1
|
Sharma M, Wagh P, Shinde T, Trimbake D, Tripathy AS. Exploring the Role of Pattern Recognition Receptors as Immunostimulatory Molecules. Immun Inflamm Dis 2025; 13:e70150. [PMID: 40396589 DOI: 10.1002/iid3.70150] [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: 05/07/2024] [Revised: 12/09/2024] [Accepted: 01/21/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Pattern recognition receptors (PRRs) are the receptors of the innate immune system that play a vital role in initiating innate immune response. PRRs recognize pathogen associated molecular patterns (PAMPs) and activate immune cells through a signaling cascade. Due to this remarkable ability to recognize pathogenic microbes and elucidation of an immune response in a well-organized manner, PRR agonizts are likely to have great potential as vaccine adjuvants. Recent advancements in vaccine development raised concerns regarding the reduced immunogenicity of various vaccines, questioning the vaccine efficacy. In such cases, the use of an adjuvant becomes crucial. Understanding the structure and downstream signaling of PRRs will provide the possibility of developing a novel therapeutic approach. METHOD The rapidly evolving field of immunology and vaccinology, coupled with the increasing focus on PRRs in disease therapy, demands a comprehensive overview. In this review, we provide all-inclusive and contemporary gist on PRRs and the applications of their agonizts. We explored the potential of PRR agonizts as vaccine adjuvant. The current review integrates the basic understanding of PRRs and recent findings highlighting emerging trends of the same. RESULT Our review highlights that combining multiple PRR agonizts could offer synergistic benefits. This approach might prove advantageous and could potentially enhance vaccine efficacy and reduce the need for excessive immunogens. CONCLUSION A comprehensive understanding of PRR subset, agonizts of PRR and their application in vaccine adjuvant. This knowledge will be significant in formulating vaccine approaches.
Collapse
Affiliation(s)
- Meenal Sharma
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, Pune, India
| | - Priyanka Wagh
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, Pune, India
| | - Tanvi Shinde
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, Pune, India
| | - Diptee Trimbake
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, Pune, India
| | - Anuradha S Tripathy
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, Pune, India
| |
Collapse
|
2
|
Jiang Y, Dhungana A, Odunfa OA, McCoun M, McGill J, Yoon I, Ogunade I. Effects of Saccharomyces cerevisiae fermentation product on ruminal fermentation, total tract digestibility, blood proinflammatory cytokines, and plasma metabolome of Holstein steers fed a high-grain diet. Transl Anim Sci 2025; 9:txaf058. [PMID: 40391287 PMCID: PMC12086543 DOI: 10.1093/tas/txaf058] [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: 12/26/2024] [Accepted: 04/23/2025] [Indexed: 05/21/2025] Open
Abstract
This study aimed to assess the impact of Saccharomyces cerevisiae fermentation product (SCFP) on digestibility, ruminal fermentation, and plasma metabolome of Holstein steers fed a high-grain diet. Steers were fed diet with 80% concentrate and 20% corn silage once daily ad libitum. Steers were stratified based on initial body weight (BW) and randomly assigned to two treatments: 1) control (CON), a basal diet without SCFP; 2) basal diet + 12 g/head/day SCFP, top-dressed. Eight rumen-cannulated Holstein steers (BW: 580 ± 29.2 kg) were enrolled in a crossover design study with 25-d treatment periods and a 24-d washout period. Dry matter intake (DMI) was calculated from daily feed offered and refusals. Blood was collected before morning feeding on day 25 of each period. Rumen fluid was collected at 0, 4, 8 and 12 h post-feeding on d 25. Fecal samples were collected from d 22 to 24 for digestibility measures. Statistical analyses were performed with the GLIMMIX procedure of SAS 9.4 (SAS, 2023). Supplementing SCFP had no effect on digestibility of organic matter (OM, P = 0.63), crude protein (CP, P = 0.97), neutral detergent fiber (NDF, P = 0.59), and acid detergent fiber (ADF, P = 0.84). Treatment did not affect fecal excretion of nitrogen (N, P = 0.69), phosphorus (P, P = 0.24), copper (Cu, P = 0.71), and zinc (Zn, P = 0.95). Supplementing SCFP increased (6.29 vs. 6.01, P = 0.01) ruminal pH compared to CON. Lactic acid concentrations were similar between treatments (P = 0.17) and low in both groups (0.09mM vs. 0.28mM). Treatment did not affect ruminal total volatile fatty acid (VFA) concentrations (P > 0.10) but decreased butyrate molar proportion (P = 0.01) and tended to increase the molar proportions of isobutyrate (P = 0.06) and isovalerate (P ≤ 0.10). Treatment had no effect on the in vitro production of proinflammatory cytokines, IL-1β (P > 0.11) and IL-6 (P > 0.12), in the whole blood in response to various toll-like receptor stimulants. Plasma pathways of purine metabolism, amino sugar and nucleotide sugar metabolism, and lysine degradation were enriched (P ≤ 0.05) by feeding SCFP. Overall, supplementing SCFP did not affect total tract digestibility, fecal excretions of macro minerals but enhanced ruminal pH in cattle fed a high-grain diet. Furthermore, feeding SCFP enriched several important plasma pathways related to protein metabolism.
Collapse
Affiliation(s)
- Yun Jiang
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Anjan Dhungana
- School of Agriculture and Natural Resources, Kentucky State University, Frankfort, KY, 40601, USA
| | - Oluwaseun A Odunfa
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Megan McCoun
- School of Agriculture and Natural Resources, Kentucky State University, Frankfort, KY, 40601, USA
| | - Jodi McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011, USA
| | | | - Ibukun Ogunade
- Division of Animal and Nutritional Science, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
3
|
Henige M, Anklam K, Aviles M, Buettner J, Henschel S, Yoon I, Wheeler J, Dawson G, McGill J, Döpfer D. The Effect of Saccharomyces cerevisiae Fermentation Product Supplementation on Pro-Inflammatory Cytokines in Holstein Friesian Cattle Experimentally Inoculated with Digital Dermatitis. Animals (Basel) 2024; 14:3260. [PMID: 39595313 PMCID: PMC11591135 DOI: 10.3390/ani14223260] [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: 10/02/2024] [Revised: 11/02/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Digital dermatitis (DD) poses a major animal welfare concern for the dairy industry, with even broader economic implications for the agricultural industry worldwide. The postbiotic, a Saccharomyces cerevisiae fermentation product (SCFP), has had a positive influence on the innate immune system of cattle, which makes it a potential candidate as a feed supplement as part of a prevention strategy for DD. This study investigated the effect of a commercial SCFP feed supplement compared to a control feed supplement on the production of pro-inflammatory cytokines (IL-1β and IL-6) by peripheral blood mononuclear cells (PBMCs) in Holstein Friesian steers experimentally infected with DD. The results showed that SCFP supplementation was associated with an overall reduced IL-1β production (p = 0.005), particularly prior to experimental inoculation with a DD lesion homogenate. However, the results of the analysis suggest that the innate immune system in the SCFP group became prepared to respond more rapidly to DD infection post-inoculation. During active (M2), chronic (M4), and focal flare-ups (M4.1) of DD, SCFP supplementation resulted in a more rapid secretion of IL-1β (M2: p = 0.038; M4/M4/1: p = 0.034). A more rapid response to DD infection for IL-6 was only found for chronic (M4) and focal flare-ups (M4.1) of DD (p = 0.006). These findings emphasize the difference in cytokine response between various stages of DD in the SCFP group compared to the control, highlighting implications for DD prevention and treatment.
Collapse
Affiliation(s)
- Marlee Henige
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.A.); (M.A.); (J.B.); (S.H.); (D.D.)
| | - Kelly Anklam
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.A.); (M.A.); (J.B.); (S.H.); (D.D.)
| | - Matthew Aviles
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.A.); (M.A.); (J.B.); (S.H.); (D.D.)
| | - Julia Buettner
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.A.); (M.A.); (J.B.); (S.H.); (D.D.)
| | - Summer Henschel
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.A.); (M.A.); (J.B.); (S.H.); (D.D.)
| | - Ilkyu Yoon
- Diamond V, Cedar Rapids, IA 52404, USA; (I.Y.); (J.W.); (G.D.)
| | - Jeffrey Wheeler
- Diamond V, Cedar Rapids, IA 52404, USA; (I.Y.); (J.W.); (G.D.)
| | - George Dawson
- Diamond V, Cedar Rapids, IA 52404, USA; (I.Y.); (J.W.); (G.D.)
| | - Jodi McGill
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA 50011, USA;
| | - Dörte Döpfer
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA; (K.A.); (M.A.); (J.B.); (S.H.); (D.D.)
| |
Collapse
|
4
|
Zhu Y, Wei L, Zwygart ACA, Gaínza P, Khac QO, Olgiati F, Kurum A, Tang L, Correia B, Tapparel C, Stellacci F. A Synthetic Multivalent Lipopeptide Derived from Pam3CSK4 with Irreversible Influenza Inhibition and Immuno-Stimulating Effects. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307709. [PMID: 38438885 DOI: 10.1002/smll.202307709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 02/23/2024] [Indexed: 03/06/2024]
Abstract
The activation of the host adaptive immune system is crucial for eliminating viruses. However, influenza infection often suppresses the innate immune response that precedes adaptive immunity, and the adaptive immune responses are typically delayed. Dendritic cells, serving as professional antigen-presenting cells, have a vital role in initiating the adaptive immune response. In this study, an immuno-stimulating antiviral system (ISAS) is introduced, which is composed of the immuno-stimulating adjuvant lipopeptide Pam3CSK4 that acts as a scaffold onto which it is covalently bound 3 to 4 influenza-inhibiting peptides. The multivalent display of peptides on the scaffold leads to a potent inhibition against H1N1 (EC50 = 20 nM). Importantly, the resulting lipopeptide, Pam3FDA, shows an irreversible inhibition mechanism. The chemical modification of peptides on the scaffold maintains Pam3CSK4's ability to stimulate dendritic cell maturation, thereby rendering Pam3FDA a unique antiviral. This is attributed to its immune activation capability, which also acts in synergy to expedite viral elimination.
Collapse
Affiliation(s)
- Yong Zhu
- Institute of Materials, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Lixia Wei
- Institute of Materials, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Arnaud Charles-Antoine Zwygart
- Department of Microbiology and Molecular Medicine, University of Geneva, CMU Rue Michel-Servet 1, Geneva 4, CH-1211, Switzerland
| | - Pablo Gaínza
- Interschool Institute of Bioengineering, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Quy Ong Khac
- Institute of Materials, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Francesca Olgiati
- Institute of Materials, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Armand Kurum
- Interschool Institute of Bioengineering, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Li Tang
- Interschool Institute of Bioengineering, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Bruno Correia
- Interschool Institute of Bioengineering, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, University of Geneva, CMU Rue Michel-Servet 1, Geneva 4, CH-1211, Switzerland
| | - Francesco Stellacci
- Institute of Materials, École Polytechnique Fédérale de Lausanne Station 12, Lausanne, CH-1015, Switzerland
| |
Collapse
|
5
|
Griesman T, McMillen CM, Negatu SG, Hulahan JJ, Whig K, Dohnalová L, Dittmar M, Thaiss CA, Jurado KA, Schultz DC, Hartman AL, Cherry S. The lipopeptide Pam3CSK4 inhibits Rift Valley fever virus infection and protects from encephalitis. PLoS Pathog 2024; 20:e1012343. [PMID: 38935789 PMCID: PMC11236204 DOI: 10.1371/journal.ppat.1012343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/10/2024] [Accepted: 06/12/2024] [Indexed: 06/29/2024] Open
Abstract
Rift Valley fever virus (RVFV) is an encephalitic bunyavirus that can infect neurons in the brain. There are no approved therapeutics that can protect from RVFV encephalitis. Innate immunity, the first line of defense against infection, canonically antagonizes viruses through interferon signaling. We found that interferons did not efficiently protect primary cortical neurons from RVFV, unlike other cell types. To identify alternative neuronal antiviral pathways, we screened innate immune ligands and discovered that the TLR2 ligand Pam3CSK4 inhibited RVFV infection, and other bunyaviruses. Mechanistically, we found that Pam3CSK4 blocks viral fusion, independent of TLR2. In a mouse model of RVFV encephalitis, Pam3CSK4 treatment protected animals from infection and mortality. Overall, Pam3CSK4 is a bunyavirus fusion inhibitor active in primary neurons and the brain, representing a new approach toward the development of treatments for encephalitic bunyavirus infections.
Collapse
Affiliation(s)
- Trevor Griesman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia Pennsylvania, United States of America
| | - Cynthia M. McMillen
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Seble Getenet Negatu
- Department of Microbiology, University of Pennsylvania, Philadelphia Pennsylvania, Unites States of America
| | - Jesse J. Hulahan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia Pennsylvania, United States of America
| | - Kanupriya Whig
- High throughput screening core, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Lenka Dohnalová
- Department of Microbiology, University of Pennsylvania, Philadelphia Pennsylvania, Unites States of America
| | - Mark Dittmar
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia Pennsylvania, United States of America
| | - Christoph A. Thaiss
- Department of Microbiology, University of Pennsylvania, Philadelphia Pennsylvania, Unites States of America
| | - Kellie A. Jurado
- Department of Microbiology, University of Pennsylvania, Philadelphia Pennsylvania, Unites States of America
| | - David C. Schultz
- High throughput screening core, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Amy L. Hartman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia Pennsylvania, United States of America
- High throughput screening core, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
6
|
Lee JS, Kim C. Role of CARD9 in Cell- and Organ-Specific Immune Responses in Various Infections. Int J Mol Sci 2024; 25:2598. [PMID: 38473845 DOI: 10.3390/ijms25052598] [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: 12/12/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
The caspase recruitment domain-containing protein 9 (CARD9) is an intracellular adaptor protein that is abundantly expressed in cells of the myeloid lineage, such as neutrophils, macrophages, and dendritic cells. CARD9 plays a critical role in host immunity against infections caused by fungi, bacteria, and viruses. A CARD9 deficiency impairs the production of inflammatory cytokines and chemokines as well as migration and infiltration, thereby increasing susceptibility to infections. However, CARD9 signaling varies depending on the pathogen causing the infection. Furthermore, different studies have reported altered CARD9-mediated signaling even with the same pathogen. Therefore, this review focuses on and elucidates the current literature on varied CARD9 signaling in response to various infectious stimuli in humans and experimental mice models.
Collapse
Affiliation(s)
- Ji Seok Lee
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, Inha University School of Medicine, Incheon 22212, Republic of Korea
- BK21, Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Chaekyun Kim
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, Inha University School of Medicine, Incheon 22212, Republic of Korea
- BK21, Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
7
|
Wada FW, Desta AF, Gebre M, Mihret W, Seyoum T, Melaku K, Alemu A, Howe R, Mulu A, Mihret A. Pneumococcal colonization and coinfecting respiratory viruses in children under 5 years in Addis Ababa, Ethiopia: a prospective case-control study. Sci Rep 2024; 14:4174. [PMID: 38378681 PMCID: PMC10879120 DOI: 10.1038/s41598-024-54256-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/10/2024] [Indexed: 02/22/2024] Open
Abstract
A comprehensive understanding of the dynamics of Streptococcus pneumoniae colonization in conjunction with respiratory virus infections is essential for enhancing our knowledge of the pathogenesis and advancing the development of effective preventive strategies. Therefore, a case-control study was carried out in Addis Ababa, Ethiopia to investigate the colonization rate of S. pneumoniae and its coinfection dynamics with respiratory viruses among children under the age of 5 years. Samples from the nasopharyngeal and/or oropharyngeal, along with socio-demographic and clinical information, were collected from 420 children under 5 years old (210 cases with lower respiratory tract infections and 210 controls with conditions other than respiratory infections.). A one-step Multiplex real-time PCR using the Allplex Respiratory Panel Assays 1-4 was performed to identify respiratory viruses and bacteria. Data analysis was conducted using STATA software version 17. The overall colonization rate of S. pneumoniae in children aged less than 5 years was 51.2% (215/420). The colonization rates in cases and controls were 54.8% (115/210) and 47.6% (100/210), respectively (p = 0.14). Colonization rates were observed to commence at an early age in children, with a colonization rate of 48.9% and 52.7% among infants younger than 6 months controls and cases, respectively. The prevalence of AdV (OR, 3.11; 95% CI [1.31-8.19]), RSV B (OR, 2.53; 95% CI [1.01-6.78]) and HRV (OR, 1.7; 95% CI [1.04-2.78]) tends to be higher in children who tested positive for S. pneumoniae compared to those who tested negative for S. pneumoniae. Further longitudinal research is needed to understand and determine interaction mechanisms between pneumococci and viral pathogens and the clinical implications of this coinfection dynamics.
Collapse
Affiliation(s)
- Fiseha Wadilo Wada
- Armauer Hansen Research Institute, Ministry of Health, Addis Ababa, Ethiopia.
- Department of Biomedical Sciences, College of Natural and Computational Sciences, Addis Ababa University, Addis Ababa, Ethiopia.
- Department of Medical Laboratory Sciences, College of Health Sciences and Medicine, Wolaita Sodo University, Wolaita Soddo, Ethiopia.
| | - Adey Feleke Desta
- Department of Biomedical Sciences, College of Natural and Computational Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Meseret Gebre
- Armauer Hansen Research Institute, Ministry of Health, Addis Ababa, Ethiopia
| | - Wude Mihret
- Armauer Hansen Research Institute, Ministry of Health, Addis Ababa, Ethiopia
| | - Tamrayehu Seyoum
- Armauer Hansen Research Institute, Ministry of Health, Addis Ababa, Ethiopia
| | - Kalkidan Melaku
- Armauer Hansen Research Institute, Ministry of Health, Addis Ababa, Ethiopia
| | - Ashenafi Alemu
- Armauer Hansen Research Institute, Ministry of Health, Addis Ababa, Ethiopia
| | - Rawleigh Howe
- Armauer Hansen Research Institute, Ministry of Health, Addis Ababa, Ethiopia
| | - Andargachew Mulu
- Armauer Hansen Research Institute, Ministry of Health, Addis Ababa, Ethiopia
| | - Adane Mihret
- Armauer Hansen Research Institute, Ministry of Health, Addis Ababa, Ethiopia
- Department of Microbiology, Immunology, and Parasitology, School of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
8
|
Lalbiaktluangi C, Yadav MK, Singh PK, Singh A, Iyer M, Vellingiri B, Zomuansangi R, Zothanpuia, Ram H. A cooperativity between virus and bacteria during respiratory infections. Front Microbiol 2023; 14:1279159. [PMID: 38098657 PMCID: PMC10720647 DOI: 10.3389/fmicb.2023.1279159] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/27/2023] [Indexed: 12/17/2023] Open
Abstract
Respiratory tract infections remain the leading cause of morbidity and mortality worldwide. The burden is further increased by polymicrobial infection or viral and bacterial co-infection, often exacerbating the existing condition. Way back in 1918, high morbidity due to secondary pneumonia caused by bacterial infection was known, and a similar phenomenon was observed during the recent COVID-19 pandemic in which secondary bacterial infection worsens the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) condition. It has been observed that viruses paved the way for subsequent bacterial infection; similarly, bacteria have also been found to aid in viral infection. Viruses elevate bacterial infection by impairing the host's immune response, disrupting epithelial barrier integrity, expression of surface receptors and adhesion proteins, direct binding of virus to bacteria, altering nutritional immunity, and effecting the bacterial biofilm. Similarly, the bacteria enhance viral infection by altering the host's immune response, up-regulation of adhesion proteins, and activation of viral proteins. During co-infection, respiratory bacterial and viral pathogens were found to adapt and co-exist in the airways of their survival and to benefit from each other, i.e., there is a cooperative existence between the two. This review comprehensively reviews the mechanisms involved in the synergistic/cooperativity relationship between viruses and bacteria and their interaction in clinically relevant respiratory infections.
Collapse
Affiliation(s)
- C. Lalbiaktluangi
- Department of Microbiology, Central University of Punjab, Bathinda, Punjab, India
| | - Mukesh Kumar Yadav
- Department of Microbiology, Central University of Punjab, Bathinda, Punjab, India
| | - Prashant Kumar Singh
- Department of Biotechnology, Mizoram University (A Central University), Pachhunga University College, Aizawl, Mizoram, India
| | - Amit Singh
- Department of Microbiology, Central University of Punjab, Bathinda, Punjab, India
| | - Mahalaxmi Iyer
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | | | - Ruth Zomuansangi
- Department of Microbiology, Central University of Punjab, Bathinda, Punjab, India
| | - Zothanpuia
- Department of Biotechnology, Mizoram University (A Central University), Pachhunga University College, Aizawl, Mizoram, India
| | - Heera Ram
- Department of Zoology, Jai Narain Vyas University, Jodhpur, India
| |
Collapse
|
9
|
Melot L, Bankamp B, Rota PA, Coughlin MM. Characterizing infection of B cells with wild-type and vaccine strains of measles virus. iScience 2023; 26:107721. [PMID: 37736039 PMCID: PMC10510084 DOI: 10.1016/j.isci.2023.107721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 04/14/2023] [Accepted: 08/22/2023] [Indexed: 09/23/2023] Open
Abstract
Acute infection with measles virus (MeV) causes transient immunosuppression often leading to secondary infections. MeV infection of B lymphocytes results in changes in the antibody repertoire and memory B cell populations for which the mechanism is unknown. In this study, we characterize the infection of primary B cells with wild-type and vaccine strains of MeV. Vaccine-infected B cells were characterized by a higher percentage of cells positive for viral protein, a higher level of viral transcription and reduced cell death compared to wild-type infected cells, regardless of B cell subtype. Vaccine-infected cells showed more production of TNF-α and IL-10 but less production of IL-8 compared to wild-type infected cells. IL-4 and IL-6 levels detected were increased during both vaccine and wild-type infection. Despite evidence of replication, measles-infected B cells did not produce detectable viral progeny. This study furthers our understanding of the outcomes of MeV infection of human B cells.
Collapse
Affiliation(s)
- Logan Melot
- Viral Vaccine Preventable Diseases Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
- Emory University, Atlanta, GA 303333, USA
| | - Bettina Bankamp
- Viral Vaccine Preventable Diseases Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Paul A. Rota
- Viral Vaccine Preventable Diseases Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
- Emory University, Atlanta, GA 303333, USA
| | - Melissa M. Coughlin
- Viral Vaccine Preventable Diseases Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| |
Collapse
|
10
|
van Teijlingen NH, Eder J, Sarrami-Forooshani R, Zijlstra-Willems EM, Roovers JPWR, van Leeuwen E, Ribeiro CMS, Geijtenbeek TBH. Immune activation of vaginal human Langerhans cells increases susceptibility to HIV-1 infection. Sci Rep 2023; 13:3283. [PMID: 36841916 PMCID: PMC9968315 DOI: 10.1038/s41598-023-30097-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 02/14/2023] [Indexed: 02/27/2023] Open
Abstract
Vaginal inflammation increases the risk for sexual HIV-1 transmission but underlying mechanisms remain unclear. In this study we assessed the impact of immune activation on HIV-1 susceptibility of primary human vaginal Langerhans cells (LCs). Vaginal LCs isolated from human vaginal tissue expressed a broad range of TLRs and became activated after exposure to both viral and bacterial TLR ligands. HIV-1 replication was restricted in immature vaginal LCs as only low levels of infection could be detected. Notably, activation of immature vaginal LCs by bacterial TLR ligands increased HIV-1 infection, whereas viral TLR ligands were unable to induce HIV-1 replication in vaginal LCs. Furthermore, mature vaginal LCs transmitted HIV-1 to CD4 T cells. This study emphasizes the role for vaginal LCs in protection against mucosal HIV-1 infection, which is abrogated upon activation. Moreover, our data suggest that bacterial STIs can increase the risk of HIV-1 acquisition in women.
Collapse
Affiliation(s)
- Nienke H. van Teijlingen
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands
| | - Julia Eder
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands ,Amsterdam Institute for Infection & Immunity, Amsterdam, The Netherlands
| | - Ramin Sarrami-Forooshani
- grid.417689.5ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. BOX, Tehran, 15179/64311 Iran
| | - Esther M. Zijlstra-Willems
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands ,Amsterdam Institute for Infection & Immunity, Amsterdam, The Netherlands
| | - Jan-Paul W. R. Roovers
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Obstetrics and Gynaecology, Meibergdreef 9, Amsterdam, The Netherlands
| | - Elisabeth van Leeuwen
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Obstetrics and Gynaecology, Meibergdreef 9, Amsterdam, The Netherlands
| | - Carla M. S. Ribeiro
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands ,Amsterdam Institute for Infection & Immunity, Amsterdam, The Netherlands
| | - Teunis B. H. Geijtenbeek
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands ,Amsterdam Institute for Infection & Immunity, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Baloyi NN, Tugizimana F, Sitole LJJ. Metabolomics assessment of vitamin D impact in Pam3CSK4 stimulation. Mol Omics 2022; 18:397-407. [DOI: 10.1039/d1mo00377a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mycobacterium tuberculosis, a causative agent of tuberculosis, is amongst the leading causes of mycobacterial mortality worldwide. Although several studies have proposed the possible therapeutic role of vitamin D in antimycobacterial...
Collapse
|
12
|
Kimoto T. Development of a safe and effective novel synthetic mucosal adjuvant SF-10 derived from physiological metabolic pathways and function of human pulmonary surfactant. Vaccine 2021; 40:544-553. [PMID: 34887132 DOI: 10.1016/j.vaccine.2021.11.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
A safe and effective mucosal adjuvant is required for vaccination against influenza A virus (IAV) infection. Previously, we described that intranasally administration of surfacten®, a medicine derived from bovine pulmonary surfactant (PS), with IAV vaccine can induce IAV-specific IgA in the respiratory tract mucosa and IgG in serum. PS is secreted by alveolar type II cells and Clara cells and serves to reduce lung surface tension. PS finished its rules is incorporated by antigen presenting cells (APCs), such as alveolar macrophages and dendritic cells, and alveolar type II cells and rapidly metabolized. We focused on the metabolic pathways and rapid metabolic turnover of PS and developed a PS-based mucosal adjuvant. First, we determined the essential components of PS adjuvanticity and found that the complex of three PS lipids and surfactant protein-C can enhance to deliver the vaccine antigen and activate APCs. Later, we improved the safety, efficacy and ease of manufacture and finally succeeded in developing SF-10. The use of SF-10 with influenza split vaccine (HAv) (HAv-SF-10) enhances HAv incorporation into APCs both in vitro and in vivo, and intranasal instillation of HAv-SF-10 induced systemic and mucosal HAv-specific immunities in not only mice but also cynomolgus monkeys. The report that PS has physiological effects on the gastrointestinal mucosa prompted us develop a new SF-10-based vaccine that can be administered orally. In this review, We summarize our work on the development of clinically effective PS-based nasal and oral mucosal adjuvants for influenza vaccine.
Collapse
Affiliation(s)
- Takashi Kimoto
- Division of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University, Tokushima, Japan.
| |
Collapse
|
13
|
Lipopeptides against COVID-19 RNA-dependent RNA polymerase using molecular docking. Biomed J 2021; 44:S15-S24. [PMID: 34871815 PMCID: PMC8641408 DOI: 10.1016/j.bj.2021.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/01/2021] [Accepted: 11/19/2021] [Indexed: 12/31/2022] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) is caused by a novel virus that is responsible for the largest pandemic in recent times. Although numerous studies have explored methods to cope with COVID-19 and targeted drugs and vaccines have been developed, the spread of disease remains rapid due to the high infectivity and mutation capability of SARS-CoV-2, the causative virus of COVID-19. Therefore, there is an urgent necessity to seek more efficient treatments and approaches to combat the disease. Methods In this study, molecular docking was used to predict the binding of different lipopeptides, which exhibit significant biological functions, to the RNA-dependent RNA polymerase (also known as nsp12) of SARS-CoV-2, the central component of coronaviral replication and transcription machinery. Results The results showed that seven lipopeptides bound to nsp12 at the same location as the FDA-approved drug remdesivir, with higher affinities. Notably, iron-chelating ferrocin A (ferrocin A–iron complex [FAC]) bound to nsp12 most tightly, releasing up to 9.1 kcal mol−1 of free energy. Protein-ligand interaction analysis revealed that FAC formed four hydrogen bonds, two hydrophobic interactions, and three salt bridges with nsp12. These active amino acids are mainly distributed in the fingers and thumb subdomains of nsp12 and are highly conserved. Conclusions Our findings suggest that the abovementioned lipopeptides can tightly bind to nsp12, and thus represent promising drug candidates for anti-coronaviral treatments with the potential to fight SARS-CoV-2.
Collapse
|
14
|
Abstract
![]()
The development of
lipopeptides (lipidated peptides) for vaccines
is discussed, including their role as antigens and/or adjuvants. Distinct
classes of lipopeptide architectures are covered including simple
linear and ligated constructs and lipid core peptides. The design,
synthesis, and immunological responses of the important class of glycerol-based
Toll-like receptor agonist lipopeptides such as Pam3CSK4, which contains three palmitoyl chains and a CSK4 hexapeptide sequence, and many derivatives of this model immunogenic
compound are also reviewed. Self-assembled lipopeptide structures
including spherical and worm-like micelles that have been shown to
act as vaccine agents are also described. The work discussed includes
examples of lipopeptides developed with model antigens, as well as
for immunotherapies to treat many infectious diseases including malaria,
influenza, hepatitis, COVID-19, and many others, as well as cancer
immunotherapies. Some of these have proceeded to clinical development.
The research discussed highlights the huge potential of, and diversity
of roles for, lipopeptides in contemporary and future vaccine development.
Collapse
Affiliation(s)
- Ian W Hamley
- Department of Chemistry, University of Reading, Whiteknights, Reading RG6 6AD, U.K
| |
Collapse
|
15
|
Girkin J, Loo SL, Esneau C, Maltby S, Mercuri F, Chua B, Reid AT, Veerati PC, Grainge CL, Wark PAB, Knight D, Jackson D, Demaison C, Bartlett NW. TLR2-mediated innate immune priming boosts lung anti-viral immunity. Eur Respir J 2021; 58:13993003.01584-2020. [PMID: 33303547 DOI: 10.1183/13993003.01584-2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/27/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND We assessed whether Toll-like receptor (TLR)2 activation boosts the innate immune response to rhinovirus infection, as a treatment strategy for virus-induced respiratory diseases. METHODS We employed treatment with a novel TLR2 agonist (INNA-X) prior to rhinovirus infection in mice, and INNA-X treatment in differentiated human bronchial epithelial cells derived from asthmatic-donors. We assessed viral load, immune cell recruitment, cytokines, type I and III interferon (IFN) production, as well as the lung tissue and epithelial cell immune transcriptome. RESULTS We show, in vivo, that a single INNA-X treatment induced innate immune priming characterised by low-level IFN-λ, Fas ligand, chemokine expression and airway lymphocyte recruitment. Treatment 7 days before infection significantly reduced lung viral load, increased IFN-β/λ expression and inhibited neutrophilic inflammation. Corticosteroid treatment enhanced the anti-inflammatory effects of INNA-X. Treatment 1 day before infection increased expression of 190 lung tissue immune genes. This tissue gene expression signature was absent with INNA-X treatment 7 days before infection, suggesting an alternate mechanism, potentially via establishment of immune cell-mediated mucosal innate immunity. In vitro, INNA-X treatment induced a priming response defined by upregulated IFN-λ, chemokine and anti-microbial gene expression that preceded an accelerated response to infection enriched for nuclear factor (NF)-κB-regulated genes and reduced viral loads, even in epithelial cells derived from asthmatic donors with intrinsic delayed anti-viral immune response. CONCLUSION Airway epithelial cell TLR2 activation induces prolonged innate immune priming, defined by early NF-κB activation, IFN-λ expression and lymphocyte recruitment. This response enhanced anti-viral innate immunity and reduced virus-induced airway inflammation.
Collapse
Affiliation(s)
- Jason Girkin
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,These authors contributed equally
| | - Su-Ling Loo
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,These authors contributed equally
| | - Camille Esneau
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Steven Maltby
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | | | - Brendon Chua
- Dept of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Andrew T Reid
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Punnam Chander Veerati
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Chris L Grainge
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Darryl Knight
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - David Jackson
- Dept of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | | | - Nathan W Bartlett
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia .,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| |
Collapse
|
16
|
Chathuranga K, Weerawardhana A, Dodantenna N, Ranathunga L, Cho WK, Ma JY, Lee JS. Inhibitory Effect of Sargassum fusiforme and Its Components on Replication of Respiratory Syncytial Virus In Vitro and In Vivo. Viruses 2021; 13:548. [PMID: 33806073 PMCID: PMC8064456 DOI: 10.3390/v13040548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/23/2022] Open
Abstract
Sargassum fusiforme, a plant used as a medicine and food, is regarded as a marine vegetable and health supplement to improve life expectancy. Here, we demonstrate that S. fusiforme extract (SFE) has antiviral effects against respiratory syncytial virus (RSV) in vitro and in vivo mouse model. Treatment of HEp2 cells with a non-cytotoxic concentration of SFE significantly reduced RSV replication, RSV-induced cell death, RSV gene transcription, RSV protein synthesis, and syncytium formation. Moreover, oral inoculation of SFE significantly improved RSV clearance from the lungs of BALB/c mice. Interestingly, the phenolic compounds eicosane, docosane, and tetracosane were identified as active components of SFE. Treatment with a non-cytotoxic concentration of these three components elicited similar antiviral effects against RSV infection as SFE in vitro. Together, these results suggest that SFE and its potential components are a promising natural antiviral agent candidate against RSV infection.
Collapse
Affiliation(s)
- Kiramage Chathuranga
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea; (K.C.); (A.W.); (N.D.); (L.R.)
| | - Asela Weerawardhana
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea; (K.C.); (A.W.); (N.D.); (L.R.)
| | - Niranjan Dodantenna
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea; (K.C.); (A.W.); (N.D.); (L.R.)
| | - Lakmal Ranathunga
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea; (K.C.); (A.W.); (N.D.); (L.R.)
| | - Won-Kyung Cho
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Korea; (W.-K.C.); (J.Y.M.)
| | - Jin Yeul Ma
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Korea; (W.-K.C.); (J.Y.M.)
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea; (K.C.); (A.W.); (N.D.); (L.R.)
| |
Collapse
|
17
|
Deliyannis G, Wong CY, McQuilten HA, Bachem A, Clarke M, Jia X, Horrocks K, Zeng W, Girkin J, Scott NE, Londrigan SL, Reading PC, Bartlett NW, Kedzierska K, Brown LE, Mercuri F, Demaison C, Jackson DC, Chua BY. TLR2-mediated activation of innate responses in the upper airways confers antiviral protection of the lungs. JCI Insight 2021; 6:140267. [PMID: 33561017 PMCID: PMC8021123 DOI: 10.1172/jci.insight.140267] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 02/03/2021] [Indexed: 12/21/2022] Open
Abstract
The impact of respiratory virus infections on global health is felt not just during a pandemic, but endemic seasonal infections pose an equal and ongoing risk of severe disease. Moreover, vaccines and antiviral drugs are not always effective or available for many respiratory viruses. We investigated how induction of effective and appropriate antigen-independent innate immunity in the upper airways can prevent the spread of respiratory virus infection to the vulnerable lower airways. Activation of TLR2, when restricted to the nasal turbinates, resulted in prompt induction of innate immune-driven antiviral responses through action of cytokines, chemokines, and cellular activity in the upper but not the lower airways. We have defined how nasal epithelial cells and recruitment of macrophages work in concert and play pivotal roles to limit progression of influenza virus to the lungs and sustain protection for up to 7 days. These results reveal underlying mechanisms of how control of viral infection in the upper airways can occur and support the implementation of strategies that can activate TLR2 in nasal passages to provide rapid protection, especially for at-risk populations, against severe respiratory infection when vaccines and antiviral drugs are not always effective or available.
Collapse
Affiliation(s)
- Georgia Deliyannis
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Chinn Yi Wong
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hayley A. McQuilten
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Annabell Bachem
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Michele Clarke
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Kylie Horrocks
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Weiguang Zeng
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jason Girkin
- Viral Immunology and Respiratory Disease group, School of Biomedical Science and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Nichollas E. Scott
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sarah L. Londrigan
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Patrick C. Reading
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Nathan W. Bartlett
- Viral Immunology and Respiratory Disease group, School of Biomedical Science and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lorena E. Brown
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | | | | | - David C. Jackson
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Brendon Y. Chua
- Department of Microbiology and Immunology, the University of Melbourne, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
18
|
Boiardi F, Stebbing J. Reducing transmission of SARS-CoV-2 with intranasal prophylaxis. EBioMedicine 2021; 63:103170. [PMID: 33340994 PMCID: PMC7750542 DOI: 10.1016/j.ebiom.2020.103170] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 01/13/2023] Open
Affiliation(s)
| | - Justin Stebbing
- Department of Surgery and Cancer, Imperial College, London, UK
| |
Collapse
|
19
|
Proud PC, Tsitoura D, Watson RJ, Chua BY, Aram MJ, Bewley KR, Cavell BE, Cobb R, Dowall S, Fotheringham SA, Ho CMK, Lucas V, Ngabo D, Rayner E, Ryan KA, Slack GS, Thomas S, Wand NI, Yeates P, Demaison C, Zeng W, Holmes I, Jackson DC, Bartlett NW, Mercuri F, Carroll MW. Prophylactic intranasal administration of a TLR2/6 agonist reduces upper respiratory tract viral shedding in a SARS-CoV-2 challenge ferret model. EBioMedicine 2021; 63:103153. [PMID: 33279857 PMCID: PMC7711201 DOI: 10.1016/j.ebiom.2020.103153] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/16/2020] [Accepted: 11/13/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The novel human coronavirus SARS-CoV-2 is a major ongoing global threat with huge economic burden. Like all respiratory viruses, SARS-CoV-2 initiates infection in the upper respiratory tract (URT). Infected individuals are often asymptomatic, yet highly infectious and readily transmit virus. A therapy that restricts initial replication in the URT has the potential to prevent progression of severe lower respiratory tract disease as well as limiting person-to-person transmission. METHODS SARS-CoV-2 Victoria/01/2020 was passaged in Vero/hSLAM cells and virus titre determined by plaque assay. Challenge virus was delivered by intranasal instillation to female ferrets at 5.0 × 106 pfu/ml. Treatment groups received intranasal INNA-051, developed by Ena Respiratory. SARS-CoV-2 RNA was detected using the 2019-nCoV CDC RUO Kit and QuantStudio™ 7 Flex Real-Time PCR System. Histopathological analysis was performed using cut tissues stained with haematoxylin and eosin (H&E). FINDINGS We show that prophylactic intra-nasal administration of the TLR2/6 agonist INNA-051 in a SARS-CoV-2 ferret infection model effectively reduces levels of viral RNA in the nose and throat. After 5 days post-exposure to SARS-CoV-2, INNA-051 significantly reduced virus in throat swabs (p=<0.0001) by up to a 24 fold (96% reduction) and in nasal wash (p=0.0107) up to a 15 fold (93% reduction) in comparison to untreated animals. INTERPRETATION The results of our study support clinical development of a therapy based on prophylactic TLR2/6 innate immune activation in the URT, to reduce SARS-CoV-2 transmission and provide protection against COVID-19. FUNDING This work was funded by Ena Respiratory, Melbourne, Australia.
Collapse
Affiliation(s)
- Pamela C Proud
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Daphne Tsitoura
- Ena Respiratory, Level 9, 31 Queen St, Melbourne, Victoria, 3000, Australia
| | - Robert J Watson
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Brendon Y Chua
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne, Victoria 3000, Australia
| | - Marilyn J Aram
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Kevin R Bewley
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Breeze E Cavell
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Rebecca Cobb
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Stuart Dowall
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Susan A Fotheringham
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Catherine M K Ho
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Vanessa Lucas
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Didier Ngabo
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Emma Rayner
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Kathryn A Ryan
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Gillian S Slack
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Stephen Thomas
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Nadina I Wand
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | - Paul Yeates
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG
| | | | - Weiguang Zeng
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne, Victoria 3000, Australia
| | - Ian Holmes
- Ena Respiratory, Level 9, 31 Queen St, Melbourne, Victoria, 3000, Australia
| | - David C Jackson
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne, Victoria 3000, Australia
| | - Nathan W Bartlett
- Viral Immunology and Respiratory Disease group and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Francesca Mercuri
- Ena Respiratory, Level 9, 31 Queen St, Melbourne, Victoria, 3000, Australia.
| | - Miles W Carroll
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, Wiltshire, United Kingdom SP4 0JG; Nuffield Dept of Medicine, Oxford University, Oxford, UK.
| |
Collapse
|
20
|
Rijsbergen LC, Rennick LJ, Laksono BM, van Run PRWA, Kuiken T, Duprex WP, de Swart RL, de Vries RD. In vivo comparison of a laboratory-adapted and clinical-isolate-based recombinant human respiratory syncytial virus. J Gen Virol 2020; 101:1037-1046. [PMID: 32692644 DOI: 10.1099/jgv.0.001468] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Human respiratory syncytial virus (HRSV) is the leading cause of severe respiratory tract disease in infants. Most HRSV infections remain restricted to the upper respiratory tract (URT), but in a small percentage of patients the infection spreads to the lower respiratory tract, resulting in bronchiolitis or pneumonia. We have a limited understanding of HRSV pathogenesis and what factors determine disease severity, partly due to the widespread use of tissue-culture-adapted viruses. Here, we studied early viral dissemination and tropism of HRSV in cotton rats, BALB/cJ mice and C57BL/6 mice. We used a novel recombinant (r) strain based on a subgroup A clinical isolate (A11) expressing EGFP [rHRSVA11EGFP(5)]. A recombinant laboratory-adapted HRSV strain [rHRSVA2EGFP(5)] was used as a direct comparison. Our results show that rHRSVA11EGFP(5) replicated to higher viral titres than laboratory-adapted rHRSVA2EGFP(5) in the URT of cotton rats and mice. HRSV-infected cells were detected as early as 2 days post-inoculation in both species in the nasal septa and lungs. Infection was predominantly present in ciliated epithelial cells in cotton rats and in the olfactory mucosa of mice. In our opinion, this study highlights that the choice of virus strain is important when studying HRSV pathogenesis in vivo and demonstrates that A11 is a representative clinical-based virus. Additionally, we show critical differences in tropism and inflammation when comparing HRSV infection of cotton rats and mice.
Collapse
Affiliation(s)
- Laurine C Rijsbergen
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Linda J Rennick
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brigitta M Laksono
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Peter R W A van Run
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Thijs Kuiken
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - W Paul Duprex
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rik L de Swart
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Rory D de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
21
|
Brealey JC, Young PR, Sloots TP, Ware RS, Lambert SB, Sly PD, Grimwood K, Chappell KJ. Bacterial colonization dynamics associated with respiratory syncytial virus during early childhood. Pediatr Pulmonol 2020; 55:1237-1245. [PMID: 32176838 DOI: 10.1002/ppul.24715] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/24/2020] [Indexed: 12/21/2022]
Abstract
Respiratory syncytial virus (RSV) is an important cause of early life acute respiratory infections. Potentially pathogenic respiratory bacteria, including Streptococcus pneumoniae, Moraxella catarrhalis, and Haemophilus influenzae are frequently detected during RSV infections and associated with increased illness severity. However, the temporal dynamics of bacterial colonization associated with RSV infection remain unclear. We used weekly nasal swab data from a prospective longitudinal birth cohort in Brisbane, Australia, to investigate bacterial colonization patterns within children aged less than 2 years in the 4-week period before and after an RSV infection. During 54 RSV infection episodes recorded in 47 children, both S. pneumoniae and M. catarrhalis were detected frequently (in 33 [61.1%] and 26 [48.1%] RSV infections, respectively). In most cases, S. pneumoniae and M. catarrhalis colonization preceded the viral infection, with the nasal load of each increasing during RSV infection. Generally, the dominant serotype of S. pneumoniae remained consistent in the 1 to 2 weeks immediately before and after RSV infection. Little evidence was found to indicate that prior colonization with either bacteria predisposed participants to developing RSV infection during the annual seasonal epidemic. Possible coacquisition events, where the bacteria species was first detected with RSV and not in the preceding 4 weeks, were observed in approximately 20% of RSV/S. pneumoniae and RSV/M. catarrhalis codetections. Taken together our results indicate that RSV generally triggered an outgrowth, rather than a new acquisition, of S. pneumoniae and M. catarrhalis from the resident microbial community.
Collapse
Affiliation(s)
- Jaelle C Brealey
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| | - Paul R Young
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland, Australia
| | - Theo P Sloots
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland, Australia.,Queensland Paediatric Infectious Diseases Laboratory, Infection Management and Prevention Service, Children's Health Queensland, South Brisbane, Queensland, Australia
| | - Robert S Ware
- School of Medicine and Infection and Immunology Division, Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - Stephen B Lambert
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland, Australia
| | - Peter D Sly
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland, Australia.,Child Health Research Centre, The University of Queensland, South Brisbane, Queensland, Australia
| | - Keith Grimwood
- School of Medicine and Infection and Immunology Division, Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia.,Departments of Infectious Diseases and Paediatrics, Gold Coast Health, Southport, Queensland, Australia
| | - Keith J Chappell
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
22
|
Lipopeptide PAM3CYS4 Synergizes N-Formyl-Met-Leu-Phe (fMLP)-Induced Calcium Transients in Mouse Neutrophils. Shock 2019; 50:493-499. [PMID: 29176405 DOI: 10.1097/shk.0000000000001062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
N-Formyl-Met-Leu-Phe (fMLP), a mimic of N-formyl oligopeptides that are released from bacteria, is a potent leukocyte chemotactic factor. It induces intracellular calcium ([Ca]i) transient that is important for various neutrophil biological functions, e.g., adhesion, ROS, and cytokine productions. Toll-like receptors (TLRs), an essential part of host innate immunity, regulate neutrophil activities, but their role in [Ca]i signaling is less clear. In the present study, we examined the effect of several TLR ligands, including Pam3Cys4 (TLR1/2), lipopolysaccharide (LPS, TLR4), and lipoteichoic acid (LTA, TLR2/6), on calcium signaling and on the fMLP-induced [Ca]i transients in mouse neutrophils loaded with Fura-2/AM. We found that unlike fMLP, the three TLR ligands tested did not elicit any detectable Ca flux. However, Pam3Cys4, but not LPS or LTA, markedly synergized the fMLP-induced [Ca]i transients, and had no effect on the host component keratinocyte-derived cytokine (KC)- or C5a-induced calcium flux. The effect of Pam3Cys4 on the fMLP-induced [Ca]i transients is by enhancing extracellular Ca influx, not intracellular Ca release. Surprisingly, deletion of TLR2 or MyD88 in neutrophils had no impact on the Pam3Cys4's effect, suggesting a TLR2-MyD88-independent mechanism. Finally, using the pan PKC activator and inhibitor, we demonstrated that PKC negatively regulated fMLP-induced [Ca]i transients and that inhibition of PKC did not prohibit Pam3Cys4's synergistic effect on the fMLP-induced calcium influx. In conclusion, the present study identified a novel synergistic effect of Pam3Cys4 on fMLP-induced [Ca]i transients, a process important for many neutrophil biological functions.
Collapse
|
23
|
Anti-Respiratory Syncytial Virus Activity of Plantago asiatica and Clerodendrum trichotomum Extracts In Vitro and In Vivo. Viruses 2019; 11:v11070604. [PMID: 31277257 PMCID: PMC6669655 DOI: 10.3390/v11070604] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/01/2019] [Accepted: 07/01/2019] [Indexed: 12/12/2022] Open
Abstract
The herbs Plantago asiatica and Clerodendrum trichotomum have been commonly used for centuries in indigenous and folk medicine in tropical and subtropical regions of the world. In this study, we show that extracts from these herbs have antiviral effects against the respiratory syncytial virus (RSV) in vitro cell cultures and an in vivo mouse model. Treatment of HEp2 cells and A549 cells with a non-cytotoxic concentration of Plantago asiatica or Clerodendrum trichotomum extract significantly reduced RSV replication, RSV-induced cell death, RSV gene transcription, RSV protein synthesis, and also blocked syncytia formation. Interestingly, oral inoculation with each herb extract significantly improved viral clearance in the lungs of BALB/c mice. Based on reported information and a high-performance liquid chromatography (HPLC) analysis, the phenolic glycoside acteoside was identified as an active chemical component of both herb extracts. An effective dose of acteoside exhibited similar antiviral effects as each herb extract against RSV in vitro and in vivo. Collectively, these results suggest that extracts of Plantago asiatica and Clerodendrum trichotomum could provide a potent natural source of an antiviral drug candidate against RSV infection.
Collapse
|
24
|
Benedikz EK, Bailey D, Cook CNL, Gonçalves-Carneiro D, Buckner MMC, Blair JMA, Wells TJ, Fletcher NF, Goodall M, Flores-Langarica A, Kingsley RA, Madsen J, Teeling J, Johnston SL, MacLennan CA, Balfe P, Henderson IR, Piddock LJV, Cunningham AF, McKeating JA. Bacterial flagellin promotes viral entry via an NF-kB and Toll Like Receptor 5 dependent pathway. Sci Rep 2019; 9:7903. [PMID: 31133714 PMCID: PMC6536546 DOI: 10.1038/s41598-019-44263-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022] Open
Abstract
Viruses and bacteria colonize hosts by invading epithelial barriers. Recent studies have shown that interactions between the microbiota, pathogens and the host can potentiate infection through poorly understood mechanisms. Here, we investigated whether diverse bacterial species could modulate virus internalization into host cells, often a rate-limiting step in establishing infections. Lentiviral pseudoviruses expressing influenza, measles, Ebola, Lassa or vesicular stomatitis virus envelope glycoproteins enabled us to study entry of viruses that exploit diverse internalization pathways. Salmonella Typhimurium, Escherichia coli and Pseudomonas aeruginosa significantly increased viral uptake, even at low bacterial frequencies. This did not require bacterial contact with or invasion of host cells. Studies determined that the bacterial antigen responsible for this pro-viral activity was the Toll-Like Receptor 5 (TLR5) agonist flagellin. Exposure to flagellin increased virus attachment to epithelial cells in a temperature-dependent manner via TLR5-dependent activation of NF-ΚB. Importantly, this phenotype was both long lasting and detectable at low multiplicities of infection. Flagellin is shed from bacteria and our studies uncover a new bystander role for this protein in regulating virus entry. This highlights a new aspect of viral-bacterial interplay with significant implications for our understanding of polymicrobial-associated pathogenesis.
Collapse
Affiliation(s)
- Elizabeth K Benedikz
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.,Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Dalan Bailey
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.,The Pirbright Institute, Guildford, Surrey, UK
| | - Charlotte N L Cook
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | | | - Michelle M C Buckner
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Jessica M A Blair
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Timothy J Wells
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Nicola F Fletcher
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Margaret Goodall
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | | | | - Jens Madsen
- Department of Child Health, University of Southampton, Southampton, UK
| | - Jessica Teeling
- Biological Sciences, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Calman A MacLennan
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Peter Balfe
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Ian R Henderson
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Laura J V Piddock
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Adam F Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.,Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Jane A McKeating
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK. .,Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
25
|
Effectiveness of the ten-valent pneumococcal Haemophilus influenzae protein D conjugate vaccine (PHiD-CV10) against all respiratory tract infections in children under two years of age. Vaccine 2019; 37:2935-2941. [DOI: 10.1016/j.vaccine.2019.04.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/03/2019] [Accepted: 04/12/2019] [Indexed: 01/17/2023]
|
26
|
Abstract
The Lpp lipoprotein of Escherichia coli is the first identified protein with a covalently linked lipid. It is chemically bound by its C-terminus to murein (peptidoglycan) and inserts by the lipid at the N-terminus into the outer membrane. As the most abundant protein in E. coli (106 molecules per cell) it plays an important role for the integrity of the cell envelope. Lpp represents the type protein of a large variety of lipoproteins found in Gram-negative and Gram-positive bacteria and in archaea that have in common the lipid structure for anchoring the proteins to membranes but otherwise strongly vary in sequence, structure, and function. Predicted lipoproteins in known prokaryotic genomes comprise 2.7% of all proteins. Lipoproteins are modified by a unique phospholipid pathway and transferred from the cytoplasmic membrane into the outer membrane by a special system. They are involved in protein incorporation into the outer membrane, protein secretion across the cytoplasmic membrane, periplasm and outer membrane, signal transduction, conjugation, cell wall metabolism, antibiotic resistance, biofilm formation, and adhesion to host tissues. They are only found in bacteria and function as signal molecules for the innate immune system of vertebrates, where they cause inflammation and elicit innate and adaptive immune response through Toll-like receptors. This review discusses various aspects of Lpp and other lipoproteins of Gram-negative and Gram-positive bacteria and archaea.
Collapse
Affiliation(s)
- Volkmar Braun
- Department of Protein Evolution, Max Planck Institute for Developmental Biology, Max Planck Ring 5, 72076, Tübingen, Germany.
| | - Klaus Hantke
- IMIT, University of Tuebingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| |
Collapse
|
27
|
Laksono BM, Grosserichter-Wagener C, de Vries RD, Langeveld SAG, Brem MD, van Dongen JJM, Katsikis PD, Koopmans MPG, van Zelm MC, de Swart RL. In Vitro Measles Virus Infection of Human Lymphocyte Subsets Demonstrates High Susceptibility and Permissiveness of both Naive and Memory B Cells. J Virol 2018; 92:e00131-18. [PMID: 29437964 PMCID: PMC5874404 DOI: 10.1128/jvi.00131-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 01/28/2018] [Indexed: 11/29/2022] Open
Abstract
Measles is characterized by a transient immune suppression, leading to an increased risk of opportunistic infections. Measles virus (MV) infection of immune cells is mediated by the cellular receptor CD150, expressed by subsets of lymphocytes, dendritic cells, macrophages, and thymocytes. Previous studies showed that human and nonhuman primate memory T cells express higher levels of CD150 than naive cells and are more susceptible to MV infection. However, limited information is available about the CD150 expression and relative susceptibility to MV infection of B-cell subsets. In this study, we assessed the susceptibility and permissiveness of naive and memory T- and B-cell subsets from human peripheral blood or tonsils to in vitro MV infection. Our study demonstrates that naive and memory B cells express CD150, but at lower frequencies than memory T cells. Nevertheless, both naive and memory B cells proved to be highly permissive to MV infection. Furthermore, we assessed the susceptibility and permissiveness of various functionally distinct T and B cells, such as helper T (TH) cell subsets and IgG- and IgA-positive memory B cells, in peripheral blood and tonsils. We demonstrated that TH1TH17 cells and plasma and germinal center B cells were the subsets most susceptible and permissive to MV infection. Our study suggests that both naive and memory B cells, along with several other antigen-experienced lymphocytes, are important target cells of MV infection. Depletion of these cells potentially contributes to the pathogenesis of measles immune suppression.IMPORTANCE Measles is associated with immune suppression and is often complicated by bacterial pneumonia, otitis media, or gastroenteritis. Measles virus infects antigen-presenting cells and T and B cells, and depletion of these cells may contribute to lymphopenia and immune suppression. Measles has been associated with follicular exhaustion in lymphoid tissues in humans and nonhuman primates, emphasizing the importance of MV infection of B cells in vivo However, information on the relative susceptibility of B-cell subsets is scarce. Here, we compared the susceptibility and permissiveness to in vitro MV infection of human naive and memory T- and B-cell subsets isolated from peripheral blood or tonsils. Our results demonstrate that both naive and memory B cells are more permissive to MV infection than T cells. The highest infection levels were detected in plasma cells and germinal center B cells, suggesting that infection and depletion of these populations contribute to reduced host resistance.
Collapse
Affiliation(s)
- Brigitta M Laksono
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Christina Grosserichter-Wagener
- Department of Immunology, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Rory D de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Simone A G Langeveld
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Maarten D Brem
- Department of Immunology, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Jacques J M van Dongen
- Department of Immunology, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Peter D Katsikis
- Department of Immunology, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Menno C van Zelm
- Department of Immunology, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - Rik L de Swart
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
28
|
Anti-inflammatory effect of glucose-lysine Maillard reaction products on intestinal inflammation model in vivo. Int Immunopharmacol 2017; 52:324-332. [PMID: 28987931 DOI: 10.1016/j.intimp.2017.09.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/12/2017] [Accepted: 09/12/2017] [Indexed: 12/15/2022]
Abstract
Inflammatory bowel diseases (IBDs) are chronic disorders that are characterized by intestinal epithelial inflammation and injury. Currently, the most employed therapies are antibiotics and anti-inflammatory drugs; however, the side effects limit long-term effectiveness. We evaluated the impact of glucose-lysine Maillard reaction products (Glc-Lys MRPs) on colitis, induced in rats by an administration of 5% dextran sulfate sodium (DSS) in drinking water. Glc-Lys MRPs ameliorate DSS-induced colitis, as determined by a decrease in disease index activity, colon weight/length ratio, nitric oxide levels in serum, recovery of body weight loss, colon length and serum lysozyme levels. Furthermore, Glc-Lys MRPs increase the glutathione content and the activity of glutathione peroxidase, superoxide dismutase and catalase, and inhibit lipid peroxidation and myeloperoxidase activity in colon tissues. In particular, Glc-Lys MRPs suppress the mRNA level of the inflammatory cytokines and nuclear factor-κB in colon tissues. This study suggests the potential of Glc-Lys MRPs in preventing or treating IBDs.
Collapse
|
29
|
Hertoghs N, Pul LV, Geijtenbeek TBH. Mucosal dendritic cells in HIV-1 susceptibility: a critical role for C-type lectin receptors. Future Virol 2017. [DOI: 10.2217/fvl-2017-0020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Sexual transmission is the major route of HIV-1 infection worldwide. The interaction of HIV-1 with mucosal dendritic cells (DCs) might determine HIV-1 susceptibility as well as initial antiviral immunity controlling virus in the chronic phase. Different DC subsets reside in mucosal tissues and express specific C-type lectin receptors (CLRs) that interact with HIV-1 with different outcomes. HIV-1 has been shown to subvert CLRs for viral transmission and immune evasion, whereas CLRs can also protect against HIV-1 infection. Here, we will discuss the role of CLRs in HIV-1 transmission and adaptive immunity, and how the CLRs dictate the function of DCs in infection. Ultimately, understanding the interplay between CLRs and HIV-1 will lead to targeted approaches in the search for preventative measures.
Collapse
Affiliation(s)
- Nina Hertoghs
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, 1105 AZ, Amsterdam, The Netherlands
| | - Lisa van Pul
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, 1105 AZ, Amsterdam, The Netherlands
| | - Teunis BH Geijtenbeek
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Lee BH, Chathuranga K, Uddin MB, Weeratunga P, Kim MS, Cho WK, Kim HI, Ma JY, Lee JS. Coptidis Rhizoma extract inhibits replication of respiratory syncytial virus in vitro and in vivo by inducing antiviral state. J Microbiol 2017; 55:488-498. [DOI: 10.1007/s12275-017-7088-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 03/15/2017] [Indexed: 02/05/2023]
|
31
|
de Vries RD, Ludlow M, de Jong A, Rennick LJ, Verburgh RJ, van Amerongen G, van Riel D, van Run PRWA, Herfst S, Kuiken T, Fouchier RAM, Osterhaus ADME, de Swart RL, Duprex WP. Delineating morbillivirus entry, dissemination and airborne transmission by studying in vivo competition of multicolor canine distemper viruses in ferrets. PLoS Pathog 2017; 13:e1006371. [PMID: 28481926 PMCID: PMC5436898 DOI: 10.1371/journal.ppat.1006371] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/18/2017] [Accepted: 04/23/2017] [Indexed: 12/19/2022] Open
Abstract
Identification of cellular receptors and characterization of viral tropism in animal models have vastly improved our understanding of morbillivirus pathogenesis. However, specific aspects of viral entry, dissemination and transmission remain difficult to recapitulate in animal models. Here, we used three virologically identical but phenotypically distinct recombinant (r) canine distemper viruses (CDV) expressing different fluorescent reporter proteins for in vivo competition and airborne transmission studies in ferrets (Mustela putorius furo). Six donor ferrets simultaneously received three rCDVs expressing green, red or blue fluorescent proteins via conjunctival (ocular, Oc), intra-nasal (IN) or intra-tracheal (IT) inoculation. Two days post-inoculation sentinel ferrets were placed in physically separated adjacent cages to assess airborne transmission. All donor ferrets developed lymphopenia, fever and lethargy, showed progressively increasing systemic viral loads and were euthanized 14 to 16 days post-inoculation. Systemic replication of virus inoculated via the Oc, IN and IT routes was detected in 2/6, 5/6 and 6/6 ferrets, respectively. In five donor ferrets the IT delivered virus dominated, although replication of two or three different viruses was detected in 5/6 animals. Single lymphocytes expressing multiple fluorescent proteins were abundant in peripheral blood and lymphoid tissues, demonstrating the occurrence of double and triple virus infections. Transmission occurred efficiently and all recipient ferrets showed evidence of infection between 18 and 22 days post-inoculation of the donor ferrets. In all cases, airborne transmission resulted in replication of a single-colored virus, which was the dominant virus in the donor ferret. This study demonstrates that morbilliviruses can use multiple entry routes in parallel, and co-infection of cells during viral dissemination in the host is common. Airborne transmission was efficient, although transmission of viruses expressing a single color suggested a bottleneck event. The identity of the transmitted virus was not determined by the site of inoculation but by the viral dominance during dissemination.
Collapse
Affiliation(s)
- Rory D. de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Martin Ludlow
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Alwin de Jong
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Linda J. Rennick
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - R. Joyce Verburgh
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Geert van Amerongen
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Debby van Riel
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Peter R. W. A. van Run
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Sander Herfst
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Thijs Kuiken
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Ron A. M. Fouchier
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Albert D. M. E. Osterhaus
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Rik L. de Swart
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - W. Paul Duprex
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
32
|
Murphy KC, Whitehead J, Falahee PC, Zhou D, Simon SI, Leach JK. Multifactorial Experimental Design to Optimize the Anti-Inflammatory and Proangiogenic Potential of Mesenchymal Stem Cell Spheroids. Stem Cells 2017; 35:1493-1504. [PMID: 28276602 DOI: 10.1002/stem.2606] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/13/2017] [Accepted: 02/19/2017] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cell therapies promote wound healing by manipulating the local environment to enhance the function of host cells. Aggregation of mesenchymal stem cells (MSCs) into three-dimensional spheroids increases cell survival and augments their anti-inflammatory and proangiogenic potential, yet there is no consensus on the preferred conditions for maximizing spheroid function in this application. The objective of this study was to optimize conditions for forming MSC spheroids that simultaneously enhance their anti-inflammatory and proangiogenic nature. We applied a design of experiments (DOE) approach to determine the interaction between three input variables (number of cells per spheroid, oxygen tension, and inflammatory stimulus) on MSC spheroids by quantifying secretion of prostaglandin E2 (PGE2 ) and vascular endothelial growth factor (VEGF), two potent molecules in the MSC secretome. DOE results revealed that MSC spheroids formed with 40,000 cells per spheroid in 1% oxygen with an inflammatory stimulus (Spheroid 1) would exhibit enhanced PGE2 and VEGF production versus those formed with 10,000 cells per spheroid in 21% oxygen with no inflammatory stimulus (Spheroid 2). Compared to Spheroid 2, Spheroid 1 produced fivefold more PGE2 and fourfold more VEGF, providing the opportunity to simultaneously upregulate the secretion of these factors from the same spheroid. The spheroids induced macrophage polarization, sprout formation with endothelial cells, and keratinocyte migration in a human skin equivalent model-demonstrating efficacy on three key cell types that are dysfunctional in chronic non-healing wounds. We conclude that DOE-based analysis effectively identifies optimal culture conditions to enhance the anti-inflammatory and proangiogenic potential of MSC spheroids. Stem Cells 2017;35:1493-1504.
Collapse
Affiliation(s)
- Kaitlin C Murphy
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Jacklyn Whitehead
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Patrick C Falahee
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Dejie Zhou
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Scott I Simon
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - J Kent Leach
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA.,Department of Orthopaedic Surgery, School of Medicine, University of California Davis, Sacramento, California, USA
| |
Collapse
|
33
|
Annamalay A, Le Souëf P. Viral-Bacterial Interactions in Childhood Respiratory Tract Infections. VIRAL INFECTIONS IN CHILDREN, VOLUME I 2017. [PMCID: PMC7122469 DOI: 10.1007/978-3-319-54033-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
34
|
Vissers M, Ahout IM, van den Kieboom CH, van der Gaast-de Jongh CE, Groh L, Cremers AJ, de Groot R, de Jonge MI, Ferwerda G. High pneumococcal density correlates with more mucosal inflammation and reduced respiratory syncytial virus disease severity in infants. BMC Infect Dis 2016; 16:129. [PMID: 26983753 PMCID: PMC4794819 DOI: 10.1186/s12879-016-1454-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/04/2016] [Indexed: 11/10/2022] Open
Abstract
Background Respiratory syncytial virus (RSV) is an important cause of lower respiratory tract infections in infants. A small percentage of the infected infants develops a severe infection, while most of these severely ill patients were previously healthy. It remains unclear why these children develop severe RSV infections. In this study, we investigate whether pneumococcal nasopharyngeal carriage patterns correlate with mucosal inflammation and severity of disease. Methods In total, 105 infants hospitalized with RSV infection were included and recovery samples were taken from 42 patients. The presence and density of Streptococcus pneumoniae was determined by RT qPCR to study its relation to viral load, inflammation (MMP-9 and IL-6) and severity of RSV disease. Results We show that pneumococcal presence or absence in the nasopharynx does not correlate with viral load, inflammation or severity of disease. However, when pneumococcus is present in patients, a higher nasopharyngeal pneumococcal density was correlated with a higher RSV load, higher MMP-9 levels and a less severe course of disease. Conclusions Our results show correlations between S. pneumoniae density and viral load, inflammation and disease severity, suggesting that pneumococcal density may be an indicator for severity in paediatric RSV disease. Electronic supplementary material The online version of this article (doi:10.1186/s12879-016-1454-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marloes Vissers
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Inge M Ahout
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Corné H van den Kieboom
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Christa E van der Gaast-de Jongh
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Laszlo Groh
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Amelieke J Cremers
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Ronald de Groot
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Marien I de Jonge
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Gerben Ferwerda
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
35
|
Streptococcus pneumoniae Enhances Human Respiratory Syncytial Virus Infection In Vitro and In Vivo. PLoS One 2015; 10:e0127098. [PMID: 25970287 PMCID: PMC4430531 DOI: 10.1371/journal.pone.0127098] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/11/2015] [Indexed: 12/29/2022] Open
Abstract
Human respiratory syncytial virus (HRSV) and Streptococcus pneumoniae are important causative agents of respiratory tract infections. Both pathogens are associated with seasonal disease outbreaks in the pediatric population, and can often be detected simultaneously in infants hospitalized with bronchiolitis or pneumonia. It has been described that respiratory virus infections may predispose for bacterial superinfections, resulting in severe disease. However, studies on the influence of bacterial colonization of the upper respiratory tract on the pathogenesis of subsequent respiratory virus infections are scarce. Here, we have investigated whether pneumococcal colonization enhances subsequent HRSV infection. We used a newly generated recombinant subgroup B HRSV strain that expresses enhanced green fluorescent protein and pneumococcal isolates obtained from healthy children in disease-relevant in vitro and in vivo model systems. Three pneumococcal strains specifically enhanced in vitro HRSV infection of primary well-differentiated normal human bronchial epithelial cells grown at air-liquid interface, whereas two other strains did not. Since previous studies reported that bacterial neuraminidase enhanced HRSV infection in vitro, we measured pneumococcal neuraminidase activity in these cultures but found no correlation with the observed infection enhancement in our model. Subsequently, a selection of pneumococcal strains was used to induce nasal colonization of cotton rats, the best available small animal model for HRSV. Intranasal HRSV infection three days later resulted in strain-specific enhancement of HRSV replication in vivo. One S. pneumoniae strain enhanced HRSV both in vitro and in vivo, and was also associated with enhanced syncytium formation in vivo. However, neither pneumococci nor HRSV were found to spread from the upper to the lower respiratory tract, and neither pathogen was transmitted to naive cage mates by direct contact. These results demonstrate that pneumococcal colonization can enhance subsequent HRSV infection, and provide tools for additional mechanistic and intervention studies.
Collapse
|
36
|
Brealey JC, Sly PD, Young PR, Chappell KJ. Viral bacterial co-infection of the respiratory tract during early childhood. FEMS Microbiol Lett 2015; 362:fnv062. [PMID: 25877546 DOI: 10.1093/femsle/fnv062] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2015] [Indexed: 12/21/2022] Open
Abstract
Acute respiratory infection (ARI) is an important cause of morbidity in children. Mixed aetiology is frequent, with pathogenic viruses and bacteria co-detected in respiratory secretions. However, the clinical significance of these viral/bacterial co-infections has long been a controversial topic. While severe bacterial pneumonia following influenza infection has been well described, associations are less clear among infections caused by viruses that are more common in young children, such as respiratory syncytial virus. Although assessing the overall contribution of bacteria to disease severity is complicated by the presence of many confounding factors in clinical studies, understanding the role of viral/bacterial co-infections in defining the outcome of paediatric ARI will potentially reveal novel treatment and prevention strategies, improving patient outcomes. This review summarizes current evidence for the clinical significance of respiratory viral/bacterial co-infections in young children, discusses possible mechanisms of cooperative interaction between these pathogens and highlights areas that require further investigation.
Collapse
Affiliation(s)
- Jaelle C Brealey
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Peter D Sly
- Queensland Children's Medical Research Institute, The University of Queensland, Brisbane, QLD 4006, Australia Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Paul R Young
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD 4072, Australia Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Keith J Chappell
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
37
|
Bovine immunoglobulin/protein isolate binds pro-inflammatory bacterial compounds and prevents immune activation in an intestinal co-culture model. PLoS One 2015; 10:e0120278. [PMID: 25830826 PMCID: PMC4382133 DOI: 10.1371/journal.pone.0120278] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 01/28/2015] [Indexed: 12/15/2022] Open
Abstract
Intestinal barrier dysfunction is associated with chronic gastrointestinal tract inflammation and diseases such as IBD and IBS. Serum-derived bovine immunoglobulin/protein isolate (SBI) is a specially formulated protein preparation (>90%) for oral administration. The composition of SBI is greater than 60% immunoglobulin including contributions from IgG, IgA, and IgM. Immunoglobulin within the lumen of the gut has been recognized to have anti-inflammatory properties and is involved in maintaining gut homeostasis. The binding of common intestinal antigens (LPS and Lipid A) and the ligand Pam3CSK4, by IgG, IgA, and IgM in SBI was shown using a modified ELISA technique. Each of these antigens stimulated IL-8 and TNF-α cytokine production by THP-1 monocytes. Immune exclusion occurred as SBI (≤50 mg/mL) bound free antigen in a dose dependent manner that inhibited cytokine production by THP-1 monocytes in response to 10 ng/mL LPS or 200 ng/mL Lipid A. Conversely, Pam3CSK4 stimulation of THP-1 monocytes was unaffected by SBI/antigen binding. A co-culture model of the intestinal epithelium consisted of a C2BBe1 monolayer separating an apical compartment from a basal compartment containing THP-1 monocytes. The C2BBe1 monolayer was permeabilized with dimethyl palmitoyl ammonio propanesulfonate (PPS) to simulate a damaged epithelial barrier. Results indicate that Pam3CSK4 was able to translocate across the PPS-damaged C2BBe1 monolayer. However, binding of Pam3CSK4 by immunoglobulins in SBI prevented Pam3CSK4 translocation across the damaged C2BBe1 barrier. These results demonstrated steric exclusion of antigen by SBI which prevented apical to basal translocation of antigen due to changes in the physical properties of Pam3CSK4, most likely as a result of immunoglobulin binding. This study demonstrates that immunoglobulins in SBI can reduce antigen-associated inflammation through immune and steric exclusion mechanisms and furthers the mechanistic understanding of how SBI might improve immune status and reduce inflammation in various intestinal disease states.
Collapse
|
38
|
Li G, Wu J, Qin X, Zhu J, Viswanathan K, Dong H, Somasundaran P, Gross RA. Chemo-enzymatic routes to lipopeptides and their colloidal properties. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:6889-6896. [PMID: 24856298 DOI: 10.1021/la500449d] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
A unique chemo-enzymatic route to lipopeptides was demonstrated herein that, relative to alternative methods such as solid-phase peptide synthesis (SPPS) and microbial synthesis, is simple, efficient, and scalable. Homo- and co-oligopeptides were synthesized from amino acid ethyl esters via protease catalysis in an aqueous media, followed by chemical coupling to fatty acids to generate a library of lipopeptides. Synthesized lipopeptides were built from hydrophobic moieties with chain lengths ranging from 8 to 18 and peptides consisting of oligo(L-Glu) or oligo(L-Glu-co-L-Leu) with an average of seven to eight repeating units. The chemical structures of the lipopeptides were characterized and confirmed by NMR and matrix-assisted laser desorption/ionization (MALDI). The colloidal and interfacial properties of these lipopeptides were characterized and compared in terms of the hydrophobic chain length, oligopeptide composition, and solution pH. The results showed correlation between the interfacial activity of the lipopeptides and the hydrophobicity of the fatty acid and oligopeptide headgroup, the effects of which have been semiquantitatively described in the manuscript. Results from these studies provide insights into design principles that can be further expanded in future work to access lipopeptides from protease-catalysis with improved control over sequence and exploring a wider range of peptide and lipid compositions to further tune lipopeptide biochemical and physical properties.
Collapse
Affiliation(s)
- Geng Li
- Department of Chemistry and Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI) , 4005B BioTechnology Building, 110 Eighth Street, Troy, New York 12180, United States
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Shen KY, Song YC, Chen IH, Leng CH, Chen HW, Li HJ, Chong P, Liu SJ. Molecular mechanisms of TLR2-mediated antigen cross-presentation in dendritic cells. THE JOURNAL OF IMMUNOLOGY 2014; 192:4233-41. [PMID: 24683188 DOI: 10.4049/jimmunol.1302850] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cross-presentation is a key function of dendritic cells (DCs), which present exogenous Ags on MHC class I molecules to prime CTL responses. The effects of TLR triggering on the cross-presentation of exogenous Ags by DCs remain unclear. In this study, we used synthetic dipalmitoylated peptides and TLR2 agonist-conjugated peptides as models to elucidate the mechanisms of TLR2-mediated cross-presentation. We observed that the internalization of dipalmitoylated peptides by bone marrow-derived DCs was facilitated by TLR2 via clathrin-mediated endocytosis. The administration of these dipalmitoylated peptide-pulsed bone marrow-derived DCs eliminated established tumors through TLR2 signaling. We further demonstrated that the induction of Ag-specific CTL responses and tumor regression by dipalmitoylated peptides was TAP independent. In addition, presentation of dipalmitoylated peptides by MHC class I molecules was blocked in the presence of an endosomal acidification inhibitor (chloroquine) or a lysosomal degradation inhibitor (Z-FL-COCHO). The endocytosed dipalmitoylated peptide also passed rapidly from early endosome Ag-1-positive endosomes to RAS-related GTP-binding protein 7 (Rab7)-associated late endosomes compared with their nonlipidated counterparts. Furthermore, we found that dipalmitoylated peptide-upregulated Rab7 expression correlated with Ag presentation via the TLR2/MyD88 pathway. Both JNK and ERK signaling pathways are required for upregulation of Rab7. In summary, our data suggest that TLR2-mediated cross-presentation occurs through the upregulation of Rab7 and a TAP-independent pathway that prime CTL responses.
Collapse
Affiliation(s)
- Kuan-Yin Shen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Vissers M, de Groot R, Ferwerda G. Severe viral respiratory infections: are bugs bugging? Mucosal Immunol 2014; 7:227-38. [PMID: 24220300 DOI: 10.1038/mi.2013.93] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 10/09/2013] [Indexed: 02/07/2023]
Abstract
Viral respiratory tract infections (RTI) pose a high burden on the youngest members of our society. Several risk factors are known for severe viral respiratory disease. However, a large proportion of the severe RTI cannot be explained by these risk factors. A growing body of evidence shows that the composition of the microbiota has a major influence on the training of both the mucosal and the systemic immune response and can thus potentially determine susceptibility for severe viral infections. In this review, we discuss the current evidence regarding the influence of bacterial colonization on the severity of viral respiratory infections.
Collapse
Affiliation(s)
- M Vissers
- 1] Department of Pediatrics, Laboratory of Pediatric Infectious Diseases, Radboud university medical center, Nijmegen, The Netherlands [2] Nijmegen Institute for Infection, Inflammation and Immunity, Radboud university medical center, Nijmegen, The Netherlands
| | - R de Groot
- 1] Department of Pediatrics, Laboratory of Pediatric Infectious Diseases, Radboud university medical center, Nijmegen, The Netherlands [2] Nijmegen Institute for Infection, Inflammation and Immunity, Radboud university medical center, Nijmegen, The Netherlands
| | - G Ferwerda
- 1] Department of Pediatrics, Laboratory of Pediatric Infectious Diseases, Radboud university medical center, Nijmegen, The Netherlands [2] Nijmegen Institute for Infection, Inflammation and Immunity, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
41
|
Li J, Lee DSW, Madrenas J. Evolving Bacterial Envelopes and Plasticity of TLR2-Dependent Responses: Basic Research and Translational Opportunities. Front Immunol 2013; 4:347. [PMID: 24191155 PMCID: PMC3808894 DOI: 10.3389/fimmu.2013.00347] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 10/08/2013] [Indexed: 12/17/2022] Open
Abstract
Innate immune mechanisms that follow early recognition of microbes influence the nature and magnitude of subsequent adaptive immune responses. Early detection of microbes depends on pattern recognition receptors that sense pathogen-associated molecular patterns or microbial-associated molecular patterns (PAMPS or MAMPs, respectively). The bacterial envelope contains MAMPs that include membrane proteins, lipopeptides, glycopolymers, and other pro-inflammatory molecules. Bacteria are selected by environmental pressures resulting in quantitative or qualitative changes in their envelope structures that often promote evasion of host immune responses and therefore, infection. However, recent studies have shown that slight, adaptive changes in MAMPs on the bacterial cell wall may result in their ability to induce the secretion not only of pro-inflammatory cytokines but also of anti-inflammatory cytokines. This effect can fine-tune the subsequent response to microbes expressing these MAMPs and lead to the establishment of a commensal state within the host rather than infectious disease. In this review, we will examine the plasticity of Toll-like receptor (TLR) 2 signaling as evidence of evolving MAMPs, using the better-characterized TLR4 as a template. We will review the role of differential dimerization of TLR2 and the arrangement of signaling complexes and co-receptors in determining the capacity of the host to recognize an array of TLR2 ligands and generate different immune responses to these ligands. Last, we will assess briefly how this plasticity may expand the array of interactions between microbes and immune systems beyond the traditional disease-causing paradigm.
Collapse
Affiliation(s)
- Junbin Li
- Microbiome and Disease Tolerance Centre, Department of Microbiology and Immunology, McGill University , Montreal, QC , Canada
| | | | | |
Collapse
|
42
|
Tien Nguyen D, Boes J, van Amerongen G, van Remmerden Y, Yüksel S, Guichelaar T, Osterhaus ADME, de Swart RL. Infection-enhancing lipopeptides do not improve intranasal immunization of cotton rats with a delta-G candidate live-attenuated human respiratory syncytial virus vaccine. Hum Vaccin Immunother 2013; 9:2578-83. [PMID: 23955280 DOI: 10.4161/hv.26096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Development of live-attenuated human respiratory syncytial virus (HRSV) vaccines has proven to be difficult. Several vaccine candidates were found to be over-attenuated and displayed limited immunogenicity. Recently, we identified three synthetic cationic lipopeptides that enhanced paramyxovirus infections in vitro. The infection enhancement proved to be mediated by enhanced virus binding to target cells. We hypothesized that these lipopeptides can be used as adjuvants to promote immune responses induced by live-attenuated paramyxovirus vaccines. This hypothesis was tested in a vaccination and challenge model in cotton rats, using a previously described recombinant live-attenuated candidate HRSV vaccine lacking the gene encoding the G glycoprotein (rHRSVΔG). Surprisingly, intranasal vaccination of cotton rats with rHRSVΔG formulated in infection-enhancing lipopeptides resulted in reduced virus loads in nasopharyngeal lavages, reduced seroconversion levels and reduced protection from wild-type HRSV challenge. In conclusion, we were unable to demonstrate the feasibility of lipopeptides as adjuvants for a candidate live-attenuated HRSV vaccine in the cotton rat model.
Collapse
Affiliation(s)
- D Tien Nguyen
- Department of Viroscience; Erasmus MC; Rotterdam, The Netherlands
| | - Jolande Boes
- National Institute of Public Health and the Environment; Bilthoven, The Netherlands
| | - Geert van Amerongen
- Department of Viroscience; Erasmus MC; Rotterdam, The Netherlands; National Institute of Public Health and the Environment; Bilthoven, The Netherlands
| | - Yvonne van Remmerden
- National Institute of Public Health and the Environment; Bilthoven, The Netherlands
| | - Selma Yüksel
- Department of Viroscience; Erasmus MC; Rotterdam, The Netherlands
| | - Teun Guichelaar
- National Institute of Public Health and the Environment; Bilthoven, The Netherlands
| | | | - Rik L de Swart
- Department of Viroscience; Erasmus MC; Rotterdam, The Netherlands
| |
Collapse
|
43
|
Shafique M, Meijerhof T, Wilschut J, de Haan A. Evaluation of an intranasal virosomal vaccine against respiratory syncytial virus in mice: effect of TLR2 and NOD2 ligands on induction of systemic and mucosal immune responses. PLoS One 2013; 8:e61287. [PMID: 23593453 PMCID: PMC3620164 DOI: 10.1371/journal.pone.0061287] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 03/06/2013] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION RSV infection remains a serious threat to newborns and the elderly. Currently, there is no vaccine available to prevent RSV infection. A mucosal RSV vaccine would be attractive as it could induce mucosal as well as systemic antibodies, capable of protecting both the upper and lower respiratory tract. Previously, we reported on a virosomal RSV vaccine for intramuscular injection with intrinsic adjuvant properties mediated by an incorporated lipophilic Toll-like receptor 2 (TLR2) ligand. However, it has not been investigated whether this virosomal RSV vaccine candidate would be suitable for use in mucosal immunization strategies and if additional incorporation of other innate receptor ligands, like NOD2-ligand, could further enhance the immunogenicity and protective efficacy of the vaccine. OBJECTIVE To explore if intranasal (IN) immunization with a virosomal RSV vaccine, supplemented with TLR2 and/or NOD2-ligands, is an effective strategy to induce RSV-specific immunity. METHODS We produced RSV-virosomes carrying TLR2 (Pam3CSK4) and/or NOD2 (L18-MDP) ligands. We tested the immunopotentiating properties of these virosomes in vitro, using TLR2- and/or NOD2-ligand-responsive murine and human cell lines, and in vivo by assessing induction of protective antibody and cellular responses upon IN immunization of BALB/c mice. RESULTS Incorporation of Pam3CSK4 and/or L18-MDP potentiates the capacity of virosomes to activate (antigen-presenting) cells in vitro, as demonstrated by NF-κB induction. In vivo, incorporation of Pam3CSK4 in virosomes boosted serum IgG antibody responses and mucosal antibody responses after IN immunization. While L18-MDP alone was ineffective, incorporation of L18-MDP in Pam3CSK4-carrying virosomes further boosted mucosal antibody responses. Finally, IN immunization with adjuvanted virosomes, particularly Pam3CSK4/L18-MDP-adjuvanted-virosomes, protected mice against infection with RSV, without priming for enhanced disease. CONCLUSION Mucosal immunization with RSV-virosomes, supplemented with incorporated TLR2- and/or NOD2-ligands, represents a promising approach to induce effective and safe RSV-specific immunity.
Collapse
Affiliation(s)
- Muhammad Shafique
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Tjarko Meijerhof
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Jan Wilschut
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Aalzen de Haan
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| |
Collapse
|
44
|
Deng S, Yu K, Zhang B, Yao Y, Liu Y, He H, Zhang H, Cui M, Fu J, Lian Z, Li N. Effects of over-expression of TLR2 in transgenic goats on pathogen clearance and role of up-regulation of lysozyme secretion and infiltration of inflammatory cells. BMC Vet Res 2012; 8:196. [PMID: 23082910 PMCID: PMC3560201 DOI: 10.1186/1746-6148-8-196] [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: 05/18/2012] [Accepted: 10/17/2012] [Indexed: 01/07/2023] Open
Abstract
Background Toll-like receptor 2 (TLR2) is important to host recognition of invading gram-positive microbes. In goats, these microbes can cause serious mastitis, anthrax, tetanus, and other problems. Transgenic goats constitutively over-expressing TLR2 in many tissues serve as a suitable model for the study of the role of TLR2 over-expression in bacterial clearance. Results Capra hircus TLR2 over-expression vector (p3S-LoxP-TLR2) was used to generate transgenic goats by egg microinjection. The integration efficiency was 8.57%. Real-time PCR and immunohistochemical results confirmed that the goats over-expressing the TLR2 gene (Tg) expressed more TLR2 than wild-type goats (WT). Monocyte-macrophages from the bloodstreams of transgenic goats were stimulated with synthetic bacterial lipoprotein (Pam3CSK4) and by the promotion of interleukin-6 (IL-6) and IL-10 expression in vitro. The oxidative damage was significantly reduced, and lysozyme (LZM) secretion was found to be up-regulated. Ear tissue samples from transgenic goats that had been stimulated with Pam3CSK4 via hypodermic injection showed that transgenic individuals can undergo the inflammation response very quickly. Conclusions Over-expression of TLR2 was found to decrease radical damage to host cells through low-level production of NO and MDA and to promote the clearance of invasive bacteria by up-regulating lysozyme secretion and filtration of inflammatory cells to the infected site.
Collapse
Affiliation(s)
- Shoulong Deng
- National key Lab of Agro-Biotechnology, China Agricultural University, Beijing 100193, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
de Vries RD, McQuaid S, van Amerongen G, Yüksel S, Verburgh RJ, Osterhaus ADME, Duprex WP, de Swart RL. Measles immune suppression: lessons from the macaque model. PLoS Pathog 2012; 8:e1002885. [PMID: 22952446 PMCID: PMC3431343 DOI: 10.1371/journal.ppat.1002885] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 07/15/2012] [Indexed: 11/19/2022] Open
Abstract
Measles remains a significant childhood disease, and is associated with a transient immune suppression. Paradoxically, measles virus (MV) infection also induces robust MV-specific immune responses. Current hypotheses for the mechanism underlying measles immune suppression focus on functional impairment of lymphocytes or antigen-presenting cells, caused by infection with or exposure to MV. We have generated stable recombinant MVs that express enhanced green fluorescent protein, and remain virulent in non-human primates. By performing a comprehensive study of virological, immunological, hematological and histopathological observations made in animals euthanized at different time points after MV infection, we developed a model explaining measles immune suppression which fits with the "measles paradox". Here we show that MV preferentially infects CD45RA(-) memory T-lymphocytes and follicular B-lymphocytes, resulting in high infection levels in these populations. After the peak of viremia MV-infected lymphocytes were cleared within days, followed by immune activation and lymph node enlargement. During this period tuberculin-specific T-lymphocyte responses disappeared, whilst strong MV-specific T-lymphocyte responses emerged. Histopathological analysis of lymphoid tissues showed lymphocyte depletion in the B- and T-cell areas in the absence of apoptotic cells, paralleled by infiltration of T-lymphocytes into B-cell follicles and reappearance of proliferating cells. Our findings indicate an immune-mediated clearance of MV-infected CD45RA(-) memory T-lymphocytes and follicular B-lymphocytes, which causes temporary immunological amnesia. The rapid oligoclonal expansion of MV-specific lymphocytes and bystander cells masks this depletion, explaining the short duration of measles lymphopenia yet long duration of immune suppression.
Collapse
Affiliation(s)
| | - Stephen McQuaid
- Tissue Pathology, Belfast Health and Social Care Trust, Queen's University of Belfast, Belfast, Northern Ireland, United Kingdom
| | | | - Selma Yüksel
- Viroscience Lab, Erasmus MC, Rotterdam, The Netherlands
| | | | | | - W. Paul Duprex
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | | |
Collapse
|
46
|
Short KR, Habets MN, Hermans PWM, Diavatopoulos DA. Interactions between Streptococcus pneumoniae and influenza virus: a mutually beneficial relationship? Future Microbiol 2012; 7:609-24. [PMID: 22568716 DOI: 10.2217/fmb.12.29] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Historically, most research on infectious diseases has focused on infections with single pathogens. However, infections with pathogens often occur in the context of pre-existing viral and bacterial infections. Clinically, this is of particular relevance for coinfections with Streptococcus pneumoniae and influenza virus, which together are an important cause of global morbidity and mortality. In recent years new evidence has emerged regarding the underlying mechanisms of influenza virus-induced susceptibility to secondary pneumococcal infections, in particular regarding the sustained suppression of innate recognition of S. pneumoniae. Conversely, it is also increasingly being recognized that there is not a unidirectional effect of the virus on S. pneumoniae, but that asymptomatic pneumococcal carriage may also affect subsequent influenza virus infection and the clinical outcome. Here, we will review both aspects of pneumococcal influenza virus infection, with a particular focus on the age-related differences in pneumococcal colonization rates and invasive pneumococcal disease.
Collapse
Affiliation(s)
- Kirsty R Short
- Department of Microbiology and Immunology, The University of Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
47
|
Hussell T, Godlee A, Salek-Ardakani S, Snelgrove RJ. Respiratory viral infections: knowledge based therapeutics. Curr Opin Immunol 2012; 24:438-43. [PMID: 22770666 DOI: 10.1016/j.coi.2012.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 05/15/2012] [Accepted: 06/06/2012] [Indexed: 12/13/2022]
Abstract
Lung viral infections continue to kill millions of people worldwide. Virus-specific properties, replication kinetics and longevity affect the subsequent vigour of innate and adaptive immunity, which contribute to clinical manifestations. The point at which lung innate immunity activates is different between individuals and is determined by age, genetics, underlying conditions and infection history. On resolution of virus-induced lung disease an 'altered state of homeostasis' exists that in turn affects the next antigenic challenge. The last two years has produced a plethora of studies on the resolution of inflammatory lung disease; highlighting potential for immune modulation. In the future a more precise etiological diagnosis, combined with a knowledge of co-morbidities and an immune signature will lead to the development of more specifically targeted therapeutics.
Collapse
Affiliation(s)
- Tracy Hussell
- Imperial College London, Leukocyte Biology Section, National Heart and Lung Institute, London SW7 2AZ, UK.
| | | | | | | |
Collapse
|
48
|
Nguyen DT, Ludlow M, van Amerongen G, de Vries RD, Yüksel S, Verburgh RJ, Osterhaus ADME, Duprex WP, de Swart RL. Evaluation of synthetic infection-enhancing lipopeptides as adjuvants for a live-attenuated canine distemper virus vaccine administered intra-nasally to ferrets. Vaccine 2012; 30:5073-80. [PMID: 22705079 DOI: 10.1016/j.vaccine.2012.05.079] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 05/11/2012] [Accepted: 05/30/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND Inactivated paramyxovirus vaccines have been associated with hypersensitivity responses upon challenge infection. For measles and canine distemper virus (CDV) safe and effective live-attenuated virus vaccines are available, but for human respiratory syncytial virus and human metapneumovirus development of such vaccines has proven difficult. We recently identified three synthetic bacterial lipopeptides that enhance paramyxovirus infections in vitro, and hypothesized these could be used as adjuvants to promote immune responses induced by live-attenuated paramyxovirus vaccines. METHODS Here, we tested this hypothesis using a CDV vaccination and challenge model in ferrets. Three groups of six animals were intra-nasally vaccinated with recombinant (r) CDV(5804P)L(CCEGFPC) in the presence or absence of the infection-enhancing lipopeptides Pam3CSK4 or PHCSK4. The recombinant CDV vaccine virus had previously been described to be over-attenuated in ferrets. A group of six animals was mock-vaccinated as control. Six weeks after vaccination all animals were challenged with a lethal dose of rCDV strain Snyder-Hill expressing the red fluorescent protein dTomato. RESULTS Unexpectedly, intra-nasal vaccination of ferrets with rCDV(5804P)L(CCEGFPC) in the absence of lipopeptides resulted in good immune responses and protection against lethal challenge infection. However, in animals vaccinated with lipopeptide-adjuvanted virus significantly higher vaccine virus loads were detected in nasopharyngeal lavages and peripheral blood mononuclear cells. In addition, these animals developed significantly higher CDV neutralizing antibody titers compared to animals vaccinated with non-adjuvanted vaccine. CONCLUSIONS This study demonstrates that the synthetic cationic lipopeptides Pam3CSK4 and PHCSK4 not only enhance paramyxovirus infection in vitro, but also in vivo. Given the observed enhancement of immunogenicity their potential as adjuvants for other live-attenuated paramyxovirus vaccines should be considered.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Intranasal
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Chlorocebus aethiops
- Distemper/immunology
- Distemper/prevention & control
- Distemper Virus, Canine/immunology
- Distemper Virus, Canine/pathogenicity
- Drug Evaluation, Preclinical
- Female
- Ferrets/immunology
- Ferrets/virology
- Lipopeptides/administration & dosage
- Lymphocytes/immunology
- Lymphocytes/virology
- Neutralization Tests/methods
- Transfection
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
- Vero Cells
- Viral Load
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- D Tien Nguyen
- Department of Virology, Erasmus MC, University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lee HK, Park DW, Bae JH, Kim HJ, Shin DG, Park JS, Lee JG, Lee SJ, Bae YS, Baek SH. RGS2 is a negative regulator of STAT3-mediated Nox1 expression. Cell Signal 2012; 24:803-9. [DOI: 10.1016/j.cellsig.2011.11.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 11/07/2011] [Indexed: 10/15/2022]
|
50
|
Berencsi III G. Fetal and Neonatal Illnesses Caused or Influenced by Maternal Transplacental IgG and/or Therapeutic Antibodies Applied During Pregnancy. MATERNAL FETAL TRANSMISSION OF HUMAN VIRUSES AND THEIR INFLUENCE ON TUMORIGENESIS 2012. [PMCID: PMC7121401 DOI: 10.1007/978-94-007-4216-1_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The human fetus is protected by the mother’s antibodies. At the end of the pregnancy, the concentration of maternal antibodies is higher in the cord blood, than in the maternal circulation. Simultaneously, the immune system of the fetus begins to work and from the second trimester, fetal IgM is produced by the fetal immune system specific to microorganisms and antigens passing the maternal-fetal barrier. The same time the fetal immune system has to cope and develop tolerance and TREG cells to the maternal microchimeric cells, latent virus-carrier maternal cells and microorganisms transported through the maternal-fetal barrier. The maternal phenotypic inheritance may hide risks for the newborn, too. Antibody mediated enhancement results in dengue shock syndrome in the first 8 month of age of the baby. A series of pathologic maternal antibodies may elicit neonatal illnesses upon birth usually recovering during the first months of the life of the offspring. Certain antibodies, however, may impair the fetal or neonatal tissues or organs resulting prolonged recovery or initiating prolonged pathological processes of the children. The importance of maternal anti-idiotypic antibodies are believed to prime the fetal immune system with epitopes of etiologic agents infected the mother during her whole life before pregnancy and delivery. The chemotherapeutical and biological substances used for the therapy of the mother will be transcytosed into the fetal body during the last two trimesters of pregnancy. The long series of the therapeutic monoclonal antibodies and conjugates has not been tested systematically yet. The available data are summarised in this chapter. The innate immunity plays an important role in fetal defence. The concentration of interferon is relative high in the placenta. This is probably one reason, why the therapeutic interferon treatment of the mother does not impair the fetal development.
Collapse
Affiliation(s)
- György Berencsi III
- , Division of Virology, National Center for Epidemiology, Gyáli Street 2-6, Budapest, 1096 Hungary
| |
Collapse
|