1
|
Pittaluga JR, Birnberg-Weiss F, Serafino A, Castro JE, Castillo LA, Martire-Greco D, Barrionuevo P, Fernández GC, Landoni VI. The RNA from Pseudomonas aeruginosa Reduces Neutrophil Responses Favoring Bacterial Survival. J Innate Immun 2024; 16:489-500. [PMID: 39293427 PMCID: PMC11521516 DOI: 10.1159/000541414] [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: 05/20/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024] Open
Abstract
INTRODUCTION Epithelial and endothelial cells modulate innate immune responses in the lung, including the arrival of neutrophils (PMN), which are crucial cells for the antibacterial host defense. Cells are exposed to prokaryotic RNA (pRNA) during bacterial infections and different pRNA may promote or attenuate the inflammatory response on different immune cells. Pseudomonas aeruginosa (PAE) can cause severe pneumonia and has several immune-evading mechanisms. The aim of this study was to determine the effects of the RNA from PAE (RNAPAE) on lung epithelial, endothelial cells, and PMN, and its impact on bacterial elimination. METHODS Purified total RNAPAE was used as a stimulus on a human lung epithelial cell line (Calu-6), human microvascular endothelial cell line HMEC-1 and isolated healthy human PMN. Activation and cytokine secretion were evaluated. In addition, PMN elimination of live ECO or PAE was determined in the presence of RNAPAE. RESULTS We found that RNAPAE either induced a pro-inflammatory response on Calu-6 and HMEC-1 or PMN. Pre-stimulation of PMN with RNAPAE diminished activation and chemotaxis induced by live bacteria. Moreover, we found that RNAPAE reduced phagocytosis of live ECO. Finally, we also found that non-degraded fragments of small RNA (<200 bp) were responsible for the PMN microbicidal attenuation during PAE elimination. CONCLUSION Our results indicated that short fragments of RNAPAE diminished the immune response of PMN favoring bacterial survival.
Collapse
Affiliation(s)
- Jose R Pittaluga
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Federico Birnberg-Weiss
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Agustina Serafino
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Joselyn E Castro
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Luis A Castillo
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Daiana Martire-Greco
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Paula Barrionuevo
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Gabriela C Fernández
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Verónica I Landoni
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
2
|
Lin B, Wang J, Zhang Y. Bacterial dynamics in the progression of caries to apical periodontitis in primary teeth of children with severe early childhood caries. Front Microbiol 2024; 15:1418261. [PMID: 39323882 PMCID: PMC11422202 DOI: 10.3389/fmicb.2024.1418261] [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: 04/16/2024] [Accepted: 07/10/2024] [Indexed: 09/27/2024] Open
Abstract
Background Early childhood caries (ECC) are a prevalent chronic disease in young children. However, there has been limited research on the microbiota in different tissue levels of the same tooth in children with ECC. This study aimed to investigate the dynamic changes in bacterial diversity during the progression of Severe Early Childhood Caries (S-ECC) within the same tooth, from the tooth surface to the root canal, by collecting tissue samples from different areas of the affected tooth. Methods Twenty primary teeth with periapical periodontitis were selected from 20 children aged 3-5 years, with 100 samples collected from the different layers: uncavitated buccal enamel surface without white spot lesion (surface), the outermost layer of the dentin carious lesion (superficial), the inner layer of carious dentin (deep), necrotic pulp tissue (pulp), and root exudate (exudate). The taxonomy of each OTU representative sequence was analyzed against the 16S rRNA database. Comparisons of alpha diversity between groups were performed. The number of shared and unique genera between groups counted. Beta diversity was contrasted to evaluate differences in bacterial community composition, and the relationships between the microbiota and samples were analyzed. The heatmap analysis of the 30 most abundant genera was used, which highlighted their relative distribution and abundance. The significantly abundant taxa (phylum to genera) of bacteria among the different groups were identified. The differences of relative abundance between bacterial genera among the five groups were analyzed. Significant Spearman correlations were noted, and visualization of the co-occurrence network was conducted. Results Bacterial 16S rRNA gene sequencing showed that most genera were present in all layers, with the number of shared genera increasing as the disease advanced. The bacterial communities and core genera in the co-occurrence network changed with progression to severe ECC. Conclusion An increase in both the quantity and complexity of bacterial interactions was observed. This study emphasized the importance of paying attention to the relationship between microbial species rather than just checking changes in bacterial species structure when investigating the role of bacteria in disease progression.
Collapse
Affiliation(s)
- Bichen Lin
- First Clinical Division, Peking University School and Hospital of Stomatology and National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Jinfeng Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yifei Zhang
- Department of Dental Materials, Peking University School and Hospital of Stomatology and National Center for Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| |
Collapse
|
3
|
Bork F, Greve CL, Youn C, Chen S, N C Leal V, Wang Y, Fischer B, Nasri M, Focken J, Scheurer J, Engels P, Dubbelaar M, Hipp K, Zalat B, Szolek A, Wu MJ, Schittek B, Bugl S, Kufer TA, Löffler MW, Chamaillard M, Skokowa J, Kramer D, Archer NK, Weber ANR. naRNA-LL37 composite DAMPs define sterile NETs as self-propagating drivers of inflammation. EMBO Rep 2024; 25:2914-2949. [PMID: 38783164 PMCID: PMC11239898 DOI: 10.1038/s44319-024-00150-5] [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: 08/24/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024] Open
Abstract
Neutrophil extracellular traps (NETs) are a key antimicrobial feature of cellular innate immunity mediated by polymorphonuclear neutrophils (PMNs). NETs counteract microbes but are also linked to inflammation in atherosclerosis, arthritis, or psoriasis by unknown mechanisms. Here, we report that NET-associated RNA (naRNA) stimulates further NET formation in naive PMNs via a unique TLR8-NLRP3 inflammasome-dependent pathway. Keratinocytes respond to naRNA with expression of psoriasis-related genes (e.g., IL17, IL36) via atypical NOD2-RIPK signaling. In vivo, naRNA drives temporary skin inflammation, which is drastically ameliorated by genetic ablation of RNA sensing. Unexpectedly, the naRNA-LL37 'composite damage-associated molecular pattern (DAMP)' is pre-stored in resting neutrophil granules, defining sterile NETs as inflammatory webs that amplify neutrophil activation. However, the activity of the naRNA-LL37 DAMP is transient and hence supposedly self-limiting under physiological conditions. Collectively, upon dysregulated NET release like in psoriasis, naRNA sensing may represent both a potential cause of disease and a new intervention target.
Collapse
Affiliation(s)
- Francesca Bork
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Carsten L Greve
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Christine Youn
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Sirui Chen
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Vinicius N C Leal
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
- Laboratory of Immunogenetics, Department of Immunology, Institute of Biomedical Science, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Berenice Fischer
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Masoud Nasri
- Division of Translational Oncology, Department of Oncology, Hematology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Otfried-Müller Str. 10, 72076, Tübingen, Germany
| | - Jule Focken
- Department of Dermatology, University Hospital Tübingen, Liebermeisterstr. 25, 72076, Tübingen, Germany
| | - Jasmin Scheurer
- Department of Dermatology, University Hospital Tübingen, Liebermeisterstr. 25, 72076, Tübingen, Germany
| | - Pujan Engels
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Marissa Dubbelaar
- Institute of Immunology, Department of Peptide-based Immunotherapy, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
- Quantitative Biology Center (QBiC), University of Tübingen, Auf der Morgenstelle 10, 72076, Tübingen, Germany
| | - Katharina Hipp
- Electron Microscopy Facility, Max Planck Institute for Biology Tübingen, Max-Planck-Ring 5, 72076, Tübingen, Germany
| | - Baher Zalat
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Andras Szolek
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Meng-Jen Wu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Birgit Schittek
- Department of Dermatology, University Hospital Tübingen, Liebermeisterstr. 25, 72076, Tübingen, Germany
- iFIT - Cluster of Excellence (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- CMFI - Cluster of Excellence (EXC 2124) "Controlling microbes to fight infection", University of Tübingen, Tübingen, Germany
| | - Stefanie Bugl
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Thomas A Kufer
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, Fruwirthstr. 12, 70593, Stuttgart, Germany
| | - Markus W Löffler
- Institute of Immunology, Department of Peptide-based Immunotherapy, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
- iFIT - Cluster of Excellence (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty, University of Tübingen, Otfried-Müller-Str. 4/1, 72076, Tübingen, Germany
| | - Mathias Chamaillard
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000, Lille, France
| | - Julia Skokowa
- Division of Translational Oncology, Department of Oncology, Hematology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Otfried-Müller Str. 10, 72076, Tübingen, Germany
- iFIT - Cluster of Excellence (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Daniela Kramer
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Alexander N R Weber
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.
- iFIT - Cluster of Excellence (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.
- CMFI - Cluster of Excellence (EXC 2124) "Controlling microbes to fight infection", University of Tübingen, Tübingen, Germany.
| |
Collapse
|
4
|
Willmann K, Moita LF. Physiologic disruption and metabolic reprogramming in infection and sepsis. Cell Metab 2024; 36:927-946. [PMID: 38513649 DOI: 10.1016/j.cmet.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/23/2024]
Abstract
Effective responses against severe systemic infection require coordination between two complementary defense strategies that minimize the negative impact of infection on the host: resistance, aimed at pathogen elimination, and disease tolerance, which limits tissue damage and preserves organ function. Resistance and disease tolerance mostly rely on divergent metabolic programs that may not operate simultaneously in time and space. Due to evolutionary reasons, the host initially prioritizes the elimination of the pathogen, leading to dominant resistance mechanisms at the potential expense of disease tolerance, which can contribute to organ failure. Here, we summarize our current understanding of the role of physiological perturbations resulting from infection in immune response dynamics and the metabolic program requirements associated with resistance and disease tolerance mechanisms. We then discuss how insight into the interplay of these mechanisms could inform future research aimed at improving sepsis outcomes and the potential for therapeutic interventions.
Collapse
Affiliation(s)
- Katharina Willmann
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Luis F Moita
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal; Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
5
|
Pradeu T, Thomma BPHJ, Girardin SE, Lemaitre B. The conceptual foundations of innate immunity: Taking stock 30 years later. Immunity 2024; 57:613-631. [PMID: 38599162 DOI: 10.1016/j.immuni.2024.03.007] [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: 01/14/2024] [Revised: 02/23/2024] [Accepted: 03/06/2024] [Indexed: 04/12/2024]
Abstract
While largely neglected over decades during which adaptive immunity captured most of the attention, innate immune mechanisms have now become central to our understanding of immunology. Innate immunity provides the first barrier to infection in vertebrates, and it is the sole mechanism of host defense in invertebrates and plants. Innate immunity also plays a critical role in maintaining homeostasis, shaping the microbiota, and in disease contexts such as cancer, neurodegeneration, metabolic syndromes, and aging. The emergence of the field of innate immunity has led to an expanded view of the immune system, which is no longer restricted to vertebrates and instead concerns all metazoans, plants, and even prokaryotes. The study of innate immunity has given rise to new concepts and language. Here, we review the history and definition of the core concepts of innate immunity, discussing their value and fruitfulness in the long run.
Collapse
Affiliation(s)
- Thomas Pradeu
- CNRS UMR 5164 ImmunoConcept, University of Bordeaux, Bordeaux, France; Department of Biological and Medical Sciences, University of Bordeaux, Bordeaux, France; Presidential Fellow, Chapman University, Orange, CA, USA.
| | - Bart P H J Thomma
- Institute for Plant Sciences, University of Cologne, Cologne, Germany
| | - Stephen E Girardin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Bruno Lemaitre
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
6
|
Shankar-Hari M, Calandra T, Soares MP, Bauer M, Wiersinga WJ, Prescott HC, Knight JC, Baillie KJ, Bos LDJ, Derde LPG, Finfer S, Hotchkiss RS, Marshall J, Openshaw PJM, Seymour CW, Venet F, Vincent JL, Le Tourneau C, Maitland-van der Zee AH, McInnes IB, van der Poll T. Reframing sepsis immunobiology for translation: towards informative subtyping and targeted immunomodulatory therapies. THE LANCET. RESPIRATORY MEDICINE 2024; 12:323-336. [PMID: 38408467 PMCID: PMC11025021 DOI: 10.1016/s2213-2600(23)00468-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/27/2023] [Accepted: 12/07/2023] [Indexed: 02/28/2024]
Abstract
Sepsis is a common and deadly condition. Within the current model of sepsis immunobiology, the framing of dysregulated host immune responses into proinflammatory and immunosuppressive responses for the testing of novel treatments has not resulted in successful immunomodulatory therapies. Thus, the recent focus has been to parse observable heterogeneity into subtypes of sepsis to enable personalised immunomodulation. In this Personal View, we highlight that many fundamental immunological concepts such as resistance, disease tolerance, resilience, resolution, and repair are not incorporated into the current sepsis immunobiology model. The focus for addressing heterogeneity in sepsis should be broadened beyond subtyping to encompass the identification of deterministic molecular networks or dominant mechanisms. We explicitly reframe the dysregulated host immune responses in sepsis as altered homoeostasis with pathological disruption of immune-driven resistance, disease tolerance, resilience, and resolution mechanisms. Our proposal highlights opportunities to identify novel treatment targets and could enable successful immunomodulation in the future.
Collapse
Affiliation(s)
- Manu Shankar-Hari
- Institute for Regeneration and Repair, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK.
| | - Thierry Calandra
- Service of Immunology and Allergy, Center of Human Immunology Lausanne, Department of Medicine and Department of Laboratory Medicine and Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | | | - Michael Bauer
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - W Joost Wiersinga
- Center for Experimental and Molecular Medicine and Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Hallie C Prescott
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Julian C Knight
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Kenneth J Baillie
- Institute for Regeneration and Repair, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Lieuwe D J Bos
- Department of Intensive Care, Academic Medical Center, Amsterdam, Netherlands
| | - Lennie P G Derde
- Intensive Care Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Simon Finfer
- Critical Care Division, The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | - Richard S Hotchkiss
- Department of Anesthesiology and Critical Care Medicine, Washington University School of Medicine in St Louis, St Louis, MO, USA
| | - John Marshall
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, ON, Canada
| | | | - Christopher W Seymour
- Department of Critical Care Medicine, The Clinical Research, Investigation, and Systems Modeling of Acute illness (CRISMA) Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fabienne Venet
- Immunology Laboratory, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | | | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris-Saclay University, Paris, France
| | - Anke H Maitland-van der Zee
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Iain B McInnes
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine and Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
7
|
Xu J, Jia Z, Xiao S, Long C, Wang L. Effects of Enterotoxigenic Escherichia coli Challenge on Jejunal Morphology and Microbial Community Profiles in Weaned Crossbred Piglets. Microorganisms 2023; 11:2646. [PMID: 38004658 PMCID: PMC10672776 DOI: 10.3390/microorganisms11112646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/25/2023] [Accepted: 10/05/2023] [Indexed: 11/26/2023] Open
Abstract
Pathogenic enterotoxigenic Escherichia coli (ETEC) is a major cause of bacterial diarrhea in weaning piglets, which are vulnerable to changes in environment and feed. This study aimed to determine the effects of the ETEC challenge on piglet growth performance, diarrhea rate, jejunal microbial profile, jejunal morphology and goblet cell distribution. A total of 13 piglets from one litter were selected on postnatal day 21 and assigned to treatments with or without ETEC challenge at 1 × 108 CFUs, as ETEC group or control group, respectively. On postnatal day 28, samples were collected, followed by the detection of serum biochemical indexes and inflammatory indicators, HE staining, PAS staining and 16S rDNA gene amplicon sequencing. Results showed that the growth performance decreased, while the diarrhea rate increased for the ETEC group. The jejunum is the main segment of the injured intestine during the ETEC challenge. Compared with the control, the ETEC group displayed fewer goblet cells in the jejunum, where goblet cells are more distributed at the crypt and less distributed at the villus. In addition, ETEC piglets possessed higher abundances of the genus Desulfovibrio, genus Oxalobacter and genus Peptococus and lower abundances of the genus Prevotella 2, genus Flavonifractor and genus Blautra. In terms of alpha diversity, Chao 1 and observed features indexes were both increased for the ETEC group. Our study provides insights into jejunal histopathological impairment and microbial variation in response to ETEC infection for weaned piglets and is a valuable reference for researchers engaged in animal health research to select stress models.
Collapse
Affiliation(s)
- Juan Xu
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410081, China; (J.X.); (Z.J.); (S.X.)
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, National Center of Technology Innovation for Synthetic Biology, Tianjin 300308, China
| | - Zhen Jia
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410081, China; (J.X.); (Z.J.); (S.X.)
| | - Shu Xiao
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410081, China; (J.X.); (Z.J.); (S.X.)
| | - Cimin Long
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410081, China; (J.X.); (Z.J.); (S.X.)
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, National Center of Technology Innovation for Synthetic Biology, Tianjin 300308, China
| | - Leli Wang
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410081, China; (J.X.); (Z.J.); (S.X.)
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, National Center of Technology Innovation for Synthetic Biology, Tianjin 300308, China
| |
Collapse
|
8
|
Bullen CK, Singh AK, Krug S, Lun S, Thakur P, Srikrishna G, Bishai WR. MDA5 RNA-sensing pathway activation by Mycobacterium tuberculosis promotes innate immune subversion and pathogen survival. JCI Insight 2023; 8:e166242. [PMID: 37725440 PMCID: PMC10619499 DOI: 10.1172/jci.insight.166242] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 09/13/2023] [Indexed: 09/21/2023] Open
Abstract
Host cytosolic sensing of Mycobacterium tuberculosis (M. tuberculosis) RNA by the RIG-I-like receptor (RLR) family perturbs innate immune control within macrophages; however, a distinct role of MDA5, a member of the RLR family, in M. tuberculosis pathogenesis has yet to be fully elucidated. To further define the role of MDA5 in M. tuberculosis pathogenesis, we evaluated M. tuberculosis intracellular growth and innate immune responses in WT and Mda5-/- macrophages. Transfection of M. tuberculosis RNA strongly induced proinflammatory cytokine production in WT macrophages, which was abrogated in Mda5-/- macrophages. M. tuberculosis infection in macrophages induced MDA5 protein expression, accompanied by an increase in MDA5 activation as assessed by multimer formation. IFN-γ-primed Mda5-/- macrophages effectively contained intracellular M. tuberculosis proliferation to a markedly greater degree than WT macrophages. Further comparisons of WT versus Mda5-/- macrophages revealed that during M. tuberculosis infection MDA5 contributed to IL-1β production and inflammasome activation and that loss of MDA5 led to a substantial increase in autophagy. In the mouse TB model, loss of MDA5 conferred host survival benefits with a concomitant reduction in M. tuberculosis bacillary burden. These data reveal that loss of MDA5 is host protective during M. tuberculosis infection in vitro and in vivo, suggesting that M. tuberculosis exploits MDA5 to subvert immune containment.
Collapse
|
9
|
Yadav S, Dalai P, Gowda S, Nivsarkar M, Agrawal-Rajput R. Azithromycin alters Colony Stimulating Factor-1R (CSF-1R) expression and functional output of murine bone marrow-derived macrophages: A novel report. Int Immunopharmacol 2023; 123:110688. [PMID: 37499396 DOI: 10.1016/j.intimp.2023.110688] [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: 11/19/2022] [Revised: 07/18/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023]
Abstract
Antibiotic treatment may lead to side effects that require mechanistic explanation. We investigated the effect of azithromycin (AZM) treatment on bone marrow-derived macrophage (Mφ) generation, their functional output, and the subsequent effect on bacterial clearance in a mouse model of S. flexneri infection. To our fascination, AZM increased PU.1, C/EBPβ, CSF-1R/pCSF-1R expressions leading to M2-skewed in vitro BMDM generation. Altered Mφ-functions like- phagocytosis, oxidative stress generation, inflammasome-activation, cytokine release, and phenotype (pro-inflammatory-M1, anti-inflammatory-M2) even in the presence of infection were observed with AZM treatment. AZM increased CD206, egr2, arg1 (M2-marker) expression and activity while reducing CD68, inducible nitric oxide (iNOS) expression, and activity (M1-marker) in Mφs during infection. Pro-inflammatory cytokines (TNF-α, IL-12, IL-1β) were reduced and anti-inflammatory IL-10 release was augmented by AZM-treated-iMφs (aiMφs) along with decreased asc, nlrp3, aim2, nlrp1a, caspase1 expressions, and caspase3 activity signifying that aMφs/aiMφs were primed towards an anti-inflammatory phenotype. Interestingly, CSF-1R blockade increased NO, IL-12, TNF-α, IL-1β, decreased TGF-β release, and CD206 expression in aiMφs. T-cell co-stimulatory molecule cd40, cd86, and cd80 expressions were decreased in ai/aM1-Mφs and co-cultured CD8+, CD4+ T-cells had decreased proliferation, t-bet, IFN-γ, IL-17, IL-2 but increased foxp3, TGF-β, IL-4 which were rescued with CSF-1R blockade. Thus AZM affected Mφ-functions and subsequent T-cell responses independent of its antibacterial actions. This was validated in the balb/c model of S. flexneri infection. We conclude that AZM skewed BMDM generation to anti-inflammatory M2-like via increased CSF-1R expression. This warrants further investigation of AZM-induced altered-Mφ-generation during intracellular infections.
Collapse
Affiliation(s)
- Shivani Yadav
- Department of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, India
| | - Parmeswar Dalai
- Department of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, India
| | - Sharath Gowda
- Department of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, India
| | | | - Reena Agrawal-Rajput
- Department of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, India.
| |
Collapse
|
10
|
Mbatha LS, Akinyelu J, Maiyo F, Kudanga T. Future prospects in mRNA vaccine development. Biomed Mater 2023; 18:052006. [PMID: 37589309 DOI: 10.1088/1748-605x/aceceb] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/02/2023] [Indexed: 08/18/2023]
Abstract
The recent advancements in messenger ribonucleic acid (mRNA) vaccine development have vastly enhanced their use as alternatives to conventional vaccines in the prevention of various infectious diseases and treatment of several types of cancers. This is mainly due to their remarkable ability to stimulate specific immune responses with minimal clinical side effects. This review gives a detailed overview of mRNA vaccines currently in use or at various stages of development, the recent advancements in mRNA vaccine development, and the challenges encountered in their development. Future perspectives on this technology are also discussed.
Collapse
Affiliation(s)
- Londiwe Simphiwe Mbatha
- Department of Biotechnology and Food Science, Durban University of Technology, PO Box 1334, Durban 4000, South Africa
| | - Jude Akinyelu
- Department of Biochemistry, Federal University Oye-Ekiti, Ekiti state, Nigeria
| | - Fiona Maiyo
- Department of Medical Sciences, Kabarak University, Nairobi, Kenya
| | - Tukayi Kudanga
- Department of Biotechnology and Food Science, Durban University of Technology, PO Box 1334, Durban 4000, South Africa
| |
Collapse
|
11
|
Gronke K, Nguyen M, Santamaria N, Schumacher J, Yang Y, Sonnert N, Leopold S, Martin AL, Hallet R, Richter K, Schubert DA, Daniel GM, Dylus D, Forkel M, Vieira SM, Schwinge D, Schramm C, Lassen KG, Piali L, Palm NW, Bieniossek C, Kriegel MA. Human Th17- and IgG3-associated autoimmunity induced by a translocating gut pathobiont. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.546430. [PMID: 37425769 PMCID: PMC10327010 DOI: 10.1101/2023.06.29.546430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Extraintestinal autoimmune diseases are multifactorial with translocating gut pathobionts implicated as instigators and perpetuators in mice. However, the microbial contributions to autoimmunity in humans remain largely unclear, including whether specific pathological human adaptive immune responses are triggered by such pathobionts. We show here that the translocating pathobiont Enterococcus gallinarum induces human IFNγ + Th17 differentiation and IgG3 subclass switch of anti- E. gallinarum RNA and correlating anti-human RNA autoantibody responses in patients with systemic lupus erythematosus and autoimmune hepatitis. Human Th17 induction by E. gallinarum is cell-contact dependent and involves TLR8-mediated human monocyte activation. In murine gnotobiotic lupus models, E. gallinarum translocation triggers IgG3 anti-RNA autoantibody titers that correlate with renal autoimmune pathophysiology and with disease activity in patients. Overall, we define cellular mechanisms of how a translocating pathobiont induces human T- and B-cell-dependent autoimmune responses, providing a framework for developing host- and microbiota-derived biomarkers and targeted therapies in extraintestinal autoimmune diseases. One Sentence Summary Translocating pathobiont Enterococcus gallinarum promotes human Th17 and IgG3 autoantibody responses linked to disease activity in autoimmune patients.
Collapse
|
12
|
Konje JC, Al Beloushi M, Ahmed B. Immunisation against COVID-19 in Pregnancy and of Women Planning Pregnancy. Viruses 2023; 15:v15030621. [PMID: 36992330 PMCID: PMC10059008 DOI: 10.3390/v15030621] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 03/03/2023] Open
Abstract
Following reports of the first human SARS-CoV2 infection in December 2019 from Wuhan Province, China, there was such rapid spread that by March 2021, the World Health Organization (WHO) had declared a pandemic. Over 6.5 million people have died from this infection worldwide, although this is most likely an underestimate. Until vaccines became available, mortality and severe morbidity were costly in terms of life lost as well as the cost of supporting the severely and acutely ill. Vaccination changed the landscape, and following worldwide adoption, life has gradually been returning to normal. The speed of production of the vaccines was unprecedented and undoubtedly ushered in a new era in the science of fighting infections. The developed vaccines were on the already known platforms for vaccine delivery: inactivated virus, virus vector, virus-like particles (VLP) subunit, DNA and mRNA. The mRNA platform was used for the first time to deliver vaccines to humans. An understanding of these platforms and the pros and cons of each are important for clinicians who are often challenged by the recipients on the advantages and risks of these vaccines. These vaccines have so far and reassuringly been shown to be safe in reproduction (with no effect on gametes) and pregnancy (not associated with congenital malformations). However, safety remains paramount and continuing vigilance is critical, especially against rare fatal complications such as vaccine-induced thrombocytopenia and myocarditis. Finally, the waning immunity months after vaccination means repeated immunisation is likely to be ongoing, but just how often and how many such revaccinations should be recommended remains uncertain. Research into other vaccines and alternate delivery methods should continue as this infection is likely to be around for a long time.
Collapse
Affiliation(s)
- Justin C. Konje
- Feto-Maternal Centre Al Markhiya, Doha P.O. Box 34181, Qatar
- Obstetrics and Gynecology Department, Weill Cornell Medicine Qatar, Doha P.O. Box 24144, Qatar
- Obstetrics and Gynaecology, Department of Health Sciences, University of Leicester, Leicester LE2 7LX, UK
- Correspondence: ; Tel.: +974-7777-8375
| | - Mariam Al Beloushi
- Women’s Wellness and Research Centre, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar
- Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar
| | - Badreldeen Ahmed
- Feto-Maternal Centre Al Markhiya, Doha P.O. Box 34181, Qatar
- Obstetrics and Gynecology Department, Weill Cornell Medicine Qatar, Doha P.O. Box 24144, Qatar
- Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
13
|
Le-Trilling VTK, Jagnjić A, Brizić I, Eilbrecht M, Wohlgemuth K, Rožmanić C, Herdman A, Hoffmann K, Westendorf AM, Hengel H, Jonjić S, Trilling M. Maternal antibodies induced by a live attenuated vaccine protect neonatal mice from cytomegalovirus. NPJ Vaccines 2023; 8:8. [PMID: 36737485 PMCID: PMC9898546 DOI: 10.1038/s41541-023-00602-4] [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: 10/06/2021] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Human cytomegalovirus (HCMV) frequently causes congenital infections, resulting in birth defects and developmental disorders. A vaccine is needed, but unavailable. We analyzed the potential of CMV mutants, lacking their STAT2 antagonists to serve as live attenuated vaccine viruses in mice. Infections with attenuated viruses elicited strong ELISA-reactive binding IgG responses and induced neutralizing antibodies as well as antibodies stimulating cellular Fcγ receptors, including the antibody-dependent cellular cytotoxicity (ADCC)-eliciting receptors FcγRIII/CD16 and FcγRIV. Accordingly, vaccinated mice were fully protected against challenge infections. Female mice vaccinated prior to gestation transmitted CMV-specific IgG to their offspring, which protected the progeny from perinatal infections in a mouse model for congenital CMV disease. To define the role of maternal antibodies, female mice either capable or incapable of producing antibodies were vaccinated and subsequently bred to males of the opposite genotype. Challenge infections of the genotypically identical F1 generation revealed the indispensability of maternal antibodies for vaccine-induced protection against cytomegaloviruses.
Collapse
Affiliation(s)
- Vu Thuy Khanh Le-Trilling
- grid.5718.b0000 0001 2187 5445Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Andreja Jagnjić
- grid.5718.b0000 0001 2187 5445Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ilija Brizić
- grid.22939.330000 0001 2236 1630Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Mareike Eilbrecht
- grid.5718.b0000 0001 2187 5445Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kerstin Wohlgemuth
- grid.5718.b0000 0001 2187 5445Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Carmen Rožmanić
- grid.22939.330000 0001 2236 1630Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Alan Herdman
- grid.5718.b0000 0001 2187 5445Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Katja Hoffmann
- grid.5963.9Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Astrid M. Westendorf
- grid.5718.b0000 0001 2187 5445Institute for Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hartmut Hengel
- grid.5963.9Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stipan Jonjić
- grid.22939.330000 0001 2236 1630Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Mirko Trilling
- grid.5718.b0000 0001 2187 5445Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
14
|
Infection and Immunity. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
15
|
Serafino A, Marin Franco JL, Maio M, Trotta A, Genoula M, Castillo LA, Birnberg Weiss F, Pittaluga JR, Balboa L, Barrionuevo P, Milillo MA. Brucella abortus RNA does not polarize macrophages to a particular profile but interferes with M1 polarization. PLoS Negl Trop Dis 2022; 16:e0010950. [PMID: 36441810 PMCID: PMC9731426 DOI: 10.1371/journal.pntd.0010950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 12/08/2022] [Accepted: 11/14/2022] [Indexed: 11/29/2022] Open
Abstract
Monocytes and macrophages play a central role in chronic brucellosis. Brucella abortus (Ba) is an intracellular pathogen that survives inside these cells. On the other hand, macrophages could be differentiated into classical (M1), alternative (M2) or other less-identified profiles. We have previously shown that Ba RNA (a bacterial viability-associated PAMP or vita-PAMP) is a key molecule by which Ba can evade the host immune response. However, we did not know if macrophages could be polarized by this vita-PAMP. To assess this, we used two different approaches: we evaluated if Ba RNA per se was able to differentiate macrophages to M1 or M2 or, given that Ba survives inside macrophages once a Th1 response is established (i.e., in the presence of IFN-γ), we also analysed if Ba RNA could interfere with M1 polarization. We found that Ba RNA alone does not polarize to M1 or M2 but activates human macrophages instead. However, our results show that Ba RNA does interfere with M1 polarization while they are being differentiated. This vita-PAMP diminished the M1-induced CD64, and MHC-II surface expression on macrophages at 48 h. This phenomenon was not associated with an alternative activation of these cells (M2), as shown by unchanged CD206, DC-SIGN and CD163 surface expression. When evaluating glucose metabolism, we found that Ba RNA did not modify M1 glucose consumption or lactate production. However, production of Nitrogen Reactive Species (NRS) did diminish in Ba RNA-treated M1 macrophages. Overall, our results show that Ba RNA could alter the proper immune response set to counterattack the bacteria that could persist in the host establishing a chronic infection.
Collapse
Affiliation(s)
- Agustina Serafino
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - José L. Marin Franco
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Mariano Maio
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Aldana Trotta
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Melanie Genoula
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Luis A. Castillo
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Federico Birnberg Weiss
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - José R. Pittaluga
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Luciana Balboa
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - Paula Barrionuevo
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| | - M. Ayelén Milillo
- Instituto de Medicina Experimental—Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Academia Nacional de Medicina; Buenos Aires, Argentina
| |
Collapse
|
16
|
Rainard P, Gilbert FB, Germon P. Immune defenses of the mammary gland epithelium of dairy ruminants. Front Immunol 2022; 13:1031785. [PMID: 36341445 PMCID: PMC9634088 DOI: 10.3389/fimmu.2022.1031785] [Citation(s) in RCA: 19] [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: 08/30/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
The epithelium of the mammary gland (MG) fulfills three major functions: nutrition of progeny, transfer of immunity from mother to newborn, and its own defense against infection. The defense function of the epithelium requires the cooperation of mammary epithelial cells (MECs) with intraepithelial leucocytes, macrophages, DCs, and resident lymphocytes. The MG is characterized by the secretion of a large amount of a nutrient liquid in which certain bacteria can proliferate and reach a considerable bacterial load, which has conditioned how the udder reacts against bacterial invasions. This review presents how the mammary epithelium perceives bacteria, and how it responds to the main bacterial genera associated with mastitis. MECs are able to detect the presence of actively multiplying bacteria in the lumen of the gland: they express pattern recognition receptors (PRRs) that recognize microbe-associated molecular patterns (MAMPs) released by the growing bacteria. Interactions with intraepithelial leucocytes fine-tune MECs responses. Following the onset of inflammation, new interactions are established with lymphocytes and neutrophils recruited from the blood. The mammary epithelium also identifies and responds to antigens, which supposes an antigen-presenting capacity. Its responses can be manipulated with drugs, plant extracts, probiotics, and immune modifiers, in order to increase its defense capacities or reduce the damage related to inflammation. Numerous studies have established that the mammary epithelium is a genuine effector of both innate and adaptive immunity. However, knowledge gaps remain and newly available tools offer the prospect of exciting research to unravel and exploit the multiple capacities of this particular epithelium.
Collapse
|
17
|
Differential Homing Receptor Profiles of Lymphocytes Induced by Attenuated versus Live Plasmodium falciparum Sporozoites. Vaccines (Basel) 2022; 10:vaccines10101768. [PMID: 36298634 PMCID: PMC9611729 DOI: 10.3390/vaccines10101768] [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: 09/10/2022] [Revised: 10/02/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
The onset of an adaptive immune response provides the signals required for differentiation of antigen-specific lymphocytes into effector cells and imprinting of these cells for re-circulation to the most appropriate anatomical site (i.e., homing). Lymphocyte homing is governed by the expression of tissue-specific lymphocyte homing receptors that bind to unique tissue-specific ligands on endothelial cells. In this study, a whole-parasite malaria vaccine (radiation-attenuated sporozoites (RAS)) was used as a model system to establish homing receptor signatures induced by the parasite delivered through mosquito bite to provide a benchmark of desirable homing receptors for malaria vaccine developers. This immunization regimen resulted in the priming of antigen-specific B cells and CD8+ T cells for homing primarily to the skin and T/B cell compartments of secondary lymphoid organs. Infection with live sporozoites, however, triggers the upregulation of homing receptor for the liver and the skin, demonstrating that there is a difference in the signal provided by attenuated vs. live sporozoites. This is the first report on imprinting of homing routes by Plasmodium sporozoites and, surprisingly, it also points to additional, yet to be identified, signals provided by live parasites that prime lymphocytes for homing to the liver. The data also demonstrate the utility of this method for assessing the potential of vaccine formulations to direct antigen-specific lymphocytes to the most relevant anatomical site, thus potentially impacting vaccine efficacy.
Collapse
|
18
|
Bagheri S, Paudel S, Wijewardana V, Kangethe RT, Cattoli G, Hess M, Liebhart D, Mitra T. Production of interferon gamma and interleukin 17A in chicken T-cell subpopulations hallmarks the stimulation with live, irradiated and killed avian pathogenic Escherichia coli. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 133:104408. [PMID: 35390358 DOI: 10.1016/j.dci.2022.104408] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 06/14/2023]
Abstract
Avian pathogenic Escherichia coli (APEC) causes colibacillosis with different clinical manifestations. The disease is associated with compromised animal welfare and results in substantial economic losses in poultry production worldwide. So far, immunological mechanisms of protection against colibacillosis are not comprehensively resolved. Therefore, the present study aimed to use an ex vivo model applying chicken mononuclear cells stimulated by live and inactivated APEC. For this purpose, an 8-color flow cytometry panel was set up to target viable chicken immune cells including CD45+, CD8α+, CD4+, TCR-γδ+, Bu-1+ cells and monocytes/macrophages along with the cytokines interferon gamma (IFN-γ) or interleukin 17A (IL-17A). The 8-color flow cytometry panel was applied to investigate the effect of live and two different types of inactivated APEC (formalin-killed APEC and irradiated APEC) on the cellular immune response. For that, mononuclear cells from spleen, lung and blood of 10-week-old specific pathogen-free layer birds were isolated and stimulated with live, irradiated or killed APEC. Intracellular cytokine staining and RT-qPCR assays were applied for the detection of IFN-γ and IL-17A protein level, as well as at mRNA level for spleenocytes. Ex vivo stimulation of isolated splenocytes, lung and peripheral blood mononuclear cells (PBMCs) from chickens with live, irradiated or killed APEC showed an increasing number of IFN-γ and IL-17A producing cells at protein and mRNA level. Phenotyping of the cells from blood and organs revealed that IFN-γ and IL-17A were mainly produced by CD8α+, TCR-γδ+ T cells as well as CD4+ T cells following stimulation with APEC. Expression level of cytokines were very similar following stimulation with live and irradiated APEC but lower when killed APEC were applied. Consequently, in the present study, an ex vivo model using mononuclear cells of chickens was applied to investigate the cellular immune response against APEC. The results suggest the relevance of IFN-γ and IL-17A production in different immune cells following APEC infection in chickens which needs to be further investigated in APEC primed birds.
Collapse
Affiliation(s)
- Sina Bagheri
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria
| | - Surya Paudel
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria
| | - Viskam Wijewardana
- Animal Production and Health Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency IAEA, Vienna, Austria
| | - Richard Thiga Kangethe
- Animal Production and Health Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency IAEA, Vienna, Austria
| | - Giovanni Cattoli
- Animal Production and Health Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency IAEA, Vienna, Austria
| | - Michael Hess
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria
| | - Dieter Liebhart
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria
| | - Taniya Mitra
- Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria.
| |
Collapse
|
19
|
Milillo MA, Velásquez LN, Barrionuevo P. Microbial RNA, the New PAMP of Many Faces. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.924719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Traditionally, pathogen-associated molecular patterns (PAMPs) were described as structural molecular motifs shared by different classes of microorganisms. However, it was later discovered that the innate immune system is also capable of distinguishing metabolically active microbes through the detection of a special class of viability-associated PAMPs (vita-PAMPs). Indeed, recognition of vita-PAMPs triggers an extra warning sign not provoked by dead bacteria. Bacterial RNA is classified as a vita-PAMP since it stops being synthesized once the microbes are eliminated. Most of the studies in the literature have focused on the pro-inflammatory capacity of bacterial RNA on macrophages, neutrophils, endothelial cells, among others. However, we, and other authors, have shown that microbial RNA also has down-modulatory properties. More specifically, bacterial RNA can reduce the surface expression of MHC class I and MHC class II on monocytes/macrophages and help evade CD8+ and CD4+ T cell-mediated immune surveillance. This phenomenon has been described for several different bacteria and parasites, suggesting that microbial RNA plays a significant immunoregulatory role in the context of many infectious processes. Thus, beyond the pro-inflammatory capacity of microbial RNA, it seems to be a crucial component in the intricate collection of immune evasion strategies. This review focuses on the different facets of the immune modulating capacity of microbial RNA.
Collapse
|
20
|
Mokuda S, Watanabe H, Kohno H, Ishitoku M, Araki K, Hirata S, Sugiyama E. N1-methylpseudouridine-incorporated mRNA enhances exogenous protein expression and suppresses immunogenicity in primary human fibroblast-like synoviocytes. Cytotechnology 2022; 74:503-514. [PMID: 35791402 PMCID: PMC9245880 DOI: 10.1007/s10616-022-00540-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 06/03/2022] [Indexed: 11/25/2022] Open
Abstract
Studies conducted using murine arthritis models have indicated that the use of in vitro-transcribed messenger RNA (IVT mRNA) is an effective therapeutic approach for joint diseases. However, the use of IVT mRNA in human synovial cells has not been widely studied. Recently, the outbreak of the novel coronavirus disease has accelerated the development of innovative mRNA vaccines, such as those containing a modified nucleic acid, N1-methylpseudouridine-5′-triphosphate (m1ψ). IVT mRNA is an attractive tool for biological experiments and drug discovery. To verify the protein expression from IVT mRNA in vitro, primary cultured fibroblast-like synoviocytes (FLS) and MH7A human synovial fibroblast cells were transfected with enhanced green fluorescent protein (EGFP) mRNA with or without m1ψ incorporation. EGFP was detected using western blotting and fluorescence microscopy. A multiplex assay was performed to comprehensively understand IVT mRNA-induced immunogenicity. Gene expression levels were measured using reverse transcription polymerase chain reaction. In both MH7A cells and FLS, cells transfected with EGFP mRNA containing m1ψ generated higher levels of EGFP than those transfected with unmodified EGFP or control mRNAs. The multiplex assay of the FLS culture supernatant and reverse transcription polymerase chain reaction for FLS revealed that both concentration and expression of IL-6, TNF-α, and CXCL10 were upregulated by unmodified EGFP mRNA, whereas they were suppressed by EGFP mRNA with m1ψ. Overall, m1ψ incorporation enhanced protein expression and decreased the expression of cytokines. These findings may contribute to arthritis research.
Collapse
Affiliation(s)
- Sho Mokuda
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Hirofumi Watanabe
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Hiroki Kohno
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Michinori Ishitoku
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kei Araki
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Shintaro Hirata
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Eiji Sugiyama
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
21
|
Cagliero J, Vernel-Pauillac F, Murray G, Adler B, Matsui M, Werts C. Pathogenic Leptospires Limit Dendritic Cell Activation Through Avoidance of TLR4 and TRIF Signaling. Front Immunol 2022; 13:911778. [PMID: 35812397 PMCID: PMC9258186 DOI: 10.3389/fimmu.2022.911778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Leptospira interrogans is a bacterial species responsible for leptospirosis, a neglected worldwide zoonosis. Mice and rats are resistant and can become asymptomatic carriers, whereas humans and some other mammals may develop severe forms of leptospirosis. Uncommon among spirochetes, leptospires contain lipopolysaccharide (LPS) in their outer membrane. LPS is highly immunogenic and forms the basis for a large number of serovars. Vaccination with inactivated leptospires elicits a protective immunity, restricted to serovars with related LPS. This protection that lasts in mice, is not long lasting in humans and requires annual boosts. Leptospires are stealth pathogens that evade the complement system and some pattern recognition receptors from the Toll-like (TLR) and Nod-Like families, therefore limiting antibacterial defense. In macrophages, leptospires totally escape recognition by human TLR4, and escape the TRIF arm of the mouse TLR4 pathway. However, very little is known about the recognition and processing of leptospires by dendritic cells (DCs), although they are crucial cells linking innate and adaptive immunity. Here we tested the activation of primary DCs derived from human monocytes (MO-DCs) and mouse bone marrow (BM-DCs) 24h after stimulation with saprophytic or different pathogenic virulent or avirulent L. interrogans. We measured by flow cytometry the expression of DC-SIGN, a lectin involved in T-cell activation, co-stimulation molecules and MHC-II markers, and pro- and anti-inflammatory cytokines by ELISA. We found that exposure to leptospires, live or heat-killed, activated dendritic cells. However, pathogenic L. interrogans, especially from the Icterohaemorraghiae Verdun strain, triggered less marker upregulation and less cytokine production than the saprophytic Leptospira biflexa. In addition, we showed a better activation with avirulent leptospires, when compared to the virulent parental strains in murine BM-DCs. We did not observe this difference in human MO-DCs, suggesting a role for TLR4 in DC stimulation. Accordingly, using BM-DCs from transgenic deficient mice, we showed that virulent Icterohaemorraghiae and Manilae serovars dampened DC activation, at least partly, through the TLR4 and TRIF pathways. This work shows a novel bacterial immune evasion mechanism to limit DC activation and further illustrates the role of the leptospiral LPS as a virulence factor.
Collapse
Affiliation(s)
- Julie Cagliero
- Institut Pasteur de Nouvelle-Calédonie, member of the Pasteur Network, Immunity and Inflammation Group (GIMIN), Noumea, New Caledonia
- Institut Pasteur, Université de Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi bactérienne, F-75015 Paris, France
- Institut Pasteur de Nouvelle-Calédonie, member of the Pasteur Network, Leptospirosis Research and Expertise Unit, Noumea, New Caledonia
| | - Frédérique Vernel-Pauillac
- Institut Pasteur, Université de Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi bactérienne, F-75015 Paris, France
| | - Gerald Murray
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Ben Adler
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Mariko Matsui
- Institut Pasteur de Nouvelle-Calédonie, member of the Pasteur Network, Immunity and Inflammation Group (GIMIN), Noumea, New Caledonia
| | - Catherine Werts
- Institut Pasteur, Université de Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi bactérienne, F-75015 Paris, France
- *Correspondence: Catherine Werts,
| |
Collapse
|
22
|
Qureischi M, Mohr J, Arellano-Viera E, Knudsen SE, Vohidov F, Garitano-Trojaola A. mRNA-based therapies: Preclinical and clinical applications. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 372:1-54. [PMID: 36064262 DOI: 10.1016/bs.ircmb.2022.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
At the fundamental level, messenger RNA (mRNA)-based therapeutics involves the delivery of in vitro-transcribed (IVT) mRNA into the cytoplasm of a target cell, where it is translated into the desired protein. IVT mRNA presents various advantages compared to DNA and recombinant protein-based approaches that make it ideal for a broad range of therapeutic applications. IVT mRNA, which is translated in the cytoplasm after transfection into cells, can encode virtually any target protein. Notably, it does not enter the nucleus, which avoids its integration into the genome and the risk of insertional mutagenesis. The large-scale production of IVT mRNA is less complex than production of recombinant proteins, and Good Manufacturing Practice-compliant mRNA production is easily scalable, ideally poising mRNA for not only off-the-shelf, but more personalized treatment approaches. IVT mRNA's safety profile, pharmacokinetics, and pharmacodynamics, including its inherent immunostimulatory capacity, can be optimized for different therapeutic applications by harnessing a wide array of optimized sequence elements, chemical modifications, purification techniques, and delivery methods. The value of IVT mRNA was recently proved during the COVID-19 pandemic when mRNA-based vaccines outperformed the efficacy of established technologies, and millions of doses were rapidly deployed. In this review, we will discuss chemical modifications of IVT mRNA and highlight numerous preclinical and clinical applications including vaccines for cancer and infectious diseases, cancer immunotherapy, protein replacement, gene editing, and cell reprogramming.
Collapse
|
23
|
Salmonella Induces the cGAS-STING-Dependent Type I Interferon Response in Murine Macrophages by Triggering mtDNA Release. mBio 2022; 13:e0363221. [PMID: 35604097 PMCID: PMC9239183 DOI: 10.1128/mbio.03632-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) elicited strong innate immune responses in macrophages. To activate innate immunity, pattern recognition receptors (PRRs) in host cells can recognize highly conserved pathogen-associated molecular patterns (PAMPs). Here, we showed that S. Typhimurium induced a robust type I interferon (IFN) response in murine macrophages. Exposure of macrophages to S. Typhimurium activated a Toll-like receptor 4 (TLR4)-dependent type I IFN response. Next, we showed that type I IFN and IFN-stimulated genes (ISGs) were elicited in a TBK1-IFN-dependent manner. Furthermore, cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) and immune adaptor protein stimulator of interferon genes (STING) were also required for the induction of type I IFN response during infection. Intriguingly, S. Typhimurium infection triggered mitochondrial DNA (mtDNA) release into the cytosol to activate the type I IFN response. In addition, we also showed that bacterial DNA was enriched in cGAS during infection, which may contribute to cGAS activation. Finally, we showed that cGAS and STING deficient mice and cells were more susceptible to S. Typhimurium infection, signifying the critical role of the cGAS-STING pathway in host defense against S. Typhimurium infection. In conclusion, in addition to TLR4-dependent innate immune response, we demonstrated that S. Typhimurium induced the type I IFN response in a cGAS-STING-dependent manner and the S. Typhimurium-induced mtDNA release was important for the induction of type I IFN. This study elucidated a new mechanism by which bacterial pathogen activated the cGAS-STING pathway and also characterized the important role of cGAS-STING during S. Typhimurium infection.
Collapse
|
24
|
Moretti J, Jia B, Hutchins Z, Roy S, Yip H, Wu J, Shan M, Jaffrey SR, Coers J, Blander JM. Caspase-11 interaction with NLRP3 potentiates the noncanonical activation of the NLRP3 inflammasome. Nat Immunol 2022; 23:705-717. [PMID: 35487985 PMCID: PMC9106893 DOI: 10.1038/s41590-022-01192-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 03/18/2022] [Indexed: 11/08/2022]
Abstract
Caspase-11 detection of intracellular lipopolysaccharide (LPS) from invasive Gram-negative bacteria mediates noncanonical activation of the NLRP3 inflammasome. While avirulent bacteria do not invade the cytosol, their presence in tissues necessitates clearance and immune system mobilization. Despite sharing LPS, only live avirulent Gram-negative bacteria activate the NLRP3 inflammasome. Here, we found that bacterial mRNA, which signals bacterial viability, was required alongside LPS for noncanonical activation of the NLRP3 inflammasome in macrophages. Concurrent detection of bacterial RNA by NLRP3 and binding of LPS by pro-caspase-11 mediated a pro-caspase-11-NLRP3 interaction before caspase-11 activation and inflammasome assembly. LPS binding to pro-caspase-11 augmented bacterial mRNA-dependent assembly of the NLRP3 inflammasome, while bacterial viability and an assembled NLRP3 inflammasome were necessary for activation of LPS-bound pro-caspase-11. Thus, the pro-caspase-11-NLRP3 interaction nucleated a scaffold for their interdependent activation explaining their functional reciprocal exclusivity. Our findings inform new vaccine adjuvant combinations and sepsis therapy.
Collapse
Affiliation(s)
- Julien Moretti
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Baosen Jia
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Zachary Hutchins
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Soumit Roy
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- 182 E 95th St., New York, NY, USA
| | - Hilary Yip
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jiahui Wu
- Department of Pharmacology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Meimei Shan
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Patch Biosciences, New York, NY, USA
| | - Samie R Jaffrey
- Department of Pharmacology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - J Magarian Blander
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
25
|
|
26
|
Lentini G, De Gaetano GV, Famà A, Galbo R, Coppolino F, Mancuso G, Teti G, Beninati C. Neutrophils discriminate live from dead bacteria by integrating signals initiated by Fprs and TLRs. EMBO J 2022; 41:e109386. [PMID: 35112724 PMCID: PMC8886525 DOI: 10.15252/embj.2021109386] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/15/2021] [Accepted: 01/12/2022] [Indexed: 11/09/2022] Open
Abstract
The mechanisms whereby neutrophils respond differentially to live and dead organisms are unknown. We show here that neutrophils produce 5- to 30-fold higher levels of the Cxcl2 chemokine in response to live bacteria, compared with killed bacteria or isolated bacterial components, despite producing similar levels of Cxcl1 or pro-inflammatory cytokines. Secretion of high levels of Cxcl2, which potently activates neutrophils by an autocrine mechanism, requires three signals. The first two signals are provided by two different sets of signal peptides released by live bacteria, which selectively activate formylated peptide receptor 1 (Fpr1) and Fpr2, respectively. Signal 3 originates from Toll-like receptor activation by microbial components present in both live and killed bacteria. Mechanistically, these signaling pathways converge at the level of the p38 MAP kinase, leading to activation of the AP-1 transcription factor and to Cxcl2 induction. Collectively, our data demonstrate that the simultaneous presence of agonists for Fpr1, Fpr2, and Toll-like receptors represents a unique signature associated with viable bacteria, which is sensed by neutrophils and induces Cxcl2-dependent autocrine cell activation.
Collapse
Affiliation(s)
- Germana Lentini
- Department of Human PathologyUniversity of MessinaMessinaItaly
| | | | - Agata Famà
- Department of Human PathologyUniversity of MessinaMessinaItaly
| | - Roberta Galbo
- Department of Chemical, Biological and Pharmaceutical SciencesUniversity of MessinaMessinaItaly
| | - Francesco Coppolino
- Department of BiomedicalDental, Morphological and Functional Imaging SciencesUniversity of MessinaMessinaItaly
| | | | | | - Concetta Beninati
- Department of Human PathologyUniversity of MessinaMessinaItaly,Scylla Biotech SrlMessinaItaly
| |
Collapse
|
27
|
Ryder CB, Kondolf HC, O’Keefe ME, Zhou B, Abbott DW. Chemical Modulation of Gasdermin-Mediated Pyroptosis and Therapeutic Potential. J Mol Biol 2022; 434:167183. [PMID: 34358546 PMCID: PMC8810912 DOI: 10.1016/j.jmb.2021.167183] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/24/2021] [Accepted: 07/29/2021] [Indexed: 12/13/2022]
Abstract
Pyroptosis, a lytic form of programmed cell death, both stimulates effective immune responses and causes tissue damage. Gasdermin (GSDM) proteins are a family of pore-forming executors of pyroptosis. While the most-studied member, GSDMD, exerts critical functions in inflammasome biology, emerging evidence demonstrates potential broad relevance for GSDM-mediated pyroptosis across diverse pathologies. In this review, we describe GSDM biology, outline conditions where inflammasomes and GSDM-mediated pyroptosis represent rational therapeutic targets, and delineate strategies to manipulate these central immunologic processes for the treatment of human disease.
Collapse
Affiliation(s)
- Christopher B. Ryder
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA 44106,Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA 44106
| | - Hannah C. Kondolf
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA 44106
| | - Meghan E. O’Keefe
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA 44106
| | - Bowen Zhou
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA 44106
| | - Derek W. Abbott
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA 44106,Corresponding author: ()
| |
Collapse
|
28
|
Ngo QV, Faass L, Sähr A, Hildebrand D, Eigenbrod T, Heeg K, Nurjadi D. Inflammatory Response Against Staphylococcus aureus via Intracellular Sensing of Nucleic Acids in Keratinocytes. Front Immunol 2022; 13:828626. [PMID: 35281009 PMCID: PMC8907419 DOI: 10.3389/fimmu.2022.828626] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/07/2022] [Indexed: 12/02/2022] Open
Abstract
Staphylococcus aureus is one of the clinically most relevant pathogens causing infections. Humans are often exposed to S. aureus. In approximately one-third of the healthy population it can be found on the skin either for long or short periods as colonizing "commensals", without inducing infections or an inflammatory immune response. While tolerating S. aureus seems to be limited to certain individuals and time periods in most cases, Staphylococcus epidermidis is tolerated permanently on the skin of almost all individuals without activating overwhelming skin inflammation. To investigate this, we co-cultured a keratinocyte cell line (HaCaT) with viable S. aureus or S. epidermidis to study the differences in the immune activation. S. aureus activated keratinocytes depicted by a profound IL-6 and IL-8 response, whereas S. epidermidis did not. Our data indicate that internalization of S. aureus and the subsequent intracellular sensing of bacterial nucleic acid may be essential for initiating inflammatory response in keratinocytes. Internalized dsRNA activates IL-6 and IL-8 release, but not TNF-α or IFNs by human keratinocytes. This is a non-specific effect of dsRNA, which can be induced using Poly(I:C), as well as RNA from S. aureus and S. epidermidis. However, only viable S. aureus were able to induce this response as these bacteria and not S. epidermidis were actively internalized by HaCaT. The stimulatory effect of S. aureus seems to be independent of the TLR3, -7 and -8 pathways.
Collapse
Affiliation(s)
- Quang Vinh Ngo
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Department of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Larissa Faass
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
- Max von Pettenkofer Institute, Chair for Medical Microbiology and Hygiene, Ludwig Maximilians University Munich, Munich, Germany
| | - Aline Sähr
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Dagmar Hildebrand
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Tatjana Eigenbrod
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Klaus Heeg
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Department of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Dennis Nurjadi
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Department of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
29
|
Liu N, Pang X, Zhang H, Ji P. The cGAS-STING Pathway in Bacterial Infection and Bacterial Immunity. Front Immunol 2022; 12:814709. [PMID: 35095914 PMCID: PMC8793285 DOI: 10.3389/fimmu.2021.814709] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/21/2021] [Indexed: 12/27/2022] Open
Abstract
Cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) (cGAMP) synthase (cGAS), along with the adaptor stimulator of interferon genes (STING), are crucial components of the innate immune system, and their study has become a research hotspot in recent years. Many biochemical and structural studies that have collectively elucidated the mechanism of activation of the cGAS-STING pathway with atomic resolution have provided insights into the roles of the cGAS-STING pathway in innate immunity and clues to the origin and evolution of the modern cGAS-STING signaling pathway. The cGAS-STING pathway has been identified to protect the host against viral infection. After detecting viral dsDNA, cGAS synthesizes a second messenger to activate STING, eliciting antiviral immune responses by promoting the expression of interferons (IFNs) and hundreds of IFN-stimulated genes (ISGs). Recently, the cGAS-STING pathway has also been found to be involved in response to bacterial infections, including bacterial pneumonia, melioidosis, tuberculosis, and sepsis. However, compared with its functions in viral infection, the cGAS-STING signaling pathway in bacterial infection is more complex and diverse since the protective and detrimental effects of type I IFN (IFN-I) on the host depend on the bacterial species and infection mode. Besides, STING activation can also affect infection prognosis through other mechanisms in different bacterial infections, independent of the IFN-I response. Interestingly, the core protein components of the mammalian cGAS-STING signaling pathway have been found in the bacterial defense system, suggesting that this widespread signaling pathway may have originated in bacteria. Here, we review recent findings related to the structures of major molecules involved in the cGAS-STING pathway and the effects of the cGAS-STING pathway in various bacterial infections and bacterial immunity, which may pave the way for the development of new antibacterial drugs that specifically kill bacteria without harmful effects on the host.
Collapse
Affiliation(s)
- Nanxin Liu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoxiao Pang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Zhang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Ji
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
30
|
Pereira M, Oh JK, Kang DK, Engstrand L, Valeriano VD. Hacking Commensal Bacteria to Consolidate the Adaptive Mucosal Immune Response in the Gut-Lung Axis: Future Possibilities for SARS-CoV-2 Protection. BIOTECH 2022; 11:3. [PMID: 35822811 PMCID: PMC9245903 DOI: 10.3390/biotech11010003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/04/2022] [Accepted: 01/26/2022] [Indexed: 11/17/2022] Open
Abstract
Infectious diseases caused by mucosal pathogens significantly increase mortality and morbidity. Thus, the possibility to target these pathogens at their primary entry points can consolidate protective immunity. Regarding SARS-CoV-2 infection, it has been observed that the upper respiratory mucosa is highly affected and that dysregulation of resident microbiota in the gut-lung axis plays a crucial role in determining symptom severity. Thus, understanding the possibility of eliciting various mucosal and adaptive immune responses allows us to effectively design bacterial mucosal vaccine vectors. Such design requires rationally selecting resident bacterial candidates as potential host carriers, evaluating effective carrier proteins for stimulating an immune response, and combining these two to improve antigenic display and immunogenicity. This review investigated mucosal vaccine vectors from 2015 to present, where a few have started to utilize Salmonella and lactic acid bacteria (LAB) to display SARS-CoV-2 Spike S proteins or fragments. Although current literature is still lacking for its studies beyond in vitro or in vivo efficiency, decades of research into these vectors show promising results. Here, we discuss the mucosal immune systems focusing on the gut-lung axis microbiome and offer new insight into the potential use of alpha streptococci in the upper respiratory tract as a vaccine carrier.
Collapse
Affiliation(s)
- Marcela Pereira
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165 Stockholm, Sweden; (M.P.); (J.K.O.); (L.E.)
| | - Ju Kyoung Oh
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165 Stockholm, Sweden; (M.P.); (J.K.O.); (L.E.)
| | - Dae-Kyung Kang
- Department of Animal Resources Science, Dankook University, Cheonan 31116, Korea;
| | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165 Stockholm, Sweden; (M.P.); (J.K.O.); (L.E.)
| | - Valerie Diane Valeriano
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165 Stockholm, Sweden; (M.P.); (J.K.O.); (L.E.)
| |
Collapse
|
31
|
Sontyana B, Shrivastava R, Battu S, Ghosh S, Mukhopadhyay S. Phagosome maturation and modulation of macrophage effector function by intracellular pathogens: target for therapeutics. Future Microbiol 2021; 17:59-76. [PMID: 34877879 DOI: 10.2217/fmb-2021-0101] [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: 11/21/2022] Open
Abstract
Macrophages are important cells that regulate various innate functions. Macrophages after engulfment of pathogens proceed for phagosome maturation and finally fuse with lysosomes to kill pathogens. Although pathogen degradation is one of the important functions of phagosomes, various immune-effector functions of macrophages are also dependent on the phagosome maturation process. This review discusses signaling processes regulating phagosome maturation as well as various effector functions of macrophages such as apoptosis, antigen presentation, autophagy and inflammasome that are dependent on the phagosome maturation process. It also discusses strategies adopted by various intracellular pathogens to counteract these functions to evade intracellular destruction mechanisms. These studies may give direction for the development of new therapeutics to control various intracellular infections.
Collapse
Affiliation(s)
- Brahmaji Sontyana
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Rohini Shrivastava
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Srikanth Battu
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India
| | - Sudip Ghosh
- Molecular Biology Unit, ICMR-National Institute of Nutrition, Jamai Osmania PO, Hyderabad, 500007, Telangana, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India
| |
Collapse
|
32
|
Madushani KP, Shanaka KASN, Yang H, Lim C, Jeong T, Tharuka MDN, Lee J. Molecular characterization, expression profile, and antiviral activity of redlip mullet (Liza haematocheila) viperin. Comp Biochem Physiol B Biochem Mol Biol 2021; 258:110699. [PMID: 34801710 DOI: 10.1016/j.cbpb.2021.110699] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/24/2021] [Accepted: 11/12/2021] [Indexed: 01/08/2023]
Abstract
Viperin is known to exhibit activity against RNA viral infection. Viral hemorrhagic septicemia virus (VHSV) is a negative-sense single-stranded RNA virus that causes severe loss in aquaculture species. Susceptible species include redlip mullets (Liza haematocheila), which has become an economically important euryhaline mugilid species in offshore aquaculture along the west coast of Korea. Although interferon-stimulated genes are suspected to act against VHSV, specific pathways or mechanisms of these antiviral actions in redlip mullets have not yet been established. In silico studies of the mullet viperin (Lhrsad2) revealed an S-adenosyl methionine binding conserved domain containing the 77CNYKCGFC84 sequence. In the tissue distribution, the highest levels of lhrsad2 expression were observed in the blood. When injected with poly(I:C), an approximately 17-fold upregulation (compared to the control) of viperin was detected in the blood after 24 h. Furthermore, non-viral immune stimuli, including Lactococcus garvieae (L. garvieae) and lipopolysaccharide (LPS), that were injected into redlip mullets were not found to induce considerable levels of viperin expression. Subcellular analysis revealed that Lhrsad2 localized to the endoplasmic reticulum (ER). To investigate Lhrsad2's antiviral effects against VHSV, cells overexpressing lhrsad2 were infected with VHSV, and then the viral titer and viral gene expression were analyzed. Both assays revealed the potential of Lhrsad2 to significantly reduce VHSV transcription and replication. In brief, the current study illustrates the remarkable ability of viperin to weaken VHSV in redlip mullet.
Collapse
Affiliation(s)
- K P Madushani
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province 63333, Republic of Korea
| | - K A S N Shanaka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province 63333, Republic of Korea
| | - Hyerim Yang
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea
| | - Chaehyeon Lim
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea
| | - Taehyug Jeong
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea
| | - M D Neranjan Tharuka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province 63333, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province 63333, Republic of Korea.
| |
Collapse
|
33
|
Greene JT, Brian BF, Senevirathne SE, Freedman TS. Regulation of myeloid-cell activation. Curr Opin Immunol 2021; 73:34-42. [PMID: 34601225 DOI: 10.1016/j.coi.2021.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 12/24/2022]
Abstract
Myeloid cells (macrophages, monocytes, dendritic cells, and granulocytes) survey the body for signs of infection and damage and regulate tissue homeostasis, organogenesis, and immunity. They express receptors that initiate the inflammatory response, send signals that alter the vascular and cytokine milieu, and oversee the recruitment, differentiation, and activation of other myeloid and adaptive immune cells. Their activation must therefore be tightly regulated, optimized for maximal innate-immune protection with a minimum of collateral tissue damage or disorganization. In this review we discuss what it means for myeloid cells to become activated, with emphasis on the receptors and signaling molecules important for the recognition of pathogen-associated and damage-associated molecular patterns. We also outline how these signals are regulated by the steric properties of proteins, by adhesive and cytoskeletal interactions, and by negative feedback to keep inflammation in check and support healthy tissue development and homeostasis. Throughout the text we highlight recent publications and reviews and direct readers therein for a comprehensive bibliography.
Collapse
Affiliation(s)
- Joseph T Greene
- Department of Pharmacology, Center for Immunology, Masonic Cancer Center, and Center for Autoimmune Diseases Research, University of Minnesota, Minneapolis, MN, 55455, United States
| | - Ben F Brian
- Department of Pharmacology, Center for Immunology, Masonic Cancer Center, and Center for Autoimmune Diseases Research, University of Minnesota, Minneapolis, MN, 55455, United States
| | - S Erandika Senevirathne
- Department of Pharmacology, Center for Immunology, Masonic Cancer Center, and Center for Autoimmune Diseases Research, University of Minnesota, Minneapolis, MN, 55455, United States
| | - Tanya S Freedman
- Department of Pharmacology, Center for Immunology, Masonic Cancer Center, and Center for Autoimmune Diseases Research, University of Minnesota, Minneapolis, MN, 55455, United States.
| |
Collapse
|
34
|
Soliman AM, Sim RH, Das S, Mahakkanukrauh P. Therapeutic Targeting of Inflammatory Pathways with Emphasis on NLRP3 Inflammasomes by Natural Products: A Novel Approach for the Treatment of Inflammatory Eye Diseases. Curr Med Chem 2021; 29:2891-2912. [PMID: 34514977 DOI: 10.2174/0929867328666210910154330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/15/2021] [Accepted: 07/23/2021] [Indexed: 11/22/2022]
Abstract
There is an increase in the incidence of inflammatory eye diseases worldwide. Several dysregulated inflammatory pathways, including the NOD-like receptor protein 3 (NLRP3) inflammasome, have been reported to contribute significantly to the pathogenesis and progression of ophthalmic diseases. Although the available allopathic/conventional medicine has demonstrated effectiveness in managing eye diseases, there is an ongoing global demand for alternative therapeutics with minimal adverse drug reactions, easy availability, increase in patient-compliance, and better disease outcome. Therefore, several studies are investigating the utilization of natural products and herbal formulations in impeding inflammatory pathways, including the NLRP3 inflammasome, in order to prevent or manage eye diseases. In the present review, we highlight the recently reported inflammatory pathways with special emphasis on NLRP3 Inflammasomes involved in the development of eye diseases. Furthermore, we present a variety of natural products and phytochemicals that were reported to interfere with these pathways and their underlying mechanisms of action. These natural products represent potential therapeutic applications for the treatment of several inflammatory eye diseases.
Collapse
Affiliation(s)
- Amro M Soliman
- Department of Biological Sciences-Physiology, Cell and Developmental Biology, University of Alberta, Edmonton, AB T6G 2R3. Canada
| | - Ru Hui Sim
- Tanglin Health Clinic, 50480 Kuala Lumpu. Malaysia
| | - Srijit Das
- Department of Human & Clinical Anatomy, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat. Oman
| | - Pasuk Mahakkanukrauh
- Department of Anatomy & Excellence Center of Osteology Research and Training, Cadaveric Surgical and Training Center, Chiang Mai University. Thailand
| |
Collapse
|
35
|
Cappelletti M, Presicce P, Feiyang M, Senthamaraikannan P, Miller LA, Pellegrini M, Sim MS, Jobe AH, Divanovic S, Way SS, Chougnet CA, Kallapur SG. The induction of preterm labor in rhesus macaques is determined by the strength of immune response to intrauterine infection. PLoS Biol 2021; 19:e3001385. [PMID: 34495952 PMCID: PMC8452070 DOI: 10.1371/journal.pbio.3001385] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 09/20/2021] [Accepted: 08/04/2021] [Indexed: 12/18/2022] Open
Abstract
Intrauterine infection/inflammation (IUI) is a major contributor to preterm labor (PTL). However, IUI does not invariably cause PTL. We hypothesized that quantitative and qualitative differences in immune response exist in subjects with or without PTL. To define the triggers for PTL, we developed rhesus macaque models of IUI driven by lipopolysaccharide (LPS) or live Escherichia coli. PTL did not occur in LPS challenged rhesus macaques, while E. coli–infected animals frequently delivered preterm. Although LPS and live E. coli both caused immune cell infiltration, E. coli–infected animals showed higher levels of inflammatory mediators, particularly interleukin 6 (IL-6) and prostaglandins, in the chorioamnion-decidua and amniotic fluid (AF). Neutrophil infiltration in the chorio-decidua was a common feature to both LPS and E. coli. However, neutrophilic infiltration and IL6 and PTGS2 expression in the amnion was specifically induced by live E. coli. RNA sequencing (RNA-seq) analysis of fetal membranes revealed that specific pathways involved in augmentation of inflammation including type I interferon (IFN) response, chemotaxis, sumoylation, and iron homeostasis were up-regulated in the E. coli group compared to the LPS group. Our data suggest that the intensity of the host immune response to IUI may determine susceptibility to PTL.
Collapse
Affiliation(s)
- Monica Cappelletti
- Divisions of Neonatology and Developmental Biology, UCLA Mattel Children’s Hospital, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, UCLA Mattel Children’s Hospital, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Ma Feiyang
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- Institute for Quantitative and Computational Biosciences–Collaboratory, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Paranthaman Senthamaraikannan
- Division of Neonatology and Pulmonary Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Lisa A. Miller
- California National Primate Research Center, University of California Davis, Davis, California, United States of America
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- Institute for Quantitative and Computational Biosciences–Collaboratory, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Myung S. Sim
- Department of Medicine Statistics Core, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Alan H. Jobe
- Division of Neonatology and Pulmonary Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Senad Divanovic
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Sing Sing Way
- Infectious Diseases, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Claire A. Chougnet
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Suhas G. Kallapur
- Divisions of Neonatology and Developmental Biology, UCLA Mattel Children’s Hospital, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
36
|
Verdecia M, Kokai-Kun JF, Kibbey M, Acharya S, Venema J, Atouf F. COVID-19 vaccine platforms: Delivering on a promise? Hum Vaccin Immunother 2021; 17:2873-2893. [PMID: 34033528 PMCID: PMC8381795 DOI: 10.1080/21645515.2021.1911204] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/24/2021] [Indexed: 12/13/2022] Open
Abstract
The emergence of the novel SARS-CoV-2 and COVID-19 has brought into sharp focus the need for a vaccine to prevent this disease. Vaccines have saved millions of lives since their introduction to the public over 200 years ago. The potential for vaccination reached new heights in the mid-20th century with the development of technologies that expanded the ability to create novel vaccines. Since then, there has been continued technological advancement in vaccine development. The resulting platforms provide the promise for solutions for many infectious diseases, including those that have been with us for decades as well as those just now emerging. Each vaccine platform represents a different technology with a unique set of advantages and challenges, especially when considering manufacturing. Therefore, it is essential to understand each platform as a separate product and process with its specific quality considerations. This review outlines the relevant platforms for developing a vaccine for SARS-CoV-2 and discusses the advantages and disadvantages of each.
Collapse
Affiliation(s)
- Mark Verdecia
- United States Pharmacopeial Convention, Rockville, MD, USA
| | | | - Maura Kibbey
- United States Pharmacopeial Convention, Rockville, MD, USA
| | - Sarita Acharya
- United States Pharmacopeial Convention, Rockville, MD, USA
| | - Jaap Venema
- United States Pharmacopeial Convention, Rockville, MD, USA
| | - Fouad Atouf
- United States Pharmacopeial Convention, Rockville, MD, USA
| |
Collapse
|
37
|
Yokota M, Häffner N, Kassier M, Brunner M, Shambat SM, Brennecke F, Schniering J, Marques Maggio E, Distler O, Zinkernagel AS, Maurer B. Staphylococcus aureus impairs dermal fibroblast functions with deleterious effects on wound healing. FASEB J 2021; 35:e21695. [PMID: 34160101 DOI: 10.1096/fj.201902836r] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/16/2021] [Accepted: 05/10/2021] [Indexed: 12/28/2022]
Abstract
Chronic wounds are a major disease burden worldwide. The breach of the epithelial barrier facilitates transition of skin commensals to invasive facultative pathogens. Therefore, we investigated the potential effects of Staphylococcus aureus (SA) on dermal fibroblasts as key cells for tissue repair. In co-culture systems combining live or heat-killed SA with dermal fibroblasts derived from the BJ-5ta cell line, healthy individuals, and patients with systemic sclerosis, we assessed tissue repair including pro-inflammatory cytokines, matrix metalloproteases (MMPs), myofibroblast functions, and host defense responses. Only live SA induced the upregulation of IL-1β/-6/-8 and MMP1/3 as co-factors of tissue degradation. Additionally, the increased cell death reduced collagen production, proliferation, migration, and contractility, prerequisite mechanisms for wound closure. Intracellular SA triggered inflammatory and type I IFN responses via intracellular dsDNA sensor molecules and MyD88 and STING signaling pathways. In conclusion, live SA affected various key tissue repair functions of dermal fibroblasts from different sources to a similar extent. Thus, SA infection of dermal fibroblasts should be taken into account for future wound management strategies.
Collapse
Affiliation(s)
- Masaya Yokota
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland.,Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Nicola Häffner
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthew Kassier
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Matthias Brunner
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Srikanth Mairpady Shambat
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Fabian Brennecke
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Janine Schniering
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Ewerton Marques Maggio
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Oliver Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Annelies Sophie Zinkernagel
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Britta Maurer
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland.,Department of Rheumatology and Immunology, University Hospital Bern, University Bern, Bern, Switzerland
| |
Collapse
|
38
|
van Dülmen M, Muthmann N, Rentmeister A. Chemo-Enzymatic Modification of the 5' Cap Maintains Translation and Increases Immunogenic Properties of mRNA. Angew Chem Int Ed Engl 2021; 60:13280-13286. [PMID: 33751748 PMCID: PMC8250829 DOI: 10.1002/anie.202100352] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Indexed: 12/19/2022]
Abstract
Eukaryotic mRNAs are emerging modalities for protein replacement therapy and vaccination. Their 5' cap is important for mRNA translation and immune response and can be naturally methylated at different positions by S-adenosyl-l-methionine (AdoMet)-dependent methyltransferases (MTases). We report on the cosubstrate scope of the MTase CAPAM responsible for methylation at the N6 -position of adenosine start nucleotides using synthetic AdoMet analogs. The chemo-enzymatic propargylation enabled production of site-specifically modified reporter-mRNAs. These cap-propargylated mRNAs were efficiently translated and showed ≈3-fold increased immune response in human cells. The same effects were observed when the receptor binding domain (RBD) of SARS-CoV-2-a currently tested epitope for mRNA vaccination-was used. Site-specific chemo-enzymatic modification of eukaryotic mRNA may thus be a suitable strategy to modulate translation and immune response of mRNAs for future therapeutic applications.
Collapse
Affiliation(s)
- Melissa van Dülmen
- Department of Chemistry and PharmacyInstitute of BiochemistryCorrensstrasse 3648149MünsterGermany
| | - Nils Muthmann
- Department of Chemistry and PharmacyInstitute of BiochemistryCorrensstrasse 3648149MünsterGermany
| | - Andrea Rentmeister
- Department of Chemistry and PharmacyInstitute of BiochemistryCorrensstrasse 3648149MünsterGermany
- Cells in Motion Interfaculty CenterUniversity of MünsterGermany
| |
Collapse
|
39
|
Dülmen M, Muthmann N, Rentmeister A. Eine chemo‐enzymatische Modifizierung der 5′‐Kappe erhält die Translation und erhöht die Immunogenität der mRNA. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202100352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Melissa Dülmen
- Fachbereich Chemie und Pharmazie Institut für Biochemie Corrensstrasse 36 48149 Münster Deutschland
| | - Nils Muthmann
- Fachbereich Chemie und Pharmazie Institut für Biochemie Corrensstrasse 36 48149 Münster Deutschland
| | - Andrea Rentmeister
- Fachbereich Chemie und Pharmazie Institut für Biochemie Corrensstrasse 36 48149 Münster Deutschland
- Cells in Motion Interfaculty Center Westfälische Wilhelms-Universität Münster Deutschland
| |
Collapse
|
40
|
Elagan SK, Almalki SJ, Alharthi MR, Mohamed MS, EL-Badawy MF. Role of Bacteria in the Incidence of Common GIT Cancers: The Dialectical Role of Integrated Bacterial DNA in Human Carcinogenesis. Infect Drug Resist 2021; 14:2003-2014. [PMID: 34103947 PMCID: PMC8179827 DOI: 10.2147/idr.s309051] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
Despite the wide medical knowledge about the direct role of many viruses in the pathogenesis of certain cancers, there is still ambiguity and hazy vision about the direct role of bacteria in cancer incidence. Understanding the role of bacteria in carcinogenesis is no longer a scientific luxury, but it has become an urgent and extremely important necessity to realize the pathogenesis of cancer caused by oncogenic bacteria as an attempt to overcome the oncogenic mechanisms exhibited by these oncogenic bacteria. This review shed the light on the indirect role of the host's inflammatory and immunological responses in the pathogenesis of bacteria-induced cancer. Also, this review discussed the indirect role of the bacterial toxins and virulence factors in the induction of common gastrointestinal cancers, such as gallbladder cancer (GBC), colorectal cancer (CRC), and gastric cancer (GC). Finally, this review dealt with the debate about the possibility of bacterial DNA integration into the human genome and cancer incidence.
Collapse
Affiliation(s)
- Sayed K Elagan
- Department of Mathematics and Statistics, College of Science, Taif University, Taif, 21944, Saudi Arabia
| | - Saad J Almalki
- Department of Mathematics and Statistics, College of Science, Taif University, Taif, 21944, Saudi Arabia
| | - M R Alharthi
- Department of Mathematics and Statistics, College of Science, Taif University, Taif, 21944, Saudi Arabia
| | - Mohamed S Mohamed
- Department of Mathematics and Statistics, College of Science, Taif University, Taif, 21944, Saudi Arabia
| | - Mohamed F EL-Badawy
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Sadat City, Sadat City, 32958, Egypt
| |
Collapse
|
41
|
Schmerer N, Schulte LN. Long noncoding RNAs in bacterial infection. WILEY INTERDISCIPLINARY REVIEWS-RNA 2021; 12:e1664. [PMID: 33989449 DOI: 10.1002/wrna.1664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 11/10/2022]
Abstract
Infectious and inflammatory diseases remain major causes of mortality and morbidity worldwide. To combat bacterial infections, the mammalian immune system employs a myriad of regulators, which secure the effective initiation of inflammatory responses while preventing pathologies due to overshooting immunity. Recently, the human genome has been shown to be pervasively transcribed and to generate thousands of still poorly characterized long noncoding RNAs (lncRNAs). A growing body of literature suggests that lncRNAs play important roles in the regulatory circuitries controlling innate and adaptive immune responses to bacterial pathogens. This review provides an overview of the roles of lncRNAs in the interaction of human and rodent host cells with bacterial pathogens. Further decoding of the lncRNA networks that underlie pathological inflammation and immune subversion could provide new insights into the host cell mechanisms and microbial strategies that determine the outcome of bacterial infections. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications.
Collapse
Affiliation(s)
- Nils Schmerer
- Institute for Lung Research, Philipps-University, Marburg, Germany
| | - Leon N Schulte
- Institute for Lung Research, Philipps-University, Marburg, Germany.,German Center for Lung Research, Giessen, Germany
| |
Collapse
|
42
|
Cole J, Angyal A, Emes RD, Mitchell TJ, Dickman MJ, Dockrell DH. Pneumolysin Is Responsible for Differential Gene Expression and Modifications in the Epigenetic Landscape of Primary Monocyte Derived Macrophages. Front Immunol 2021; 12:573266. [PMID: 34046027 PMCID: PMC8145618 DOI: 10.3389/fimmu.2021.573266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 04/16/2021] [Indexed: 12/17/2022] Open
Abstract
Epigenetic modifications regulate gene expression in the host response to a diverse range of pathogens. The extent and consequences of epigenetic modification during macrophage responses to Streptococcus pneumoniae, and the role of pneumolysin, a key Streptococcus pneumoniae virulence factor, in influencing these responses, are currently unknown. To investigate this, we infected human monocyte derived macrophages (MDMs) with Streptococcus pneumoniae and addressed whether pneumolysin altered the epigenetic landscape and the associated acute macrophage transcriptional response using a combined transcriptomic and proteomic approach. Transcriptomic analysis identified 503 genes that were differentially expressed in a pneumolysin-dependent manner in these samples. Pathway analysis highlighted the involvement of transcriptional responses to core innate responses to pneumococci including modules associated with metabolic pathways activated in response to infection, oxidative stress responses and NFκB, NOD-like receptor and TNF signalling pathways. Quantitative proteomic analysis confirmed pneumolysin-regulated protein expression, early after bacterial challenge, in representative transcriptional modules associated with innate immune responses. In parallel, quantitative mass spectrometry identified global changes in the relative abundance of histone post translational modifications (PTMs) upon pneumococcal challenge. We identified an increase in the relative abundance of H3K4me1, H4K16ac and a decrease in H3K9me2 and H3K79me2 in a PLY-dependent fashion. We confirmed that pneumolysin blunted early transcriptional responses involving TNF-α and IL-6 expression. Vorinostat, a histone deacetylase inhibitor, similarly downregulated TNF-α production, reprising the pattern observed with pneumolysin. In conclusion, widespread changes in the macrophage transcriptional response are regulated by pneumolysin and are associated with global changes in histone PTMs. Modulating histone PTMs can reverse pneumolysin-associated transcriptional changes influencing innate immune responses, suggesting that epigenetic modification by pneumolysin plays a role in dampening the innate responses to pneumococci.
Collapse
Affiliation(s)
- Joby Cole
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield, Sheffield, United Kingdom
- Sheffield Teaching Hospitals NHS FT, Sheffield, United Kingdom
- The Florey Institute, University of Sheffield, Sheffield, United Kingdom
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Adrienn Angyal
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield, Sheffield, United Kingdom
| | - Richard D. Emes
- Advanced Data Analysis Centre, University of Nottingham, Nottingham, United Kingdom
- School of Veterinary Medicine and Science University of Nottingham, Nottingham, United Kingdom
| | - Tim John Mitchell
- Institute of Microbiology and Infection, University of Birmingham, Edinburgh, United Kingdom
| | - Mark J. Dickman
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, United Kingdom
| | - David H. Dockrell
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
43
|
Homeostatic regulation of T follicular helper and antibody response to particle antigens by IL-1Ra of medullary sinus macrophage origin. Proc Natl Acad Sci U S A 2021; 118:2019798118. [PMID: 33875594 DOI: 10.1073/pnas.2019798118] [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: 11/18/2022] Open
Abstract
Hepatitis B virus (HBV) vaccines are composed of surface antigen HBsAg that spontaneously assembles into subviral particles. Factors that impede its humoral immunity in 5% to 10% of vaccinees remain elusive. Here, we showed that the low-level interleukin-1 receptor antagonist (IL-1Ra) can predict antibody protection both in mice and humans. Mechanistically, murine IL-1Ra-inhibited T follicular helper (Tfh) cell expansion and subsequent germinal center (GC)-dependent humoral immunity, resulting in significantly weakened protection against the HBV challenge. Compared to soluble antigens, HBsAg particle antigen displayed a unique capture/uptake and innate immune activation, including IL-1Ra expression, preferably of medullary sinus macrophages. In humans, a unique polymorphism in the RelA/p65 binding site of IL-1Ra enhancer associated IL-1Ra levels with ethnicity-dependent vaccination outcome. Therefore, the differential IL-1Ra response to particle antigens probably creates a suppressive milieu for Tfh/GC development, and neutralization of IL-1Ra would resurrect antibody response in HBV vaccine nonresponders.
Collapse
|
44
|
Development of epithelial cholinergic chemosensory cells of the urethra and trachea of mice. Cell Tissue Res 2021; 385:21-35. [PMID: 33616728 PMCID: PMC8270884 DOI: 10.1007/s00441-021-03424-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 01/24/2021] [Indexed: 12/24/2022]
Abstract
Cholinergic chemosensory cells (CCC) are infrequent epithelial cells with immunosensor function, positioned in mucosal epithelia preferentially near body entry sites in mammals including man. Given their adaptive capacity in response to infection and their role in combatting pathogens, we here addressed the time points of their initial emergence as well as their postnatal development from first exposure to environmental microbiota (i.e., birth) to adulthood in urethra and trachea, utilizing choline acetyltransferase (ChAT)-eGFP reporter mice, mice with genetic deletion of MyD88, toll-like receptor-2 (TLR2), TLR4, TLR2/TLR4, and germ-free mice. Appearance of CCC differs between the investigated organs. CCC of the trachea emerge during embryonic development at E18 and expand further after birth. Urethral CCC show gender diversity and appear first at P6-P10 in male and at P11-P20 in female mice. Urethrae and tracheae of MyD88- and TLR-deficient mice showed significantly fewer CCC in all four investigated deficient strains, with the effect being most prominent in the urethra. In germ-free mice, however, CCC numbers were not reduced, indicating that TLR2/4-MyD88 signaling, but not vita-PAMPs, governs CCC development. Collectively, our data show a marked postnatal expansion of CCC populations with distinct organ-specific features, including the relative impact of TLR2/4-MyD88 signaling. Strong dependency on this pathway (urethra) correlates with absence of CCC at birth and gender-specific initial development and expansion dynamics, whereas moderate dependency (trachea) coincides with presence of first CCC at E18 and sex-independent further development.
Collapse
|
45
|
Galen JE, Wahid R, Buskirk AD. Strategies for Enhancement of Live-Attenuated Salmonella-Based Carrier Vaccine Immunogenicity. Vaccines (Basel) 2021; 9:162. [PMID: 33671124 PMCID: PMC7923097 DOI: 10.3390/vaccines9020162] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/11/2021] [Accepted: 02/15/2021] [Indexed: 12/28/2022] Open
Abstract
The use of live-attenuated bacterial vaccines as carriers for the mucosal delivery of foreign antigens to stimulate the mucosal immune system was first proposed over three decades ago. This novel strategy aimed to induce immunity against at least two distinct pathogens using a single bivalent carrier vaccine. It was first tested using a live-attenuated Salmonella enterica serovar Typhi strain in clinical trials in 1984, with excellent humoral immune responses against the carrier strain but only modest responses elicited against the foreign antigen. Since then, clinical trials with additional Salmonella-based carrier vaccines have been conducted. As with the original trial, only modest foreign antigen-specific immunity was achieved in most cases, despite the incorporation of incremental improvements in antigen expression technologies and carrier design over the years. In this review, we will attempt to deconstruct carrier vaccine immunogenicity in humans by examining the basis of bacterial immunity in the human gastrointestinal tract and how the gut detects and responds to pathogens versus benign commensal organisms. Carrier vaccine design will then be explored to determine the feasibility of retaining as many characteristics of a pathogen as possible to elicit robust carrier and foreign antigen-specific immunity, while avoiding over-stimulation of unacceptably reactogenic inflammatory responses.
Collapse
Affiliation(s)
- James E. Galen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Rezwanul Wahid
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Amanda D. Buskirk
- Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Process and Facilities, Division of Microbiology Assessment II, U.S. Food and Drug Administration, Silver Spring, MD 20903, USA;
| |
Collapse
|
46
|
Agnew A, Nulty C, Creagh EM. Regulation, Activation and Function of Caspase-11 during Health and Disease. Int J Mol Sci 2021; 22:ijms22041506. [PMID: 33546173 PMCID: PMC7913190 DOI: 10.3390/ijms22041506] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 02/04/2023] Open
Abstract
Caspase-11 is a pro-inflammatory enzyme that is stringently regulated during its expression and activation. As caspase-11 is not constitutively expressed in cells, it requires a priming step for its upregulation, which occurs following the stimulation of pathogen and cytokine receptors. Once expressed, caspase-11 activation is triggered by its interaction with lipopolysaccharide (LPS) from Gram-negative bacteria. Being an initiator caspase, activated caspase-11 functions primarily through its cleavage of key substrates. Gasdermin D (GSDMD) is the primary substrate of caspase-11, and the GSDMD cleavage fragment generated is responsible for the inflammatory form of cell death, pyroptosis, via its formation of pores in the plasma membrane. Thus, caspase-11 functions as an intracellular sensor for LPS and an immune effector. This review provides an overview of caspase-11—describing its structure and the transcriptional mechanisms that govern its expression, in addition to its activation, which is reported to be regulated by factors such as guanylate-binding proteins (GBPs), high mobility group box 1 (HMGB1) protein, and oxidized phospholipids. We also discuss the functional outcomes of caspase-11 activation, which include the non-canonical inflammasome, modulation of actin dynamics, and the initiation of blood coagulation, highlighting the importance of inflammatory caspase-11 during infection and disease.
Collapse
|
47
|
Fang X, Wang Y, Zhang Y, Li Y, Kwak-Kim J, Wu L. NLRP3 Inflammasome and Its Critical Role in Gynecological Disorders and Obstetrical Complications. Front Immunol 2021; 11:555826. [PMID: 33584639 PMCID: PMC7876052 DOI: 10.3389/fimmu.2020.555826] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
Inflammasomes, intracellular, multimeric protein complexes, are assembled when damage signals stimulate nucleotide-binding oligomerization domain receptors (NLRs). Several inflammasomes have been reported, including the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3), NLRP1, NLRP7, ice protease-activating factor (IPAF), absent in melanoma 2 (AIM2) and NLR family CARD domain-containing protein 4 (NLRC4). Among these inflammasomes, the NLRP3 inflammasome is the most well-studied in terms of structure and function. Unlike other inflammasomes that can only be activated by a finite number of pathogenic microorganisms, the NLRP3 inflammasome can be activated by the imbalance of the internal environment and a large number of metabolites. The biochemical function of NLRP3 inflammasome is to activate cysteine-requiring aspartate proteinase-1 (caspase-1), which converts pro-IL-1β and pro-IL-18 into their active forms, namely, IL-1β and IL-18, which are then released into the extracellular space. The well-established, classic role of NLRP3 inflammasome has been implicated in many disorders. In this review, we discuss the current understanding of NLRP3 inflammasome and its critical role in gynecological disorders and obstetrical complications.
Collapse
Affiliation(s)
- Xuhui Fang
- Center for Reproductive Medicine, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, China
| | - Yanshi Wang
- Center for Reproductive Medicine, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, China
| | - Yu Zhang
- Center for Reproductive Medicine, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, China
| | - Yelin Li
- Center for Reproductive Medicine, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, China
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL, United States.,Center for Cancer Cell Biology, Immunology and Infection Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Li Wu
- Center for Reproductive Medicine, Anhui Provincial Hospital affiliated to Anhui Medical University, Hefei, China
| |
Collapse
|
48
|
Kitsios GD, Yang H, Yang L, Qin S, Fitch A, Wang XH, Fair K, Evankovich J, Bain W, Shah F, Li K, Methé B, Benos PV, Morris A, McVerry BJ. Respiratory Tract Dysbiosis Is Associated with Worse Outcomes in Mechanically Ventilated Patients. Am J Respir Crit Care Med 2021; 202:1666-1677. [PMID: 32717152 DOI: 10.1164/rccm.201912-2441oc] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Rationale: Host inflammatory responses have been strongly associated with adverse outcomes in critically ill patients, but the biologic underpinnings of such heterogeneous responses have not been defined.Objectives: We examined whether respiratory tract microbiome profiles are associated with host inflammation and clinical outcomes of acute respiratory failure.Methods: We collected oral swabs, endotracheal aspirates (ETAs), and plasma samples from mechanically ventilated patients. We performed 16S ribosomal RNA gene sequencing to characterize upper and lower respiratory tract microbiota and classified patients into host-response subphenotypes on the basis of clinical variables and plasma biomarkers of innate immunity and inflammation. We derived diversity metrics and composition clusters with Dirichlet multinomial models and examined our data for associations with subphenotypes and clinical outcomes.Measurements and Main Results: Oral and ETA microbial communities from 301 mechanically ventilated subjects had substantial heterogeneity in α and β diversity. Dirichlet multinomial models revealed a cluster with low α diversity and enrichment for pathogens (e.g., high Staphylococcus or Pseudomonadaceae relative abundance) in 35% of ETA samples, associated with a hyperinflammatory subphenotype, worse 30-day survival, and longer time to liberation from mechanical ventilation (adjusted P < 0.05), compared with patients with higher α diversity and relative abundance of typical oral microbiota. Patients with evidence of dysbiosis (low α diversity and low relative abundance of "protective" oral-origin commensal bacteria) in both oral and ETA samples (17%, combined dysbiosis) had significantly worse 30-day survival and longer time to liberation from mechanical ventilation than patients without dysbiosis (55%; adjusted P < 0.05).Conclusions: Respiratory tract dysbiosis may represent an important, modifiable contributor to patient-level heterogeneity in systemic inflammatory responses and clinical outcomes.
Collapse
Affiliation(s)
- Georgios D Kitsios
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine and University of Pittsburgh Medical Center.,Center for Medicine and the Microbiome
| | - Haopu Yang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine and University of Pittsburgh Medical Center.,Department of Computational and Systems Biology, and.,Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Libing Yang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine and University of Pittsburgh Medical Center.,Center for Medicine and the Microbiome.,Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shulin Qin
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine and University of Pittsburgh Medical Center.,Center for Medicine and the Microbiome
| | | | - Xiao-Hong Wang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine and University of Pittsburgh Medical Center
| | - Katherine Fair
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine and University of Pittsburgh Medical Center
| | - John Evankovich
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine and University of Pittsburgh Medical Center
| | - William Bain
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine and University of Pittsburgh Medical Center
| | - Faraaz Shah
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine and University of Pittsburgh Medical Center.,School of Medicine, Tsinghua University, Beijing, China; and
| | - Kelvin Li
- Center for Medicine and the Microbiome
| | - Barbara Methé
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine and University of Pittsburgh Medical Center.,Center for Medicine and the Microbiome
| | | | - Alison Morris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine and University of Pittsburgh Medical Center.,Center for Medicine and the Microbiome.,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Bryan J McVerry
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine and University of Pittsburgh Medical Center.,Center for Medicine and the Microbiome
| |
Collapse
|
49
|
Hamanaka G, Chung KK, Arai K. Do phagocytotic mechanisms regulate soluble factor secretion in microglia? Neural Regen Res 2021; 16:974-975. [PMID: 33229739 PMCID: PMC8178763 DOI: 10.4103/1673-5374.297067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Gen Hamanaka
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Kelly K Chung
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
50
|
Chudzik A, Paściak M. Bacterial extracellular vesicles as cell-cell communication
mediators. POSTEP HIG MED DOSW 2020. [DOI: 10.5604/01.3001.0014.6165] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Extracellular vesicles constitute a heterogeneous group of nanoparticles, released by both prokaryotic
and eukaryotic cells, which perform various biological functions and participate in cell-cell
communication. Bacterial extracellular vesicles are made of lipids, proteins and nucleic acids.
There are a number of hypotheses for the formation of extracellular vesicles, but the mechanisms
of biogenesis of these structures remain unclear. Hardly soluble metabolites or signaling molecules,
DNA and RNA are vesicles cargo. Extracellular vesicles have a protective function, they can
eliminate other bacterial cells and participate in horizontal gene transfer. The enzymes contained
inside the vesicles facilitate the acquisition of nutrients and help colonize various ecological niches.
Signal molecules carried in the vesicles enable biofilm formation. In the secreted extracellular
vesicles pathogenic microorganisms carry virulence factors, including toxins, into the host cells.
Via vesicles, bacteria can also modulate the host immune system. Bacterial extracellular vesicles
are promising vaccine candidates and can be used as drug carriers. The review discusses the current
knowledge concerning biogenesis, composition, preparation methods, physiological functions
and potential applications of extracellular vesicles secreted by prokaryotic cells.
Collapse
Affiliation(s)
- Anna Chudzik
- Instytut Immunologii i Terapii Doświadczalnej im. Ludwika Hirszfelda Polskiej Akademii Nauk, Wrocław
| | - Mariola Paściak
- Instytut Immunologii i Terapii Doświadczalnej im. Ludwika Hirszfelda Polskiej Akademii Nauk, Wrocław
| |
Collapse
|