minnesota School of Medicine, Minneapolis, Minnesota Denver Veterans Affairs Medical Center, Denver, CO 80220
| | - Jeremy T. Rahkola
- Mucosal and Vaccine Research Colorado (MAVRC), University of Colorado Denver, Infectious Diseases, Aurora, CO 80045
| | - Andrew G. Plaut
- Department of Medicine and Division of Gastroenterology, Tufts-New England Medical Center, Boston, Massachusetts
| | - Jeffrey N. Weiser
- Departments of Microbiology and Pediatrics University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
22
|
Huang L, Xu QA, Liu C, Fan MW, Li YH. Anti-caries DNA vaccine-induced secretory immunoglobulin A antibodies inhibit formation of Streptococcus mutans biofilms in vitro. Acta Pharmacol Sin 2013; 34:239-46. [PMID: 23274411 DOI: 10.1038/aps.2012.145] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
AIM To investigate the effects of anti-caries DNA vaccine-induced salivary secretory immunoglobulin A (S-IgA) antibodies on Streptococcus mutans (S. mutans) adherence and biofilms formation in vitro. METHODS Adult female Wistar rats were intranasally immunized with the anti-caries DNA vaccine pGJA-P/VAX. Their saliva samples were collected at different times after the immunization, and S-IgA antibody level in the saliva and its inhibition on S. mutans adherence were examined. The effects of S-IgA in the saliva with the strongest inhibitory effects were examined at 3 different stages, ie acquired pellicles, biofilm formation and production of mature biofilms. The number of viable bacteria and depth of the biofilm at 16 h in each stage were determined using counting colony forming units and using a confocal laser scanning microscopy (CLSM). The participation of S-IgA in acquired pellicles and its aggregation with S. mutans were also observed under CLSM. RESULTS The S-IgA titer in saliva reached its peak and exhibited the strongest inhibition on S. mutans adhesion at 10 weeks after the immunization. The colonies and depth of the biofilm in the saliva-pretreated group were 41.79% and 41.02%, respectively, less than the control group. The colonies and depth of the biofilm in the co-culture group were 27.4% and 22.81% less than the control group. The assembly of S. mutans and S-IgA was observed under CLSM after co-cultivation. In the mature-stage biofilm, no differences were observed between the different groups. CONCLUSION These results demonstrate that the anti-caries DNA vaccine induces the production of specific S-IgA antibodies that may prevent dental caries by inhibiting the initial adherence of S. mutans onto tooth surfaces, thereby reducing the accumulation of S. mutans on the acquired pellicles.
Collapse
|
23
|
Millares L, Marin A, Garcia-Aymerich J, Sauleda J, Belda J, Monsó E. Specific IgA and metalloproteinase activity in bronchial secretions from stable chronic obstructive pulmonary disease patients colonized by Haemophilus influenzae. Respir Res 2012; 13:113. [PMID: 23228114 PMCID: PMC3546904 DOI: 10.1186/1465-9921-13-113] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 12/05/2012] [Indexed: 12/23/2022] Open
Abstract
Background Haemophilus influenzae is the most common colonizing bacteria of the bronchial tree in chronic obstructive pulmonary disease (COPD), and positive cultures for this potentially pathogenic microorganism (PPM) has been associated with local inflammation changes that may influence the relationships between H. influenzae and the bronchial mucosa. Methods A cross-sectional analysis of stable COPD patients enrolled in the Phenotype and Course of Chronic Obstructive Pulmonary Disease (PAC-COPD) Study, focusing on bronchial colonization by H. influenzae, was performed. Specific IgA against the PPM was measured by optical density, and metalloproteinase-9 (MMP-9) and tissue inhibitor of metalloproteinase-1 (TIMP-1) using ELISA in sputum samples. Levels in patients colonized by H. influenzae and non-colonized patients were compared. Results Sputum supernatant for the measurement of specific IgA against H. influenzae was available from 54 stable COPD patients, who showed levels of specific IgA significantly lower in colonized (n=21) than in non-colonized patients (n=33) (15 [4-37] versus 31 [10-75], p=0.033, Mann-Whitney U test). Proenzyme MMP-9 was measured in 44 patients, and it was higher in colonized (n=12, 1903 [1488-6699] ng/ml) than in non-colonized patients (n=32, 639 [373-972] ng/ml) (p<0.001, Mann-Whitney U test). Active form of MMP-9 was also higher in colonized (126 [25-277] ng/ml) than in non-colonized patients (39 [14-68] ng/ml) (p=0.021, Mann-Whitney U test), and the molar ratio between proenzyme MMP-9 and TIMP-1 was above 1 (2.1 [0.1-12.5]) in colonized patients, significantly higher than the ratio found in non-colonized patients (0.2 [0.08-0.5]) (p=0.030, Mann-Whitney U test). Conclusions Clinically stable COPD patients colonized by H. influenzae had lower levels of specific IgA against the microorganism and higher values of the active form of MMP-9 in their sputum supernatant than non-colonized patients. Bronchial colonization by H. influenzae may cause structural changes in the extracellular matrix through a defective defense and the production of active metalloproteinases.
Collapse
|
24
|
Occurrence and evolution of the paralogous zinc metalloproteases IgA1 protease, ZmpB, ZmpC, and ZmpD in Streptococcus pneumoniae and related commensal species. mBio 2012; 3:mBio.00303-12. [PMID: 23033471 PMCID: PMC3518915 DOI: 10.1128/mbio.00303-12] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The distribution, genome location, and evolution of the four paralogous zinc metalloproteases, IgA1 protease, ZmpB, ZmpC, and ZmpD, in Streptococcus pneumoniae and related commensal species were studied by in silico analysis of whole genomes and by activity screening of 154 representatives of 20 species. ZmpB was ubiquitous in the Mitis and Salivarius groups of the genus Streptococcus and in the genera Gemella and Granulicatella, with the exception of a fragmented gene in Streptococcus thermophilus, the only species with a nonhuman habitat. IgA1 protease activity was observed in all members of S. pneumoniae, S. pseudopneumoniae, S. oralis, S. sanguinis, and Gemella haemolysans, was variably present in S. mitis and S. infantis, and absent in S. gordonii, S. parasanguinis, S. cristatus, S. oligofermentans, S. australis, S. peroris, and S. suis. Phylogenetic analysis of 297 zmp sequences and representative housekeeping genes provided evidence for an unprecedented selection for genetic diversification of the iga, zmpB, and zmpD genes in S. pneumoniae and evidence of very frequent intraspecies transfer of entire genes and combination of genes. Presumably due to their adaptation to a commensal lifestyle, largely unaffected by adaptive mucosal immune factors, the corresponding genes in commensal streptococci have remained conserved. The widespread distribution and significant sequence diversity indicate an ancient origin of the zinc metalloproteases predating the emergence of the humanoid species. zmpB, which appears to be the ancestral gene, subsequently duplicated and successfully diversified into distinct functions, is likely to serve an important but yet unknown housekeeping function associated with the human host. The paralogous zinc metalloproteases IgA1 protease, ZmpB, ZmpC, and ZmpD have been identified as crucial for virulence of the human pathogen Streptococcus pneumoniae. This study maps the presence of the corresponding genes and enzyme activities in S. pneumoniae and in related commensal species of the genera Streptococcus, Gemella, and Granulicatella. The distribution, genome location, and sequence diversification indicate that zmpB is the ancestral gene predating the evolution of today’s humanoid species. The ZmpB protease may play an important but yet unidentified role in the association of streptococci of the Mitis and Salivarius groups with their human host, as it is ubiquitous in these two groups, except for a fragmented gene in Streptococcus thermophilus, the only species not associated with humans. The relative sequence diversification of the IgA1 protease, ZmpB, and ZmpD is striking evidence of differences in selection for diversification of these surface-exposed proteins in the pathogen S. pneumoniae compared to the closely related commensal streptococci.
Collapse
|
25
|
Dang Z, Feng J, Yagi K, Sugimoto C, Li W, Oku Y. Mucosal adjuvanticity of fibronectin-binding peptide (FBP) fused with Echinococcus multilocularis tetraspanin 3: systemic and local antibody responses. PLoS Negl Trop Dis 2012; 6:e1842. [PMID: 23029596 PMCID: PMC3459843 DOI: 10.1371/journal.pntd.0001842] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 08/15/2012] [Indexed: 12/24/2022] Open
Abstract
Background Studies have shown that a bacterial fibronectin attachment protein (FAP) is able to stimulate strong systemic and mucosal antibody responses when it is used alone or co-administrated with other antigens (Ags). Thus, it has been suggested to be a promising adjuvant candidate for the development of efficient vaccines. However, the co-administered Ags and FAP were cloned, expressed and purified individually to date. In a recent study, we first evaluated the adjuvanticity of a fibronectin-binding peptide (FBP, 24 amino acids) of Mycobacterium avium FAP fused with Echinococcus multilocularis tetraspanin 3 (Em-TSP3) by detecting systemic and local antibody responses in intranasally (i.n.) immunized BALB/c mice. Methodology/Principal Findings Em-TSP3 and FBP fragments were linked with a GSGGSG linker and expressed as a single fusion protein (Em-TSP3-FBP) using the pBAD/Thio-TOPO expression vector. BALB/c mice were immunized i.n. with recombinant Em-TSP3-FBP (rEm-TSP3-FBP) and rEm-TSP3+CpG and the systemic and local antibody responses were detected by ELISA. The results showed that both rEm-TSP3-FBP and rEm-TSP3+CpG evoked strong serum IgG (p<0.001) and IgG1 responses (p<0.001), whereas only the latter induced a high level IgG2α production (p<0.001), compared to that of rEm-TSP3 alone without any adjuvant. There were no significant differences in IgG and IgG1 production between the groups. Low level of serum IgA and IgM were detected in both groups. The tendency of Th1 and Th2 cell immune responses were assessed via detecting the IgG1/IgG2α ratio after the second and third immunizations. The results indicated that i.n. immunization with rEm-TSP3-FBP resulted in an increased IgG1/IgG2α ratio (a Th2 tendency), while rEm-TSP3+CpG caused a rapid Th1 response that later shifted to a Th2 response. Immunization with rEm-TSP3-FBP provoked significantly stronger IgA antibody responses in intestine (p<0.05), lung (p<0.001) and spleen (p<0.001) compared to those by rEm-TSP3+CpG. Significantly high level IgA antibodies were detected in nasal cavity (p<0.05) and liver (p<0.05) samples from both groups when compared to rEm-TSP3 alone without any adjuvant, with no significant difference between them. Conclusions I.n. administration of rEm-TSP3-FBP can induce strong systemic and mucosal antibody responses in immunized BALB/c mice, suggesting that fusion of Em-TSP3 with FBP is a novel, prospective strategy for developing safe and efficient human mucosal vaccines against alveolar echinococcosis (AE). Echinococcus metacestodes form a laminated layer and develop strategies to escape host immune responses once the infection established on the liver of intermediated host. One of the most important strategies is thought to be immunoregulation, where some molecules (e.g., antigen B) impair dendritic cell (DC) differentiation and polarize immature DC maturation towards a non-protective Th2 cell response. Therefore, it is more feasible to kill Echinococcus oncospheres in the early stage of infection in the intestine and blood. Systemic and local immune responses are believed to play a crucial role on oncosphere exclusion. Among antigen delivery systems, i.n. administration is the most efficient one, inducing both systemic and a full-range of mucosal immune responses. FAP is necessary to M. avium and S. pyogenes to efficiently attach and invade epithelial cells, and has been suggested as a potent vaccine adjuvant. Mucosal immune responses are induced after FAP binds to the fibronectin protein of host microfold (M) cells and DCs are activated. We developed a one-step delivery system where FAP and other Ags can be expressed, purified and immunized as one protein. The systemic and, in particular, the mucosal antibody responses induced by the fusion protein were detected to evaluate the adjuvanticity of FBP.
Collapse
Affiliation(s)
- Zhisheng Dang
- College of Life and Environmental Sciences, Minzu University of China, Beijing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
26
|
Smoum R, Rubinstein A, Dembitsky VM, Srebnik M. Boron containing compounds as protease inhibitors. Chem Rev 2012; 112:4156-220. [PMID: 22519511 DOI: 10.1021/cr608202m] [Citation(s) in RCA: 322] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Reem Smoum
- The School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel.
| | | | | | | |
Collapse
|
27
|
Dang Z, Yagi K, Oku Y, Kouguchi H, Kajino K, Matsumoto J, Nakao R, Wakaguri H, Toyoda A, Yin H, Sugimoto C. A pilot study on developing mucosal vaccine against alveolar echinococcosis (AE) using recombinant tetraspanin 3: Vaccine efficacy and immunology. PLoS Negl Trop Dis 2012; 6:e1570. [PMID: 22479658 PMCID: PMC3313938 DOI: 10.1371/journal.pntd.0001570] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 02/05/2012] [Indexed: 12/13/2022] Open
Abstract
Background We have previously evaluated the vaccine efficacies of seven tetraspanins of Echinococcus multilocularis (Em-TSP1–7) against alveolar echinococcosis (AE) by subcutaneous (s.c.) administration with Freund's adjuvant. Over 85% of liver cyst lesion number reductions (CLNR) were achieved by recombinant Em-TSP1 (rEm-TSP1) and -TSP3 (rEm-TSP3). However, to develop an efficient and safe human vaccine, the efficacy of TSP mucosal vaccines must be thoroughly evaluated. Methodology/Principal Findings rEm-TSP1 and -TSP3 along with nontoxic CpG ODN (CpG oligodeoxynucleotides) adjuvant were intranasally (i.n.) immunized to BALB/c mice and their vaccine efficacies were evaluated by counting liver CLNR (experiment I). 37.1% (p<0.05) and 62.1% (p<0.001) of CLNR were achieved by these two proteins, respectively. To study the protection-associated immune responses induced by rEm-TSP3 via different immunization routes (i.n. administration with CpG or s.c. immunization with Freund's adjuvant), the systemic and mucosal antibody responses were detected by ELISA (experiment II). S.c. and i.n. administration of rEm-TSP3 achieved 81.9% (p<0.001) and 62.8% (p<0.01) CLNR in the liver, respectively. Both the immunization routes evoked strong serum IgG, IgG1 and IgG2α responses; i.n. immunization induced significantly higher IgA responses in nasal cavity and intestine compared with s.c. immunization (p<0.001). Both immunization routes induced extremely strong liver IgA antibody responses (p<0.001). The Th1 and Th2 cell responses were assessed by examining the IgG1/IgG2α ratio at two and three weeks post-immunization. S.c. immunization resulted in a reduction in the IgG1/IgG2α ratio (Th1 tendency), whereas i.n. immunization caused a shift from Th1 to Th2. Moreover, immunohistochemistry showed that Em-TSP1 and -TSP3 were extensively located on the surface of E. multilocularis cysts, protoscoleces and adult worms with additional expression of Em-TSP3 in the inner part of protoscoleces and oncospheres. Conclusions Our study indicated that i.n. administration of rEm-TSP3 with CpG is able to induce both systemic and local immune responses and thus provides significant protection against AE. Humans and rodents become infected with E. multilocularis by oral ingesting of the eggs, which then develop into cysts in the liver and progress an endless proliferation. Untreated AE has a fatality rate of >90% in humans. Tetraspanins have been identified in Schistosoma and showed potential as the prospective vaccine candidates. In our recent study, we first identified seven tetraspanins in E. multilocularis and evaluated their protective efficacies as vaccines against AE when subcutaneously administered to BALB/c mice. Mucosal immunization of protective proteins is able to induce strong local and systemic immune responses, which might play a crucial role in protecting humans against E. multilocularis infection via the intestine, blood and liver. We focused on Em-TSP3, which achieved significant vaccine efficacy via both s.c. and i.n. routes. The adjuvanticity of nontoxic CpG OND as i.n. vaccine adjuvant was evaluated. The widespread expression of Em-TSP3 in all the developmental stages of E. multilocularis, and the strong local and systemic immune responses evoked by i.n. administration of rEm-TSP3 with CpG OND adjuvant suggest that this study might open the way for developing efficient, nontoxic human mucosal vaccines against AE.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Helminth/blood
- Antigens, Helminth/genetics
- Antigens, Helminth/immunology
- Echinococcosis
- Echinococcosis, Hepatic/prevention & control
- Echinococcus multilocularis/isolation & purification
- Enzyme-Linked Immunosorbent Assay
- Freund's Adjuvant/administration & dosage
- Glycoproteins/genetics
- Glycoproteins/immunology
- Immunity, Mucosal
- Immunoglobulin A/analysis
- Immunoglobulin G/blood
- Intestinal Mucosa/immunology
- Liver/parasitology
- Male
- Mice
- Mice, Inbred BALB C
- Nasal Mucosa/immunology
- Oligodeoxyribonucleotides/administration & dosage
- Pilot Projects
- Tetraspanins/genetics
- Tetraspanins/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Zhisheng Dang
- Division of Collaboration and Education, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Grazing Animal Diseases MOA, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Gansu, People's Republic of China
| | - Kinpei Yagi
- Department of Biological Science, Hokkaido Institute of Public Health, Sapporo, Hokkaido, Japan
| | - Yuzaburo Oku
- Parasitology Laboratory, School of Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Hirokazu Kouguchi
- Department of Biological Science, Hokkaido Institute of Public Health, Sapporo, Hokkaido, Japan
| | - Kiichi Kajino
- Division of Collaboration and Education, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Jun Matsumoto
- Laboratory of Medical Zoology, Nihon University College of Bioresource Sciences, Fujisawa, Japan
| | - Ryo Nakao
- Division of Collaboration and Education, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hiroyuki Wakaguri
- Department of Medical Genome Science, Graduate School of Frontier Science, The University of Tokyo, Tokyo, Japan
| | - Atsushi Toyoda
- RIKEN Genomic Sciences Center, Yokohama, Kanagawa, Japan
- Comparative Genomics Laboratory, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Hong Yin
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Grazing Animal Diseases MOA, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Gansu, People's Republic of China
| | - Chihiro Sugimoto
- Division of Collaboration and Education, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
- * E-mail:
| |
Collapse
|
28
|
Spahich NA, St Geme JW. Structure and function of the Haemophilus influenzae autotransporters. Front Cell Infect Microbiol 2011; 1:5. [PMID: 22919571 PMCID: PMC3417375 DOI: 10.3389/fcimb.2011.00005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 09/03/2011] [Indexed: 12/31/2022] Open
Abstract
Autotransporters are a large class of proteins that are found in the outer membrane of Gram-negative bacteria and are almost universally implicated in virulence. These proteins consist of a C-terminal β-domain that is embedded in the outer membrane and an N-terminal domain that is exposed on the bacterial surface and is endowed with effector function. In this article, we review and compare the structural and functional characteristics of the Haemophilus influenzae IgA1 protease and Hap monomeric autotransporters and the H. influenzae Hia and Hsf trimeric autotransporters. All of these proteins play a role in colonization of the upper respiratory tract and in the pathogenesis of H. influenzae disease.
Collapse
Affiliation(s)
- Nicole A Spahich
- Department of Pediatrics, Children's Health Center, Duke University Medical Center Durham, NC, USA
| | | |
Collapse
|
29
|
How pathogen-derived cysteine proteases modulate host immune responses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 712:192-207. [PMID: 21660666 PMCID: PMC7123607 DOI: 10.1007/978-1-4419-8414-2_12] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In mammals, cysteine proteases are essential for the induction and development of both innate and adaptive immune responses. These proteases play a role in antigen-and pathogen-recognition and elimination, signal processing and cell homeostasis. Many pathogens also secrete cysteine proteases that often act on the same target proteins as the mammalian proteases and thereby can modulate host immunity from initial recognition to effector mechanisms. Pathogen-derived proteases range from nonspecific proteases that degrade multiple proteins involved in the immune response to enzymes that are very specific in their mode of action. Here, we overview current knowledge of pathogen-derived cysteine proteases that modulate immune responses by altering the normal function of key receptors or pathways in the mammalian immune system.
Collapse
|
30
|
Vonck RA, Darville T, O'Connell CM, Jerse AE. Chlamydial infection increases gonococcal colonization in a novel murine coinfection model. Infect Immun 2011; 79:1566-77. [PMID: 21245268 PMCID: PMC3067530 DOI: 10.1128/iai.01155-10] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 11/30/2010] [Accepted: 01/07/2011] [Indexed: 12/30/2022] Open
Abstract
Genital tract infections caused by Neisseria gonorrhoeae and Chlamydia trachomatis serovars D to K occur at high incidence in many areas of the world. Despite high rates of coinfection with these pathogens, investigations of host-parasite interactions have focused on each pathogen individually. We describe here a coinfection model in which female BALB/c mice were first infected with the mouse Chlamydia species C. muridarum and then inoculated with N. gonorrhoeae following treatment with water-soluble 17β-estradiol to promote long-term gonococcal infection. Viable gonococci and chlamydiae were recovered for an average of 8 to 10 days, and diplococci and chlamydial inclusions were observed in lower genital tract tissue by immunohistochemical staining. Estradiol treatment reduced proinflammatory cytokine and chemokine levels in chlamydia-infected mice; however, coinfected mice had a higher percentage of vaginal neutrophils compared to mice infected with either pathogen alone. We detected no difference in pathogen-specific antibody levels due to coinfection. Interestingly, significantly more gonococci were recovered from coinfected mice compared to mice infected with N. gonorrhoeae alone. We found no evidence that C. muridarum increases gonococcal adherence to, or invasion of, immortalized murine epithelial cells. However, increased vaginal concentrations of inflammatory mediators macrophage inflammatory protein 2 and tumor necrosis factor alpha were detected in C. muridarum-infected mice prior to inoculation with N. gonorrhoeae concurrently with the downregulation of cathelicidin-related antimicrobial peptide and secretory leukocyte peptidase inhibitor genes. We conclude that female mice can be successfully infected with both C. muridarum and N. gonorrhoeae and that chlamydia-induced alterations in host innate responses may enhance gonococcal infection.
Collapse
Affiliation(s)
- Rachel A. Vonck
- Department of Microbiology and Immunology, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, Maryland 20814, Departments of Pediatrics and Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15201, Department of Pediatrics, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15201
| | - T. Darville
- Department of Microbiology and Immunology, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, Maryland 20814, Departments of Pediatrics and Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15201, Department of Pediatrics, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15201
| | - C. M. O'Connell
- Department of Microbiology and Immunology, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, Maryland 20814, Departments of Pediatrics and Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15201, Department of Pediatrics, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15201
| | - Ann E. Jerse
- Department of Microbiology and Immunology, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, Maryland 20814, Departments of Pediatrics and Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15201, Department of Pediatrics, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15201
| |
Collapse
|
31
|
Corthésy B. Role of secretory immunoglobulin A and secretory component in the protection of mucosal surfaces. Future Microbiol 2010; 5:817-29. [PMID: 20441552 DOI: 10.2217/fmb.10.39] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The contribution of secretory immunoglobulin A (SIgA) antibodies in the defense of mucosal epithelia plays an important role in preventing pathogen adhesion to host cells, therefore blocking dissemination and further infection. This mechanism, referred to as immune exclusion, represents the dominant mode of action of the antibody. However, SIgA antibodies combine multiple facets, which together confer properties extending from intracellular and serosal neutralization of antigens, activation of non-inflammatory pathways and homeostatic control of the endogenous microbiota. The sum of these features suggests that future opportunities for translational application from research-based knowledge to clinics include the mucosal delivery of bioactive antibodies capable of preserving immunoreactivity in the lung, gastrointestinal tract, the genito-urinary tract for the treatment of infections. This article covers topics dealing with the structure of SIgA, the dissection of its mode of action in epithelia lining different mucosal surfaces and its potential in immunotherapy against infectious pathogens.
Collapse
Affiliation(s)
- Blaise Corthésy
- R&D Laboratory of the Department of Immunology & Allergy, University State Hospital (CHUV), Rue du Bugnon 46, 1011 Lausanne, Switzerland.
| |
Collapse
|
32
|
Ambatipudi KS, Lu B, Hagen FK, Melvin JE, Yates JR. Quantitative analysis of age specific variation in the abundance of human female parotid salivary proteins. J Proteome Res 2009; 8:5093-102. [PMID: 19764810 PMCID: PMC2834885 DOI: 10.1021/pr900478h] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human saliva is a protein-rich, easily accessible source of potential local and systemic biomarkers to monitor changes that occur under pathological conditions; however, little is known about the changes in abundance associated with normal aging. In this study, we performed a comprehensive proteomic profiling of pooled saliva collected from the parotid glands of healthy female subjects, divided into two age groups 1 and 2 (20-30 and 55-65 years old, respectively). Hydrophobic charge interaction chromatography was used to separate high- from low-abundance proteins prior to characterization of the parotid saliva using multidimensional protein identification technology (MudPIT). Collectively, 532 proteins were identified in the two age groups. Of these proteins, 266 were identified exclusively in one age group, while 266 proteins were common to both groups. The majority of the proteins identified in the two age groups belonged to the defense and immune response category. Of note, several defense related proteins (e.g., lysozyme, lactoferrin and histatin-1) were significantly more abundant in group 2 as determined by G-test. Selected representative mass spectrometric findings were validated by Western blot analysis. Our study reports the first quantitative analysis of differentially regulated proteins in ductal saliva collected from young and older female subjects. This study supports the use of high-throughput proteomics as a robust discovery tool. Such results provide a foundation for future studies to identify specific salivary proteins which may be linked to age-related diseases specific to women.
Collapse
Affiliation(s)
- Kiran S. Ambatipudi
- Center for Oral Biology, University of Rochester Medical Center; Rochester, New York 14642
| | - Bingwen Lu
- Department of Chemical Physiology, The Scripps Research Institute; 10550 North Torrey Pines Road, SR-11, La Jolla, CA 92037
| | - Fred K Hagen
- Center for Oral Biology, University of Rochester Medical Center; Rochester, New York 14642
| | - James E. Melvin
- Center for Oral Biology, University of Rochester Medical Center; Rochester, New York 14642
| | - John R. Yates
- Department of Chemical Physiology, The Scripps Research Institute; 10550 North Torrey Pines Road, SR-11, La Jolla, CA 92037
| |
Collapse
|
33
|
Mestecky J, Russell MW. Specific antibody activity, glycan heterogeneity and polyreactivity contribute to the protective activity of S-IgA at mucosal surfaces. Immunol Lett 2009; 124:57-62. [PMID: 19524784 PMCID: PMC2697127 DOI: 10.1016/j.imlet.2009.03.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 03/26/2009] [Indexed: 01/19/2023]
Abstract
An explanation of the principles and mechanisms involved in peaceful co-existence between animals and the huge, diverse, and ever-changing microbiota that resides on their mucosal surfaces represents a challenging puzzle that is fundamental in everyday survival. In addition to mechanical barriers and a variety of innate defense factors, mucosal immunoglobulins (Igs) provide protection by two complementary mechanisms: specific antibody activity and innate, Ig glycan-mediated binding, both of which serve to contain the mucosal microbiota in its physiological niche. Thus, the interaction of bacterial ligands with IgA glycans constitutes a discrete mechanism that is independent of antibody specificity and operates primarily in the intestinal tract. This mucosal site is by far the most heavily colonized with an enormously diverse bacterial population, as well as the most abundant production site for antibodies, predominantly of the IgA isotype, in the entire immune system. In embodying both adaptive and innate immune mechanisms within a single molecule, S-IgA maintains comprehensive protection of mucosal surfaces with economy of structure and function.
Collapse
Affiliation(s)
- Jiri Mestecky
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35295-2170, USA.
| | | |
Collapse
|
34
|
Park HY, Suh GY, Chung MP, Kim H, Kwon OJ, Chung MJ, Kim TS, Lee KS, Koh WJ. Comparison of clinical and radiographic characteristics between nodular bronchiectatic form of nontuberculous mycobacterial lung disease and diffuse panbronchiolitis. J Korean Med Sci 2009; 24:427-32. [PMID: 19543504 PMCID: PMC2698187 DOI: 10.3346/jkms.2009.24.3.427] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2008] [Accepted: 07/25/2008] [Indexed: 12/02/2022] Open
Abstract
The nodular bronchiectatic form of nontuberculous mycobacterial (NTM) lung disease and diffuse panbronchiolits (DPB) show similar clinical and radiographic findings. The present study was performed to clarify the clinicoradiographic similarities as well as the differences between NTM lung disease and DPB. The initial clinicoradiographic features of 78 patients with the nodular bronchiectatic form of NTM lung disease (41 patients with Mycobacterium avium complex infection and 37 patients with Mycobacterium abscessus infection) were compared with those of 35 patients with DPB. Old age, female sex, a history of tuberculosis treatment, and hemoptysis were related to NTM lung disease while exertional dyspnea, coarse crackles, history of sinusitis, obstructive abnormalities in pulmonary function tests, and hypoxemia were related to DPB. The number of lobes involved with bronchiolitis and bronchiectasis on chest computed tomography were more numerous in DPB patients. There is considerable overlap in the clinical and radiographic appearances of the nodular bronchiectatic form of NTM lung disease and DPB, although some clinicoradiographic features differ between two diseases. The correct diagnosis, including aggressive microbiologic evaluation, should be made for the appropriate management of patients presenting with bilateral bronchiectasis and bronchiolitis.
Collapse
Affiliation(s)
- Hye Yun Park
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Gee Young Suh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Man Pyo Chung
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hojoong Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - O Jung Kwon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Myung Jin Chung
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Sung Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyung Soo Lee
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won-Jung Koh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
35
|
Abstract
Immunoglobulin A (IgA) is the most enigmatic of immunoglobulins. It is by far the most abundant of human Igs, being present in the blood plasma at concentrations approximating 2–3mg/mL, as well as the dominant isotype in most secretions where its output amounts to some 5–8g/day in adults. Furthermore, its evolutionary origins appear to precede the synapsid– diapsid divergence in tetrapod phylogeny (>300 million years ago) because it is present in both mammals and birds and therefore possibly also in reptiles (reviewed in Peppard et al., 2005); an IgA-like molecule has now been identified in a lizard (Deza et al., 2007).
Collapse
|
36
|
Shen K, Gladitz J, Antalis P, Dice B, Janto B, Keefe R, Hayes J, Ahmed A, Dopico R, Ehrlich N, Jocz J, Kropp L, Yu S, Nistico L, Greenberg DP, Barbadora K, Preston RA, Post JC, Ehrlich GD, Hu FZ. Characterization, distribution, and expression of novel genes among eight clinical isolates of Streptococcus pneumoniae. Infect Immun 2006; 74:321-30. [PMID: 16368987 PMCID: PMC1346598 DOI: 10.1128/iai.74.1.321-330.2006] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Eight low-passage-number Streptococcus pneumoniae clinical isolates, each of a different serotype and a different multilocus sequence type, were obtained from pediatric participants in a pneumococcal vaccine trial. Comparative genomic analyses were performed with these strains and two S. pneumoniae reference strains. Individual genomic libraries were constructed for each of the eight clinical isolates, with an average insert size of approximately 1 kb. A total of 73,728 clones were picked for arraying, providing more than four times genomic coverage per strain. A subset of 4,793 clones were sequenced, for which homology searches revealed that 750 (15.6%) of the sequences were unique with respect to the TIGR4 reference genome and 263 (5.5%) clones were unrelated to any available streptococcal sequence. Hypothetical translations of the open reading frames identified within these novel sequences showed homologies to a variety of proteins, including bacterial virulence factors not previously identified in S. pneumoniae. The distribution and expression patterns of 58 of these novel sequences among the eight clinical isolates were analyzed by PCR- and reverse transcriptase PCR-based analyses, respectively. These unique sequences were nonuniformly distributed among the eight isolates, and transcription of these genes in planktonic cultures was detected in 81% (172/212) of their genic occurrences. All 58 novel sequences were transcribed in one or more of the clinical strains, suggesting that they all correspond to functional genes. Sixty-five percent (38/58) of these sequences were found in 50% or less of the clinical strains, indicating a significant degree of genomic plasticity among natural isolates.
Collapse
Affiliation(s)
- Kai Shen
- Center for Genomic Sciences, Allegheny-Singer Research Institute, Allegheny General Hospital, 320 East North Avenue, 11th Floor South Tower, Pittsburgh, PA 15212, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Henderson IR, Navarro-Garcia F, Desvaux M, Fernandez RC, Ala'Aldeen D. Type V protein secretion pathway: the autotransporter story. Microbiol Mol Biol Rev 2004; 68:692-744. [PMID: 15590781 PMCID: PMC539010 DOI: 10.1128/mmbr.68.4.692-744.2004] [Citation(s) in RCA: 604] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Gram-negative bacteria possess an outer membrane layer which constrains uptake and secretion of solutes and polypeptides. To overcome this barrier, bacteria have developed several systems for protein secretion. The type V secretion pathway encompasses the autotransporter proteins, the two-partner secretion system, and the recently described type Vc or AT-2 family of proteins. Since its discovery in the late 1980s, this family of secreted proteins has expanded continuously, due largely to the advent of the genomic age, to become the largest group of secreted proteins in gram-negative bacteria. Several of these proteins play essential roles in the pathogenesis of bacterial infections and have been characterized in detail, demonstrating a diverse array of function including the ability to condense host cell actin and to modulate apoptosis. However, most of the autotransporter proteins remain to be characterized. In light of new discoveries and controversies in this research field, this review considers the autotransporter secretion process in the context of the more general field of bacterial protein translocation and exoprotein function.
Collapse
Affiliation(s)
- Ian R Henderson
- Division of Immunity and Infection, University of Birmingham, Birmingham B15 2TT, UK.
| | | | | | | | | |
Collapse
|
38
|
Zhang Q, Finn A. Mucosal immunology of vaccines against pathogenic nasopharyngeal bacteria. J Clin Pathol 2004; 57:1015-21. [PMID: 15452151 PMCID: PMC1770445 DOI: 10.1136/jcp.2004.016253] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2004] [Indexed: 11/04/2022]
Abstract
The introduction of Haemophilus influenzae type b conjugate vaccines during the 1990s was followed by dramatic decreases both in the incidence of Haemophilus influenzae type b related invasive disease and in nasopharyngeal carriage of the organism. The extent of this effect has been influenced by the fact that Haemophilus influenzae type b conjugate vaccines reduce nasopharyngeal carriage and induce herd immunity. Based on the success of Haemophilus influenzae type b conjugate vaccines, chemical conjugation has been applied to the development of pneumococcal and meningococcal polysaccharide conjugate vaccines. Evidence has begun to accumulate that these new polysaccharide based conjugate vaccines can also reduce nasopharyngeal carriage and can induce immune responses at the local mucosal level, which may be responsible for these effects. This article reviews recent studies on mucosal immune responses induced by polysaccharide based vaccines and some protein vaccine antigens against several pathogenic nasopharyngeal bacteria, and discusses the mechanisms and functions of these immune responses that may help our understanding of mucosal immune responses to both immunisation and infection.
Collapse
Affiliation(s)
- Q Zhang
- Department of Clinical Sciences South Bristol, Institute of Child Health, University of Bristol, UBHT Education Centre, Upper Maudlin Street, Bristol BS2 8AE, UK
| | | |
Collapse
|
39
|
Sahai MA, Setiadi DH, Chass GA, Pai EF, Penke B, Csizmadia IG. A model study of the IgA hinge region: an exploratory study of selected backbone conformations of MeCO-l-Pro-l-Thr-NH-Me. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/j.theochem.2003.08.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
40
|
Naglik JR, Challacombe SJ, Hube B. Candida albicans secreted aspartyl proteinases in virulence and pathogenesis. Microbiol Mol Biol Rev 2003; 67:400-28, table of contents. [PMID: 12966142 PMCID: PMC193873 DOI: 10.1128/mmbr.67.3.400-428.2003] [Citation(s) in RCA: 821] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candida albicans is the most common fungal pathogen of humans and has developed an extensive repertoire of putative virulence mechanisms that allows successful colonization and infection of the host under suitable predisposing conditions. Extracellular proteolytic activity plays a central role in Candida pathogenicity and is produced by a family of 10 secreted aspartyl proteinases (Sap proteins). Although the consequences of proteinase secretion during human infections is not precisely known, in vitro, animal, and human studies have implicated the proteinases in C. albicans virulence in one of the following seven ways: (i) correlation between Sap production in vitro and Candida virulence, (ii) degradation of human proteins and structural analysis in determining Sap substrate specificity, (iii) association of Sap production with other virulence processes of C. albicans, (iv) Sap protein production and Sap immune responses in animal and human infections, (v) SAP gene expression during Candida infections, (vi) modulation of C. albicans virulence by aspartyl proteinase inhibitors, and (vii) the use of SAP-disrupted mutants to analyze C. albicans virulence. Sap proteins fulfill a number of specialized functions during the infective process, which include the simple role of digesting molecules for nutrient acquisition, digesting or distorting host cell membranes to facilitate adhesion and tissue invasion, and digesting cells and molecules of the host immune system to avoid or resist antimicrobial attack by the host. We have critically discussed the data relevant to each of these seven criteria, with specific emphasis on how this proteinase family could contribute to Candida virulence and pathogenesis.
Collapse
Affiliation(s)
- Julian R Naglik
- Department of Oral Medicine, Pathology & Immunology, GKT Dental Institute, Kings College London, London, United Kingdom.
| | | | | |
Collapse
|
41
|
Watanabe K, Fujimura M, Kasahara K, Yasui M, Myou S, Watanabe A, Nakao S. Characteristics of pulmonary Mycobacterium avium-intracellulare complex (MAC) infection in comparison with those of tuberculosis. Respir Med 2003; 97:654-9. [PMID: 12814150 DOI: 10.1053/rmed.2003.1496] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
To clarify the clinical features of pulmonary Mycobacterium avium-intracellulare complex (MAC) infection, we retrospectively evaluated clinical manifestations, laboratory data, chest and maxillary sinus computed tomographic scans and induced sputum findings in 26 with MAC infection in comparison with 104 patients with tuberculosis (TB) infection. We found that carbohydrate antigen 19-9 (CA 19-9) and immunoglobulin A (IgA) in the serum and percentage of neutrophils in the sputum were significantly higher, and sinusitis was significantly more frequent in patients with MAC compared with patients with TB. MAC infection might be more strongly associated with impaired upper and lower airway defense mechanism in comparison with TB.
Collapse
Affiliation(s)
- K Watanabe
- Division of Pulmonary Medicine, Department of Internal Medicine, Sinminato Municipal Hospital, Japan.
| | | | | | | | | | | | | |
Collapse
|
42
|
Zhang Y, Pacheco S, Acuna CL, Switzer KC, Wang Y, Gilmore X, Harriman GR, Mbawuike IN. Immunoglobulin A-deficient mice exhibit altered T helper 1-type immune responses but retain mucosal immunity to influenza virus. Immunology 2002; 105:286-94. [PMID: 11918690 PMCID: PMC1782659 DOI: 10.1046/j.0019-2805.2001.01368.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
We have previously demonstrated that immunoglobulin A (IgA)(-/-) knockout (KO) mice exhibit levels of susceptibility to influenza virus infection that are similar to those of their normal IgA(+/+) littermates. To understand the mechanism of this apparent mucosal immunity without IgA, immunoglobulin isotype and T helper 1 (Th1)-type [interferon-gamma (IFN-gamma)] and Th2-type [interleukin (IL)-4, IL-5)] cytokine responses to influenza vaccine were evaluated. Intranasal immunization with influenza virus subunit vaccine plus cholera toxin/cholera toxin B subunit (CT/CTB) induced significant influenza virus-specific immunoglobulin G (IgG) antibody in the serum and nasal passages of both IgA(-/-) and IgA(+/+) mice, while IgA antibodies were induced only in IgA(+/+) mice. IgA KO mice exhibited an IgG1 subclass haemagglutinin (HA)-specific response but no detectable IgG2a and IgG2b responses. In contrast, IgA(+/+) mice exhibited significant IgG1 as well as IgG2a responses. This indicates a predominant Th2-type response in IgA KO mice compared to normal mice. Following stimulation with influenza virus in vitro, splenic lymphocytes from immunized IgA(-/-) mice produced significantly lower levels of IFN-gamma than IgA(+/+) mice (P < 0.001), but elaborated similar levels of IL-4 and IL-5. This was true at both protein and mRNA levels. Immunized mice were challenged intranasally with a small inoculum of influenza virus to allow deposition of virus in the nasal mucosal passages. Compared to non-immunized mice, immunized IgA(-/-) and IgA(+/+) mice exhibited significant, but similar levels of reduction in virus titres in the nose and lung. These results demonstrate that in addition to IgA deficiency, IgA gene deletion also resulted in down-regulated Th1-type immune responses and confirm our previous data that IgA antibody is not indispensable for the prevention of influenza virus infection.
Collapse
Affiliation(s)
- Yongxin Zhang
- Influenza Research Center, Respiratory Pathogens Research Unit, Department of Molecular Virology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Many pathogens must surmount an epithelial cell barrier in order to establish an infection. While much has been learned about the interaction of bacterial pathogens with cultured epithelial cells, the influence of cell polarity on these events has only recently been appreciated. This review outlines bacterial-host epithelial cell interactions in the context of the distinct apical and basolateral surfaces of the polarized epithelium that lines the lumens of our organs.
Collapse
Affiliation(s)
- B I Kazmierczak
- Department of Medicine, University of California, San Francisco, California 94143-0654, USA.
| | | | | |
Collapse
|
44
|
Scinicariello F, Attanasio R. Intraspecies heterogeneity of immunoglobulin alpha-chain constant region genes in rhesus macaques. Immunology 2001; 103:441-8. [PMID: 11529934 PMCID: PMC1783266 DOI: 10.1046/j.1365-2567.2001.01251.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Immunoglobulin A (IgA) is the major antibody class present in external secretions and is also an important component of serum immunoglobulins. On mucosal surfaces, IgA represents a first line of defence by neutralizing invading pathogens. The number of IgA constant-region genes (C alpha) present in different mammalian species is variable. Immunoglobulin C alpha genes differ mainly in the sequences located in the hinge region. IgA molecules, whose hinge regions are remarkably similar to those of the respective human molecules, are present in hominoid primates. In this report, we show that two alleles of a single immunoglobulin C alpha are present in rhesus macaques (Macaca mulatta). In addition, we show that intraspecies immunoglobulin C alpha allelic polymorphism is very high in this non-human primate species. Specifically, five different hinge regions, some of which are proline-rich, were identified from a total of eight rhesus macaque immunoglobulin C alpha-chains. The five hinge regions were different from those present in hominoid primates, both in length and in sequence. These results represent the first example of high levels of intraspecies immunoglobulin constant-region variability and suggest that IgAs of variable structure and function may be present in rhesus macaques. As rhesus macaques are widely used as animal models for the development of vaccines for acquired immune deficiency syndrome (AIDS), the possible presence of structurally and functionally variable IgA molecules in different animals should be taken into account when designing experimental strategies to induce mucosal antibody responses to human immunodeficiency virus (HIV).
Collapse
|
45
|
Zhang JR, Mostov KE, Lamm ME, Nanno M, Shimida S, Ohwaki M, Tuomanen E. The polymeric immunoglobulin receptor translocates pneumococci across human nasopharyngeal epithelial cells. Cell 2000; 102:827-37. [PMID: 11030626 DOI: 10.1016/s0092-8674(00)00071-4] [Citation(s) in RCA: 284] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The polymeric immunoglobulin receptor (pIgR) plays a crucial role in mucosal immunity against microbial infection by transporting polymeric immunoglobulins (pIg) across the mucosal epithelium. We report here that the human pIgR (hpIgR) can bind to a major pneumococcal adhesin, CbpA. Expression of hpIgR in human nasopharyngeal cells and MDCK cells greatly enhanced pneumococcal adherence and invasion. The hpIgR-mediated bacterial adherence and invasion were abolished by either insertional knockout of cbpA or antibodies against either hpIgR or CbpA. In contrast, rabbit pIgR (rpIgR) did not bind to CbpA and its expression in MDCK cells did not enhance pneumococcal adherence and invasion. These results suggest that pneumococci are a novel example of a pathogen co-opting the pIg transcytosis machinery to promote translocation across a mucosal barrier.
Collapse
Affiliation(s)
- J R Zhang
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Partidos CD. Intranasal vaccines: forthcoming challenges. PHARMACEUTICAL SCIENCE & TECHNOLOGY TODAY 2000; 3:273-281. [PMID: 10916147 DOI: 10.1016/s1461-5347(00)00281-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The mucosal epithelium of the upper respiratory tract constitutes an effective physical barrier to many pathogens. Its mucosal-associated lymphoid tissue is of particular importance for the protection and integrity of mucosal surfaces and the body's interior. Understanding the factors that influence the induction and regulation of mucosal immune responses will facilitate the design of vaccines capable of eliciting the appropriate type of protective immune response.
Collapse
Affiliation(s)
- CD Partidos
- UPR 9021 CNRS, Immunochimie des Peptides et des Virus, Institut de Biologie Moleculaire et Cellulaire, 15 rue René Descartes, F-67084 Strasbourg Cedex, France
| |
Collapse
|
47
|
Abstract
Group A streptococci are model extracellular gram-positive pathogens responsible for pharyngitis, impetigo, rheumatic fever, and acute glomerulonephritis. A resurgence of invasive streptococcal diseases and rheumatic fever has appeared in outbreaks over the past 10 years, with a predominant M1 serotype as well as others identified with the outbreaks. emm (M protein) gene sequencing has changed serotyping, and new virulence genes and new virulence regulatory networks have been defined. The emm gene superfamily has expanded to include antiphagocytic molecules and immunoglobulin-binding proteins with common structural features. At least nine superantigens have been characterized, all of which may contribute to toxic streptococcal syndrome. An emerging theme is the dichotomy between skin and throat strains in their epidemiology and genetic makeup. Eleven adhesins have been reported, and surface plasmin-binding proteins have been defined. The strong resistance of the group A streptococcus to phagocytosis is related to factor H and fibrinogen binding by M protein and to disarming complement component C5a by the C5a peptidase. Molecular mimicry appears to play a role in autoimmune mechanisms involved in rheumatic fever, while nephritis strain-associated proteins may lead to immune-mediated acute glomerulonephritis. Vaccine strategies have focused on recombinant M protein and C5a peptidase vaccines, and mucosal vaccine delivery systems are under investigation.
Collapse
Affiliation(s)
- M W Cunningham
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
48
|
Fujieda S, Suzuki S, Sunaga H, Yamamoto H, Seki M, Sugimoto H, Saito H. Induction of IgA against Haemophilus parainfluenzae antigens in tonsillar mononuclear cells from patients with IgA nephropathy. Clin Immunol 2000; 95:235-43. [PMID: 10866131 DOI: 10.1006/clim.2000.4864] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Much evidence suggests that IgA production in vivo and in vitro is enhanced in patients with IgA nephropathy (IgAN). We have demonstrated glomerular deposition of the outer membranes of Haemophilus parainfluenzae (HP) antigens (OMHP) and the presence of HP-specific IgA in the serum of patients with IgAN. In this study, we investigated the production of IgA and several cytokines by tonsillar mononuclear cells (TMC) from IgAN patients induced by stimulation with OMHP. The spontaneous production of total IgA and TGF-beta by TMC from IgAN patients was higher than that by TMC from patients with chronic tonsillitis (CT) (P < 0.05). Stimulation with OMHP in vitro enhanced the production of HP-specific IgA by TMC from IgAN patients (P < 0.01), but not by TMC from CT patients. OMHP stimulation also enhanced the production of TGF-beta and IL-10 by TMC from IgAN patients (P < 0.001). These results suggest that the infection of HP in the tonsil may be involved in the etiology of IgAN.
Collapse
Affiliation(s)
- S Fujieda
- Department of Otorhinolaryngology, Fukui Medical University, Matsuoka, Yoshida, Japan.
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Amebiasis is a major cause of morbidity and mortality throughout the tropical world. Entamoeba histolytica is now recognized as a separate species from the morphologically identical E. dispar, which cannot invade. Cysteine proteinases are a key virulence factor of E. histolytica and play a role in intestinal invasion by degrading the extracellular matrix and circumventing the host immune response through cleavage of secretory immunoglobulin A (sIgA), IgG, and activation of complement. Cysteine proteinases are encoded by at least seven genes, several of which are found in E. histolytica but not E. dispar. A number of new animal models, including the formation of liver abscesses in SCID mice and intestinal infection in human intestinal xenografts, have proven useful to confirm the critical role of cysteine proteinases in invasion. Detailed structural analysis of cysteine proteinases should provide further insights into their biochemical function and may facilitate the design of specific inhibitors which could be used as potential chemotherapeutic agents in the future.
Collapse
|
50
|
Hammerschmidt S, Tillig MP, Wolff S, Vaerman JP, Chhatwal GS. Species-specific binding of human secretory component to SpsA protein of Streptococcus pneumoniae via a hexapeptide motif. Mol Microbiol 2000; 36:726-36. [PMID: 10844660 DOI: 10.1046/j.1365-2958.2000.01897.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
SpsA, a pneumococcal surface protein belonging to the family of choline-binding proteins, interacts specifically with secretory immunglobulin A (SIgA) via the secretory component (SC). SIgA and free SC from mouse, rat, rabbit and guinea-pig failed to interact with SpsA indicating species-specific binding to human SIgA and SC. SpsA is the only pneumococcal receptor molecule for SIgA and SC as confirmed by complete loss of SIgA and SC binding to a spsA mutant. Analysis of recombinant SpsA fusion proteins showed that the binding domain is located in the N-terminal region of SpsA. By the use of different truncated N-terminal SpsA fusion proteins, the minimum binding domain was shown to be composed of 112 amino acids (residues 172-283). The sequence of this 112-amino-acids domain was used to spot synthesize 34 overlapping peptides, consisting of 15 amino acids each, with an offset of three amino acids on a cellulose membrane. One of the peptides reacted specifically with both SIgA and SC. By using a second membrane with immobilized synthetic peptides of decreasing length containing parts of the identified 15-amino-acid motif a hexapeptide, YRNYPT was identified as the binding motif for SC and SIgA. SpsA proteins with a size smaller than the assay-positive domain of 112 amino acids were able to inhibit the interaction of SIgA and pneumococci provided they contained the binding motif. The results indicated that the hexapeptide YRNYPT located in SpsA of pneumococcal strain type 1 (ATCC 33400) between amino acids 198 and 203 is involved in SIgA and SC binding. Because synthetic peptides containing only parts of the hexapeptide also assayed positive, these results further suggest that at least the amino acids YPT of the identified hexapeptide are critical for binding to SC and SIgA. Amino acid substitutions in the identified putative binding motif abolished SC-/SIgA-binding activity of the mutated SpsA protein, confirming the functional activity of this hexapeptide and the critical role of the amino acids YPT in SC and SIgA binding. Identification of this motif, which is highly conserved in SpsA protein among different serotypes, might contribute towards a new peptide based vaccine strategy.
Collapse
Affiliation(s)
- S Hammerschmidt
- Department of Microbial Pathogenesis, GBF-German Research Centre for Biotechnology, D-38124 Braunschweig, Germany
| | | | | | | | | |
Collapse
|