1
|
Meerschaert KA, Chiu IM. The gut-brain axis and pain signalling mechanisms in the gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2025; 22:206-221. [PMID: 39578592 DOI: 10.1038/s41575-024-01017-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
Visceral pain is a major clinical problem and one of the most common reasons patients with gastrointestinal disorders seek medical help. Peripheral sensory neurons that innervate the gut can detect noxious stimuli and send signals to the central nervous system that are perceived as pain. There is a bidirectional communication network between the gastrointestinal tract and the nervous system that mediates pain through the gut-brain axis. Sensory neurons detect mechanical and chemical stimuli within the intestinal tissues, and receive signals from immune cells, epithelial cells and the gut microbiota, which results in peripheral sensitization and visceral pain. This Review focuses on molecular communication between these non-neuronal cell types and neurons in visceral pain. These bidirectional interactions can be dysregulated during gastrointestinal diseases to exacerbate visceral pain. We outline the anatomical pathways involved in pain processing in the gut and how cell-cell communication is integrated into this gut-brain axis. Understanding how bidirectional communication between the gut and nervous system is altered during disease could provide new therapeutic targets for treating visceral pain.
Collapse
Affiliation(s)
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Liang Y, Adamson C, Feng S, Qiao Y. Exploring the Impact of Amidation Status in Meso-Diaminopimelic-Acid-Containing Disaccharide Peptidoglycan Fragments on Host Innate Immune Activation. ACS Chem Biol 2025; 20:69-76. [PMID: 39749870 DOI: 10.1021/acschembio.4c00700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Bacterial peptidoglycan, the essential cell surface polymer that protects bacterial integrity, also serves as the molecular pattern recognized by the host's innate immune system. Although the minimal motifs of bacterial peptidoglycan fragments (PGNs) that activate mammalian NOD1 and NOD2 sensors are well-known and often represented by small canonical ligands, the immunostimulatory effects of natural PGNs, which are structurally more complex and potentially can simultaneously activate both the NOD1 and NOD2 signaling pathways in hosts, have not been comprehensively investigated. In particular, many bacteria incorporate additional structural modifications in peptidoglycans to evade host immune surveillance, resulting in diverse structural variations among natural PGNs that may influence their biological effects in hosts. The focus of this study is on the amidation status of γ-d-glutamic acid and meso-diaminopimelic acid (mDAP) at the second and third positions of stem peptides in peptidoglycan, which represent key structural features that vary across different bacterial species. With four synthetic mDAP-containing disaccharide PGNs of different amidation states, we systematically investigated their structure-activity relationship in stimulating host innate immune responses in vitro. Our findings revealed that the amidation of disaccharide PGNs has distinct effects on NOD1 and NOD2 induction, along with their differential immunostimulatory activities in macrophage cells. Additionally, we found that, like the canonical NOD2 ligand, natural PGNs confer immune tolerance to LPS, and amidation states do not affect this outcome. Overall, our work highlights the potential immunological implications of these differentially amidated mDAP-type disaccharide PGNs in host-microbe crosstalk.
Collapse
Affiliation(s)
- Yaquan Liang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637371, Singapore
| | - Christopher Adamson
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637371, Singapore
| | - Shiliu Feng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637371, Singapore
| | - Yuan Qiao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637371, Singapore
| |
Collapse
|
3
|
Guo S, Zeng M, Zhang C, Fan Y, Ran M, Song Z. Genome-wide characterization and comparative expression profiling of dual-specificity phosphatase genes in yellow catfish ( Pelteobagrus fulvidraco) after infection with exogenous Aeromonas hydrophila. Front Immunol 2024; 15:1481696. [PMID: 39606227 PMCID: PMC11598348 DOI: 10.3389/fimmu.2024.1481696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/15/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction Dual-specificity phosphatases (DUSPs) are crucial regulators in many mammals, managing dephosphorylation and inactivation of mitogen-activated protein kinases (MAPKs) and playing essential roles in immune responses. However, their presence and functions in teleosts, like the yellow catfish (Pelteobagrus fulvidraco), remain unexplored. Methods In this study, eight pfDusp genes (pfDusp1-7 and pfDusp10) were identified in yellow catfish. We characterized their molecular features, conserved protein sequences, and chromosomal localization through genome-wide analyses, and we examined their expression patterns in immune responses. Results Our findings reveal two conserved motifs, Leu-Phe-Leu-Gly and Ala-Tyr-Leu-Met, within the DSPc domain of DUSP proteins. The genes were mapped across seven chromosomes without evidence of duplication. Comparative analysis showed high conservation of Dusp genes across vertebrates, with evolutionary analysis suggesting Dusp3 as a potential intermediate form. Dusp transcripts were significantly upregulated in the kidney post-A. hydrophila infection. Discussion These results suggest the involvement of Dusp genes in the immune response of yellow catfish to bacterial pathogens, providing insights into their evolutionary significance and potential applications in aquaculture and molecular breeding.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhaobin Song
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College
of Life Sciences, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Aparecida Santos L, de Castro Dutra J, Picoli Marinho E, Cosme Cotta Malaquias L, Nascimento Gomes B, Caravita Grisolia J, Andrade Dias N, Burger E. Celecoxib exhibits antifungal effect against Paracoccidioides brasiliensis both directly and indirectly by activating neutrophil responses. Int Immunopharmacol 2024; 138:112606. [PMID: 38963980 DOI: 10.1016/j.intimp.2024.112606] [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: 04/10/2024] [Revised: 06/06/2024] [Accepted: 06/29/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Celecoxib, an anti-inflammatory drug, combined therapies using antimicrobials and immune modulator drugs are being studied. OBJECTIVE To assess whether Celecoxib has direct in vitro antifungal effect against the Paracoccidioides brasiliensis, the causative agent of Paracoccidioidomycosis-(PCM) and also if it improves the in vivo activity of neutrophils-(PMN) in an experimental murine subcutaneous-(air pouch) model of the disease. METHODS The antifungal activity of Celecoxib(6 mg/mL) on P. brasiliensis-(Pb18) was evaluated using the microdilution technique. Splenocytes co-cultured with Pb18 and treated with Celecoxib(6 mg/mL) were co-cultured for 24, 48 and 72-hours. Swiss mice were inoculated with Pb18 and treated with Celecoxib(6 mg/kg) in the subcutaneous air pouch. Neutrophils were collected from the air pouch. Mitochondrial activity, reactive oxygen production, catalase, peroxidase, cytokines and chemokines, nitrogen species, total protein, microbicidal activity of PMNs and viable Pb18 cells numbers were analyzed. RESULTS Celecoxib had no cytotoxic effect on splenocytes co-cultured with Pb18, but had a marked direct antifungal effect, inhibiting fungal growth both in vitro and in vivo. Celecoxib interaction with immune system cells in the air pouch, it leads to activation of PMNs, as confirmed by several parameters (mitochondrial activity, reactive oxygen species, peroxidase, KC and IL-6 increase, killing constant and phagocytosis). Celecoxib was able to reduce IL-4, IL-10 and IL-12 cytokine production. The number of recovered viable Pb18 decreased dramatically. CONCLUSIONS This is the first report of the direct antifungal activity of Celecoxib against P. brasiliensis. The use of Celecoxib opens a new possibility for future treatment of PCM.
Collapse
Affiliation(s)
- Lauana Aparecida Santos
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL. Alfenas, MG - Brazil CEP - 37130-001, Brazil
| | - Julia de Castro Dutra
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL. Alfenas, MG - Brazil CEP - 37130-001, Brazil
| | - Enrico Picoli Marinho
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL. Alfenas, MG - Brazil CEP - 37130-001, Brazil
| | - Luiz Cosme Cotta Malaquias
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL. Alfenas, MG - Brazil CEP - 37130-001, Brazil
| | - Bruno Nascimento Gomes
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL. Alfenas, MG - Brazil CEP - 37130-001, Brazil
| | - Julianne Caravita Grisolia
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL. Alfenas, MG - Brazil CEP - 37130-001, Brazil
| | - Nayara Andrade Dias
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL. Alfenas, MG - Brazil CEP - 37130-001, Brazil
| | - Eva Burger
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL. Alfenas, MG - Brazil CEP - 37130-001, Brazil.
| |
Collapse
|
5
|
Torrillo PA, Lieberman TD. Reversions mask the contribution of adaptive evolution in microbiomes. eLife 2024; 13:e93146. [PMID: 39240756 PMCID: PMC11379459 DOI: 10.7554/elife.93146] [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: 09/29/2023] [Accepted: 07/30/2024] [Indexed: 09/08/2024] Open
Abstract
When examining bacterial genomes for evidence of past selection, the results depend heavily on the mutational distance between chosen genomes. Even within a bacterial species, genomes separated by larger mutational distances exhibit stronger evidence of purifying selection as assessed by dN/dS, the normalized ratio of nonsynonymous to synonymous mutations. Here, we show that the classical interpretation of this scale dependence, weak purifying selection, leads to problematic mutation accumulation when applied to available gut microbiome data. We propose an alternative, adaptive reversion model with opposite implications for dynamical intuition and applications of dN/dS. Reversions that occur and sweep within-host populations are nearly guaranteed in microbiomes due to large population sizes, short generation times, and variable environments. Using analytical and simulation approaches, we show that adaptive reversion can explain the dN/dS decay given only dozens of locally fluctuating selective pressures, which is realistic in the context of Bacteroides genomes. The success of the adaptive reversion model argues for interpreting low values of dN/dS obtained from long timescales with caution as they may emerge even when adaptive sweeps are frequent. Our work thus inverts the interpretation of an old observation in bacterial evolution, illustrates the potential of mutational reversions to shape genomic landscapes over time, and highlights the importance of studying bacterial genomic evolution on short timescales.
Collapse
Affiliation(s)
- Paul A Torrillo
- Institute for Medical Engineering and Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
- Department of Civil and Environmental Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Tami D Lieberman
- Institute for Medical Engineering and Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
- Department of Civil and Environmental Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
- Broad Institute of MIT and HarvardCambridgeUnited States
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
| |
Collapse
|
6
|
Ferreira MDS, Gonçalves DDS, Mendoza SR, de Oliveira GA, Pontes B, la Noval CRD, Honorato L, Ramos LFC, Nogueira FCS, Domont GB, Casadevall A, Nimrichter L, Peralta JM, Guimaraes AJ. β-1,3-Glucan recognition by Acanthamoeba castellanii as a putative mechanism of amoeba-fungal interactions. Appl Environ Microbiol 2024; 90:e0173623. [PMID: 38259076 PMCID: PMC10880599 DOI: 10.1128/aem.01736-23] [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: 10/03/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
In this study, we conducted an in-depth analysis to characterize potential Acanthamoeba castellanii (Ac) proteins capable of recognizing fungal β-1,3-glucans. Ac specifically anchors curdlan or laminarin, indicating the presence of surface β-1,3-glucan-binding molecules. Using optical tweezers, strong adhesion of laminarin- or curdlan-coated beads to Ac was observed, highlighting their adhesive properties compared to controls (characteristic time τ of 46.9 and 43.9 s, respectively). Furthermore, Histoplasma capsulatum (Hc) G217B, possessing a β-1,3-glucan outer layer, showed significant adhesion to Ac compared to a Hc G186 strain with an α-1,3-glucan outer layer (τ of 5.3 s vs τ 83.6 s). The addition of soluble β-1,3-glucan substantially inhibited this adhesion, indicating the involvement of β-1,3-glucan recognition. Biotinylated β-1,3-glucan-binding proteins from Ac exhibited higher binding to Hc G217B, suggesting distinct recognition mechanisms for laminarin and curdlan, akin to macrophages. These observations hinted at the β-1,3-glucan recognition pathway's role in fungal entrance and survival within phagocytes, supported by decreased fungal viability upon laminarin or curdlan addition in both phagocytes. Proteomic analysis identified several Ac proteins capable of binding β-1,3-glucans, including those with lectin/glucanase superfamily domains, carbohydrate-binding domains, and glycosyl transferase and glycosyl hydrolase domains. Notably, some identified proteins were overexpressed upon curdlan/laminarin challenge and also demonstrated high affinity to β-1,3-glucans. These findings underscore the complexity of binding via β-1,3-glucan and suggest the existence of alternative fungal recognition pathways in Ac.IMPORTANCEAcanthamoeba castellanii (Ac) and macrophages both exhibit the remarkable ability to phagocytose various extracellular microorganisms in their respective environments. While substantial knowledge exists on this phenomenon for macrophages, the understanding of Ac's phagocytic mechanisms remains elusive. Recently, our group identified mannose-binding receptors on the surface of Ac that exhibit the capacity to bind/recognize fungi. However, the process was not entirely inhibited by soluble mannose, suggesting the possibility of other interactions. Herein, we describe the mechanism of β-1,3-glucan binding by A. castellanii and its role in fungal phagocytosis and survival within trophozoites, also using macrophages as a model for comparison, as they possess a well-established mechanism involving the Dectin-1 receptor for β-1,3-glucan recognition. These shed light on a potential parallel evolution of pathways involved in the recognition of fungal surface polysaccharides.
Collapse
Affiliation(s)
- Marina da Silva Ferreira
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Diego de Souza Gonçalves
- Programa de Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Susana Ruiz Mendoza
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Gabriel Afonso de Oliveira
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Bruno Pontes
- Instituto de Ciências Biomédicas e Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Claudia Rodríguez-de la Noval
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Leandro Honorato
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Luis Felipe Costa Ramos
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Fábio C. S. Nogueira
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Gilberto B. Domont
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Leonardo Nimrichter
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
- Laboratório de Glicobiologia de Eucariotos, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
- Rede Micologia RJ - Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Niterói, Rio de Janeiro, Brazil
| | - Jose Mauro Peralta
- Programa de Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
| | - Allan J. Guimaraes
- Laboratório de Bioquímica e Imunologia das Micoses, Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Niterói, Rio de Janeiro, Brazil
- Rede Micologia RJ - Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Niterói, Rio de Janeiro, Brazil
- Pós-Graduação em Microbiologia e Parasitologia Aplicadas, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Kanaparthi D, Lampe M, Krohn JH, Zhu B, Klingl A, Lueders T. The reproduction of gram-negative protoplasts and the influence of environmental conditions on this process. iScience 2023; 26:108149. [PMID: 37942012 PMCID: PMC10628739 DOI: 10.1016/j.isci.2023.108149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/31/2023] [Accepted: 10/02/2023] [Indexed: 11/10/2023] Open
Abstract
Bacterial protoplasts are known to reproduce independently of canonical molecular biological processes. Although their reproduction is thought to be influenced by environmental conditions, the growth of protoplasts in their natural habitat has never been empirically studied. Here, we studied the life cycle of protoplasts in their native environment. Contrary to the previous perception that protoplasts reproduce in an erratic manner, cells in our study reproduced in a defined sequence of steps, always leading to viable daughter cells. Their reproduction can be explained by an interplay between intracellular metabolism, the physicochemical properties of cell constituents, and the nature of cations in the growth media. The efficiency of reproduction is determined by the environmental conditions. Under favorable environmental conditions, protoplasts reproduce with nearly similar efficiency to cells that possess a cell wall. In short, here we demonstrate the simplest method of cellular reproduction and the influence of environmental conditions on this process.
Collapse
Affiliation(s)
- Dheeraj Kanaparthi
- Max-Planck Institute for Biochemistry, Munich, Germany
- Chair of Ecological Microbiology, BayCeer, University of Bayreuth, Bayreuth, Germany
- Excellence Cluster ORIGINS, Garching, Germany
| | - Marko Lampe
- Advanced Light Microscopy Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jan-Hagen Krohn
- Max-Planck Institute for Biochemistry, Munich, Germany
- Excellence Cluster ORIGINS, Garching, Germany
| | - Baoli Zhu
- Chair of Ecological Microbiology, BayCeer, University of Bayreuth, Bayreuth, Germany
- Key Laboratory of Agro-ecological Processes in Subtropical Regions, CAS, Changsha, China
| | - Andreas Klingl
- Department of Biology, LMU, Planegg-Martinsried, Germany
| | - Tillmann Lueders
- Chair of Ecological Microbiology, BayCeer, University of Bayreuth, Bayreuth, Germany
| |
Collapse
|
8
|
Egusa M, Watanabe S, Li H, Zewude DA, Ifuku S, Kaminaka H. Production of copper nanoparticle-immobilized chitin nanofibers and their role in plant disease control. JOURNAL OF PESTICIDE SCIENCE 2023; 48:86-92. [PMID: 37745172 PMCID: PMC10513960 DOI: 10.1584/jpestics.d23-001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/25/2023] [Indexed: 09/26/2023]
Abstract
Chitin is used in agriculture to improve crop production; however, its use is limited due to difficulties in its handling. A chitin nanofiber (CNF) overcomes this issue and, due to its elicitor activity, has great potential for crop protection. To expand CNF utilization, a copper nanoparticles-based antimicrobic CNF (CuNPs/CNF) was prepared using a chemical reduction method. The formation of CuNPs was confirmed via scanning electron microscopy. Thermogravimetric analysis revealed that the amount of CuNPs on the CNF was dose-dependent on the precursor salt, copper acetate. CuNPs endowed the CNF with strong antimicrobial activity against Alternaria brassicicola and Pectobacterium carotovorum. Moreover, the CuNPs/CNF reduced pathogen infection in cabbage. The antimicrobial activity and disease prevention of the CuNPs/CNF was increased compared to the corresponding CNF or commercial agrochemical Bordeaux treatment. These results indicate that CuNPs conferred antimicrobial activity on the CNF and increased the efficacy of plant disease protection.
Collapse
Affiliation(s)
| | | | - Hujun Li
- Department of Engineering, Graduate School of Sustainability Science, Tottori University
| | - Dagmawi Abebe Zewude
- Department of Engineering, Graduate School of Sustainability Science, Tottori University
- Unused Bioresource Utilization Center, Tottori University
| | - Shinsuke Ifuku
- Department of Engineering, Graduate School of Sustainability Science, Tottori University
- Center for Research on Green Sustainable Chemistry, Tottori University
- Unused Bioresource Utilization Center, Tottori University
| | - Hironori Kaminaka
- Faculty of Agriculture, Tottori University
- Unused Bioresource Utilization Center, Tottori University
| |
Collapse
|
9
|
Xu X, Qiao Y, Peng Q, Shi B. Probiotic Properties of Loigolactobacillus coryniformis NA-3 and In Vitro Comparative Evaluation of Live and Heat-Killed Cells for Antioxidant, Anticancer and Immunoregulatory Activities. Foods 2023; 12:foods12051118. [PMID: 36900635 PMCID: PMC10001366 DOI: 10.3390/foods12051118] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Some Latiactobacilli are often used as probiotics due to their functional activities, including antioxidant, anticancer and immunoregulation effect. Loigolactobacillus coryniformis NA-3 obtained from our laboratory is a promising probiotic according to the previous study. Coculture, the Oxford cup test and disk-diffusion methods were used to evaluate the probiotic properties and antibiotic resistance of L. coryniformis NA-3. The antioxidant activities of live and heat-killed L. coryniformis NA-3 were assessed via radicals' scavenging ability. The potential anticancer and immunoregulatory capacity was determined in vitro using cell lines. The results indicate that L. coryniformis NA-3 has antibacterial activity and cholesterol removal ability and is sensitive to most antibiotics. Dead L. coryniformis NA-3 can scavenge free radicals as well as live strains. Live L. coryniformis NA-3 can significantly inhibit the proliferation of colon cancer cells; however, dead cells cannot. After RAW 264.7 macrophages were treated with live and heat-killed L. coryniformis NA-3, the production of NO, IL-6, TNF-α and reactive oxygen species (ROS) was induced. The increased expression of inducible nitric oxide synthase (iNOS) in treated macrophages mediates the production of NO. In conclusion, L. coryniformis NA-3 showed potential probiotic properties, and the heat-killed strain also exhibited activities similar to those of live bacteria, suggesting the possible value of its further application in the food processing and pharmaceutical industries.
Collapse
|
10
|
Growth performance, feed utilization, hematological parameters, and histological features of Nile tilapia ( Oreochromis niloticus) fed diets with supplementary herbal extracts under prolonged water exchange. ANNALS OF ANIMAL SCIENCE 2023. [DOI: 10.2478/aoas-2023-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Abstract
Nile tilapia is known for its relative tolerance to some biotic and abiotic stressors. However, long-period water exchange may impair the health status and, thereby, the growth performance and feed utilization. In this regard, using herbal extracts may help to relieve the adverse impacts of low water quality on the productivity of this fish species. A 100-day feeding trial was designed to examine the effects of dietary supplements of Aloe vera leaves (AVE), and chamomile flowers, Matricaria chamomilla (CFE) extracts on water quality, growth performance, and well-being of Nile tilapia. Fish (3.95±0.05 g, n=1200) were distributed into five groups (15 fiberglass tanks, 2 m3) under a water exchange rate of 20% every two days for standard control (T0) without herbal supplements. In groups (T1, T2, T3, and T4), a long period water exchange period of 50% every month, wherein T1 (stressed control), T2, T3, and T4 groups were fed a diet supplemented with 0% herbal extract, 1% AVE, 1% CFE and 0.5% AVE +0.5% CFE, respectively. Results showed that feeding on a T2 diet exhibited a clear improvement under a long period of water exchange stress, and it is almost similar to their counterparts under normal conditions (T0) in terms of specific growth rate, survival rate, feed intake, feed conversion ratio, and protein efficiency ratio. Moreover, the lowest levels of aspartate aminotransferase were obtained with T2 and T0. In addition, the highest white blood cell count and hemoglobin values were recorded with group T0, followed by T2, and T4, while no significant difference between T3 and T1. Fish under stressed conditions without herbal extracts (T1) showed the lowest performance and survival rate compared to T0 and T2 groups. In conclusion, feeding diet supplied with 1% A. vera extract to stressed fish restored their performance and well-being to the level of their counterparts under normal conditions.
Collapse
|
11
|
Dimova T, Dimitrova V, Grozdanov P, Markova N. Placentа of BCG-Vaccinated Women in early Pregnancy is Colonized with Non-Immunogenic Mycobacterial L-forms. Am J Reprod Immunol 2023; 89:e13650. [PMID: 36331422 DOI: 10.1111/aji.13650] [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: 05/23/2022] [Revised: 10/20/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
PROBLEM Long-lived mycobacterial L-forms (mL-forms) could be detected in the blood of BCG-vaccinated people. We have previously found mL-forms in term placentas and blood of neonates, delivered by healthy BCG-vaccinated mothers as first formal demonstration that BCG vaccination in the childhood of the woman could affect her placentobiome during pregnancy. Of note, the isolated mL-forms reverted to the cell-walled state of the parental BCG bacilli in vitro. METHOD OF STUDY Here, we analyzed triple samples of blood, decidua and chorion taken from BCG-vaccinated pregnant women, directed to elective abortions (6-12 gestation weeks). The colonization of the primary samples with mycobacterial L-forms (mL-forms) was evaluated using microbiological isolation and subsequent identification by real time PCR and morphological characterization by light microscopy and SEM. The potential of early placenta-derived mL-forms to expand mycobacteria-reactive γδ T cells in vitro was assessed using FACS, whereas their immunogenicity in vivo was followed up after i.p. inoculation in rats. RESULTS Our results showed two important findings: 1) viable filterable mL-forms varying in size, shape and proliferation modes are capable of colonizing the gestational tissues of BCG-vaccinated women early in pregnancy and 2) early placenta-derived mL-forms are not as immunogenic as walled M. bovis BCG bacilli, shown by lack of stimulation of mycobacteria-reactive γδ T cells co-cultured with early placenta-derived mL-forms and inefficient internalization of mL-forms by rat's peritoneal phagocytes in vivo. CONCLUSION Although generally thought to be reduced in virulence, mL-forms could provide a reservoir, hidden from the immune system especially in an immune privileged niche like placenta.
Collapse
Affiliation(s)
- Tanya Dimova
- Institute of Biology and Immunology of Reproduction "Acad. K. Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Violeta Dimitrova
- Medical University, University Obstetrics and Gynecology Hospital "Maichin Dom", Sofia, Bulgaria
| | - Petar Grozdanov
- Stephan Angeloff Institute of Microbiology Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Nadya Markova
- Stephan Angeloff Institute of Microbiology Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
12
|
Silva AJD, Rocha CKDS, de Freitas AC. Standardization and Key Aspects of the Development of Whole Yeast Cell Vaccines. Pharmaceutics 2022; 14:pharmaceutics14122792. [PMID: 36559285 PMCID: PMC9781213 DOI: 10.3390/pharmaceutics14122792] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
In the context of vaccine development, improving antigenic presentation is critical for the activation of specific immune responses and the success of immunization, in addition to selecting an appropriate target. In this sense, different strategies have been developed and improved. Among them is the use of yeast cells as vehicles for the delivery of recombinant antigens. These vaccines, named whole yeast vaccines (WYVs), can induce humoral and cellular immune responses, with the additional advantage of dispensing with the use of adjuvants due to the immunostimulatory properties of their cell wall components. However, there are some gaps in the methodologies for obtaining and validating recombinant strains and vaccine formulations. The standardization of these parameters is an important factor for WYVs approval by regulatory agencies and, consequently, their licensing. This review aimed to provide an overview of the main parameters to consider when developing a yeast-based vaccine, addressing some available tools, and highlighting the main variables that can influence the vaccine production process.
Collapse
Affiliation(s)
- Anna Jéssica Duarte Silva
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | | | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
- Correspondence: ; Tel.: +55-81996067671
| |
Collapse
|
13
|
Kang SJ, Yang J, Lee NY, Lee CH, Park IB, Park SW, Lee HJ, Park HW, Yun HS, Chun T. Monitoring Cellular Immune Responses after Consumption of Selected
Probiotics in Immunocompromised Mice. Food Sci Anim Resour 2022; 42:903-914. [PMID: 36133633 PMCID: PMC9478974 DOI: 10.5851/kosfa.2022.e44] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/02/2022] [Accepted: 08/11/2022] [Indexed: 11/25/2022] Open
Abstract
Probiotics are currently considered as one of tools to modulate immune responses
under specific clinical conditions. The purpose of this study was to evaluate
whether oral administration of three different probiotics
(Lactiplantibacillus plantarum CJLP243, CJW55-10, and
CJLP475) could evoke a cell-mediated immunity in immunodeficient mice. Before
conducting in vivo experiments, we examined the in
vitro potency of these probiotics for macrophage activation. After
co-culture with these probiotics, bone marrow derived macrophages (BMDMs)
produced significant amounts of proinflammatory cytokines including
interleukin-6 (IL-6), IL-12, and tumor necrosis factor-α (TNF-α).
Levels of inducible nitric oxide synthase (inos) and
co-stimulatory molecules (CD80 and CD86) were also upregulated in BMDMs after
treatment with some of these probiotics. To establish an immunocompromised
animal model, we intraperitoneally injected mice with cyclophosphamide on day 0
and again on day 2. Starting day 3, we orally administered probiotics every day
for the last 15 d. After sacrificing experimental mice on day 18, splenocytes
were isolated and co-cultured with these probiotics for 3 d to measure levels of
several cytokines and immune cell proliferation. Results clearly indicated that
the consumption of all three probiotic strains promoted secretion of
interferon-γ (IFN-γ), IL-1β, IL-6, IL-12, and TNF-α.
NK cell cytotoxicity and proliferation of immune cells were also increased.
Taken together, our data strongly suggest that consumption of some probiotics
might induce cell-mediated immune responses in immunocompromised mice.
Collapse
Affiliation(s)
- Seok-Jin Kang
- Department of Biotechnology, College of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
- Institute of Animal Molecular
Biotechnology, Korea University, Seoul 02841, Korea
| | - Jun Yang
- Department of Biotechnology, College of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| | - Na-Young Lee
- Department of Biotechnology, College of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| | - Chang-Hee Lee
- Department of Biotechnology, College of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
- Institute of Animal Molecular
Biotechnology, Korea University, Seoul 02841, Korea
| | - In-Byung Park
- Department of Biotechnology, College of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
- Institute of Animal Molecular
Biotechnology, Korea University, Seoul 02841, Korea
| | - Si-Won Park
- Department of Biotechnology, College of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| | - Hyeon Jeong Lee
- Department of Biotechnology, College of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| | - Hae-Won Park
- Department of Biotechnology, College of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
| | | | - Taehoon Chun
- Department of Biotechnology, College of
Life Sciences and Biotechnology, Korea University, Seoul
02841, Korea
- Corresponding author: Taehoon
Chun, Department of Biotechnology, College of Life Sciences and Biotechnology,
Korea University, Seoul 02841, Korea, Tel: +82-2-3290-3069, E-mail:
| |
Collapse
|
14
|
Wang X, Liu P, Jiang Y, Han B, Yan L. The prophylactic effects of monoclonal antibodies targeting the cell wall Pmt4 protein epitopes of Candida albicans in a murine model of invasive candidiasis. Front Microbiol 2022; 13:992275. [PMID: 36081783 PMCID: PMC9446456 DOI: 10.3389/fmicb.2022.992275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Candida albicans (C. albicans) is the most prevalent opportunistic human pathogen, accounting for approximately half of all clinical cases of candidemia. Resistance to the existing antifungal drugs is a major challenge in clinical therapy, necessitating the development and identification of novel therapeutic agents and potential treatment strategies. Monoclonal antibody-based immunotherapy represents a promising therapeutic strategy against disseminated candidiasis. Protein mannosyltransferase (Pmt4) encodes mannosyltransferases initiating O-mannosylation of secretory proteins and is essential for cell wall composition and virulence of C. albicans. Therefore, the Pmt4 protein of C. albicans is an attractive target for the discovery of alternative antibody agents against invasive C. albicans infections. In the present study, we found that monoclonal antibodies (mAbs) C12 and C346 specifically targeted the recombinant protein mannosyltransferase 4 (rPmt4p) of C. albicans. These mAbs were produced and secreted by hybridoma cells isolated from the spleen of mice that were initially immunized with the purified rPmt4p to generate IgG antibodies. The mAbs C12 and C346 exhibited high affinity to C. albicans whole cells. Remarkably, these mAbs reduced the fungal burden, alleviated inflammation in the kidneys, and prolonged the survival rate significantly in the murine model of systemic candidiasis. Moreover, they could activate macrophage opsonophagocytic killing and neutrophil killing of C. albicans strain in vitro. These results suggested that anti-rPmt4p mAbs may provide immunotherapeutic interventions against disseminated candidiasis via opsonophagocytosis and opsonic killing activity. Our findings provide evidence for mAbs as a therapeutic option for the treatment of invasive candidiasis.
Collapse
Affiliation(s)
- Xiaojuan Wang
- School of Pharmacy, Naval Medical University, Shanghai, China
- Department of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
| | - Peng Liu
- Department of Gastroenterology, Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bing Han
- Department of Pharmacy, Minhang Hospital, Fudan University, Shanghai, China
- *Correspondence: Bing Han,
| | - Lan Yan
- School of Pharmacy, Naval Medical University, Shanghai, China
- Lan Yan,
| |
Collapse
|
15
|
Nguyen TT, Nguyen PT, Pham MN, Razafindralambo H, Hoang QK, Nguyen HT. Synbiotics: a New Route of Self-production and Applications to Human and Animal Health. Probiotics Antimicrob Proteins 2022; 14:980-993. [PMID: 35650337 DOI: 10.1007/s12602-022-09960-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2022] [Indexed: 01/17/2023]
Abstract
Synbiotics are preparations in which prebiotics are added to probiotics to achieve superior performance and benefits on the host. A new route of their formation is to induce the prebiotic biosynthesis within the probiotic for synbiotic self-production or autologous synbiotics. The aim of this review paper is first to overview the basic concept and (updated) definitions of synergistic synbiotics, and then to focus particularly on the prebiotic properties of probiotic wall components while describing the environmental factors/stresses that stimulate autologous synbiotics, that is, the biosynthesis of prebiotic-forming microcapsule by probiotic bacteria, and finally to present some of their applications to human and animal health.
Collapse
Affiliation(s)
- Thi-Tho Nguyen
- Hutech Institute of Applied Science, HUTECH University, Ho Chi Minh City, Vietnam
| | - Phu-Tho Nguyen
- An Giang University, An Giang, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Minh-Nhut Pham
- Hutech Institute of Applied Science, HUTECH University, Ho Chi Minh City, Vietnam
| | | | - Quoc-Khanh Hoang
- Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Huu-Thanh Nguyen
- An Giang University, An Giang, Vietnam.
- Vietnam National University, Ho Chi Minh City, Vietnam.
| |
Collapse
|
16
|
Cruz KCP, Enekegho LO, Stuart DT. Bioengineered Probiotics: Synthetic Biology Can Provide Live Cell Therapeutics for the Treatment of Foodborne Diseases. Front Bioeng Biotechnol 2022; 10:890479. [PMID: 35656199 PMCID: PMC9152101 DOI: 10.3389/fbioe.2022.890479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/29/2022] [Indexed: 11/15/2022] Open
Abstract
The rising prevalence of antibiotic resistant microbial pathogens presents an ominous health and economic challenge to modern society. The discovery and large-scale development of antibiotic drugs in previous decades was transformational, providing cheap, effective treatment for what would previously have been a lethal infection. As microbial strains resistant to many or even all antibiotic drug treatments have evolved, there is an urgent need for new drugs or antimicrobial treatments to control these pathogens. The ability to sequence and mine the genomes of an increasing number of microbial strains from previously unexplored environments has the potential to identify new natural product antibiotic biosynthesis pathways. This coupled with the power of synthetic biology to generate new production chassis, biosensors and “weaponized” live cell therapeutics may provide new means to combat the rapidly evolving threat of drug resistant microbial pathogens. This review focuses on the application of synthetic biology to construct probiotic strains that have been endowed with functionalities allowing them to identify, compete with and in some cases kill microbial pathogens as well as stimulate host immunity. Weaponized probiotics may have the greatest potential for use against pathogens that infect the gastrointestinal tract: Vibrio cholerae, Staphylococcus aureus, Clostridium perfringens and Clostridioides difficile. The potential benefits of engineered probiotics are highlighted along with the challenges that must still be met before these intriguing and exciting new therapeutic tools can be widely deployed.
Collapse
|
17
|
Bacterial Microarrays for Examining Bacterial Glycosignatures and Recognition by Host Lectins. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2460:147-160. [PMID: 34972935 DOI: 10.1007/978-1-0716-2148-6_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The surface of bacteria displays diverse carbohydrate structures that may significantly differ among bacteria with the same cell wall architecture and even among strains of a given bacterial species. These structures are often recognized by lectins of the innate immune system for triggering defense responses, although some bacterial pathogens exploit recognition by host lectins for favoring infection. Bacterial microarrays are a useful tool for profiling accessible bacterial surface glycans and for exploring their recognition by innate immune lectins. The use of array-printed bacterial cells enables evaluation of the recognition of the glycan epitopes in their natural presentation, i.e., preserving their real density and accessibility. Glycosylation patterns of bacterial surfaces can be examined by testing the binding to the bacterial arrays of a panel of lectins with known carbohydrate-binding preferences, and the recognition of surface glycans by innate immune lectins can easily be assessed using similar binding assays.
Collapse
|
18
|
Ebersole JL, Kirakodu S, Nguyen L, Gonzalez OA. Gingival Transcriptome of Innate Antimicrobial Factors and the Oral Microbiome With Aging and Periodontitis. FRONTIERS IN ORAL HEALTH 2022; 3:817249. [PMID: 35330821 PMCID: PMC8940521 DOI: 10.3389/froh.2022.817249] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/04/2022] [Indexed: 12/30/2022] Open
Abstract
The epithelial barrier at mucosal sites comprises an important mechanical protective feature of innate immunity, and is intimately involved in communicating signals of infection/tissue damage to inflammatory and immune cells in these local environments. A wide array of antimicrobial factors (AMF) exist at mucosal sites and in secretions that contribute to this innate immunity. A non-human primate model of ligature-induced periodontitis was used to explore characteristics of the antimicrobial factor transcriptome (n = 114 genes) of gingival biopsies in health, initiation and progression of periodontal lesions, and in samples with clinical resolution. Age effects and relationship of AMF to the dominant members of the oral microbiome were also evaluated. AMF could be stratified into 4 groups with high (n = 22), intermediate (n = 29), low (n = 18) and very low (n = 45) expression in healthy adult tissues. A subset of AMF were altered in healthy young, adolescent and aged samples compared with adults (e.g., APP, CCL28, DEFB113, DEFB126, FLG2, PRH1) and were affected across multiple age groups. With disease, a greater number of the AMF genes were affected in the adult and aged samples with skewing toward decreased expression, for example WDC12, PGLYRP3, FLG2, DEFB128, and DEF4A/B, with multiple age groups. Few of the AMF genes showed a >2-fold increase with disease in any age group. Selected AMF exhibited significant positive correlations across the array of AMF that varied in health and disease. In contrast, a rather limited number of the AMF significantly correlated with members of the microbiome; most prominent in healthy samples. These correlated microbes were different in younger and older samples and differed in health, disease and resolution samples. The findings supported effects of age on the expression of AMF genes in healthy gingival tissues showing a relationship to members of the oral microbiome. Furthermore, a dynamic expression of AMF genes was related to the disease process and showed similarities across the age groups, except for low/very low expressed genes that were unaffected in young samples. Targeted assessment of AMF members from this large array may provide insight into differences in disease risk and biomolecules that provide some discernment of early transition to disease.
Collapse
Affiliation(s)
- Jeffrey L. Ebersole
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, NV, United States
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
| | - Sreenatha Kirakodu
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
| | - Linh Nguyen
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, NV, United States
| | - Octavio A. Gonzalez
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
- Division of Periodontology, College of Dentistry, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
19
|
Sutton G, Fogel GB, Abramson B, Brinkac L, Michael T, Liu ES, Thomas S. Horizontal transfer and evolution of wall teichoic acid gene cassettes in Bacillus subtilis. F1000Res 2022; 10:354. [PMID: 35035886 PMCID: PMC8753576 DOI: 10.12688/f1000research.51874.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/06/2021] [Indexed: 12/31/2022] Open
Abstract
Background: Wall teichoic acid (WTA) genes are essential for production of cell walls in gram-positive bacteria and necessary for survival and variability in the cassette has led to recent antibiotic resistance acquisition in pathogenic bacteria. Methods: Using a pan-genome approach, we examined the evolutionary history of WTA genes in
Bacillus subtilis ssp.
subtilis. Results: Our analysis reveals an interesting pattern of evolution from the type-strain WTA gene cassette possibly resulting from horizontal acquisition from organisms with similar gene sequences. The WTA cassettes have a high level of variation which may be due to one or more independent horizontal transfer events during the evolution of
Bacillus subtilis ssp.
subtilis. This swapping of entire WTA cassettes and smaller regions within the WTA cassettes is an unusual feature in the evolution of the
Bacillus subtilis genome and highlights the importance of horizontal transfer of gene cassettes through homologous recombination within
B. subtilis or other bacterial species. Conclusions: Reduced sequence conservation of these WTA cassettes may indicate a modified function like the previously documented WTA ribitol/glycerol variation. An improved understanding of high-frequency recombination of gene cassettes has ramifications for synthetic biology and the use of
B. subtilis in industry.
Collapse
Affiliation(s)
- Granger Sutton
- J. Craig Venter Institute, Rockville, Maryland, 20850, USA
| | - Gary B Fogel
- Natural Selection, Inc., San Diego, CA, 92121, USA
| | - Bradley Abramson
- The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | | | - Todd Michael
- The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Enoch S Liu
- Natural Selection, Inc., San Diego, CA, 92121, USA
| | | |
Collapse
|
20
|
OUP accepted manuscript. Glycobiology 2022; 32:712-719. [DOI: 10.1093/glycob/cwac027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/05/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
|
21
|
Garrido V, Piñero‐Lambea C, Rodriguez‐Arce I, Paetzold B, Ferrar T, Weber M, Garcia‐Ramallo E, Gallo C, Collantes M, Peñuelas I, Serrano L, Grilló M, Lluch‐Senar M. Engineering a genome-reduced bacterium to eliminate Staphylococcus aureus biofilms in vivo. Mol Syst Biol 2021; 17:e10145. [PMID: 34612607 PMCID: PMC8493563 DOI: 10.15252/msb.202010145] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 01/25/2023] Open
Abstract
Bacteria present a promising delivery system for treating human diseases. Here, we engineered the genome-reduced human lung pathogen Mycoplasma pneumoniae as a live biotherapeutic to treat biofilm-associated bacterial infections. This strain has a unique genetic code, which hinders gene transfer to most other bacterial genera, and it lacks a cell wall, which allows it to express proteins that target peptidoglycans of pathogenic bacteria. We first determined that removal of the pathogenic factors fully attenuated the chassis strain in vivo. We then designed synthetic promoters and identified an endogenous peptide signal sequence that, when fused to heterologous proteins, promotes efficient secretion. Based on this, we equipped the chassis strain with a genetic platform designed to secrete antibiofilm and bactericidal enzymes, resulting in a strain capable of dissolving Staphylococcus aureus biofilms preformed on catheters in vitro, ex vivo, and in vivo. To our knowledge, this is the first engineered genome-reduced bacterium that can fight against clinically relevant biofilm-associated bacterial infections.
Collapse
Affiliation(s)
- Victoria Garrido
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Institute of Agrobiotechnology (IdAB; CSIC‐Navarra Government)MutilvaSpain
| | - Carlos Piñero‐Lambea
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Pulmobiotics LtdBarcelonaSpain
| | - Irene Rodriguez‐Arce
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Institute of Agrobiotechnology (IdAB; CSIC‐Navarra Government)MutilvaSpain
| | - Bernhard Paetzold
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- S‐Biomedic N.V.BeerseBelgium
| | - Tony Ferrar
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Marc Weber
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Eva Garcia‐Ramallo
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Pulmobiotics LtdBarcelonaSpain
| | - Carolina Gallo
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - María Collantes
- RADIOMIN Research GroupClínica Universidad de NavarraPamplonaSpain
- IdiSNA, Navarra Institute for Health ResearchPamplonaSpain
| | - Iván Peñuelas
- RADIOMIN Research GroupClínica Universidad de NavarraPamplonaSpain
- IdiSNA, Navarra Institute for Health ResearchPamplonaSpain
| | - Luis Serrano
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- ICREABarcelonaSpain
| | - María‐Jesús Grilló
- Institute of Agrobiotechnology (IdAB; CSIC‐Navarra Government)MutilvaSpain
| | - María Lluch‐Senar
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Pulmobiotics LtdBarcelonaSpain
- Basic Sciences DepartmentFaculty of Medicine and Health SciencesUniversitat Internacional de CatalunyaSant Cugat del VallèsSpain
| |
Collapse
|
22
|
Mechanism of Staphylococcus aureus peptidoglycan O-acetyltransferase A as an O-acyltransferase. Proc Natl Acad Sci U S A 2021; 118:2103602118. [PMID: 34480000 DOI: 10.1073/pnas.2103602118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 07/23/2021] [Indexed: 01/05/2023] Open
Abstract
The O-acetylation of exopolysaccharides, including the essential bacterial cell wall polymer peptidoglycan, confers resistance to their lysis by exogenous hydrolases. Like the enzymes catalyzing the O-acetylation of exopolysaccharides in the Golgi of animals and fungi, peptidoglycan O-acetyltransferase A (OatA) is predicted to be an integral membrane protein comprised of a membrane-spanning acyltransferase-3 (AT-3) domain and an extracytoplasmic domain; for OatA, these domains are located in the N- and C-terminal regions of the enzyme, respectively. The recombinant C-terminal domain (OatAC) has been characterized as an SGNH acetyltransferase, but nothing was known about the function of the N-terminal AT-3 domain (OatAN) or its homologs associated with other acyltransferases. We report herein the experimental determination of the topology of Staphylococcus aureus OatAN, which differs markedly from that predicted in silico. We present the biochemical characterization of OatAN as part of recombinant OatA and demonstrate that acetyl-CoA serves as the substrate for OatAN Using in situ and in vitro assays, we characterized 35 engineered OatA variants which identified a catalytic triad of Tyr-His-Glu residues. We trapped an acetyl group from acetyl-CoA on the catalytic Tyr residue that is located on an extracytoplasmic loop of OatAN Further enzymatic characterization revealed that O-acetyl-Tyr represents the substrate for OatAC We propose a model for OatA action involving the translocation of acetyl groups from acetyl-CoA across the cytoplasmic membrane by OatAN and their subsequent intramolecular transfer to OatAC for the O-acetylation of peptidoglycan via the concerted action of catalytic Tyr and Ser residues.
Collapse
|
23
|
Bhat SV, Price JDW, Dahms TES. AFM-Based Correlative Microscopy Illuminates Human Pathogens. Front Cell Infect Microbiol 2021; 11:655501. [PMID: 34026660 PMCID: PMC8138568 DOI: 10.3389/fcimb.2021.655501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/08/2021] [Indexed: 12/25/2022] Open
Abstract
Microbes have an arsenal of virulence factors that contribute to their pathogenicity. A number of challenges remain to fully understand disease transmission, fitness landscape, antimicrobial resistance and host heterogeneity. A variety of tools have been used to address diverse aspects of pathogenicity, from molecular host-pathogen interactions to the mechanisms of disease acquisition and transmission. Current gaps in our knowledge include a more direct understanding of host-pathogen interactions, including signaling at interfaces, and direct phenotypic confirmation of pathogenicity. Correlative microscopy has been gaining traction to address the many challenges currently faced in biomedicine, in particular the combination of optical and atomic force microscopy (AFM). AFM, generates high-resolution surface topographical images, and quantifies mechanical properties at the pN scale under physiologically relevant conditions. When combined with optical microscopy, AFM probes pathogen surfaces and their physical and molecular interaction with host cells, while the various modes of optical microscopy view internal cellular responses of the pathogen and host. Here we review the most recent advances in our understanding of pathogens, recent applications of AFM to the field, how correlative AFM-optical microspectroscopy and microscopy have been used to illuminate pathogenicity and how these methods can reach their full potential for studying host-pathogen interactions.
Collapse
Affiliation(s)
- Supriya V Bhat
- Department of Chemistry and Biochemistry, University of Regina, Regina, SK, Canada
| | - Jared D W Price
- Department of Chemistry and Biochemistry, University of Regina, Regina, SK, Canada
| | - Tanya E S Dahms
- Department of Chemistry and Biochemistry, University of Regina, Regina, SK, Canada
| |
Collapse
|
24
|
Selenium-Containing Polysaccharides—Structural Diversity, Biosynthesis, Chemical Modifications and Biological Activity. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11083717] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Selenosugars are a group of sugar derivatives of great structural diversity (e.g., molar masses, selenium oxidation state, and selenium binding), obtained as a result of biosynthesis, chemical modification of natural compounds, or chemical synthesis. Seleno-monosaccharides and disaccharides are known to be non-toxic products of the natural metabolism of selenium compounds in mammals. In the case of the selenium-containing polysaccharides of natural origin, their formation is also postulated as a form of detoxification of excess selenium in microorganisms, mushroom, and plants. The valency of selenium in selenium-containing polysaccharides can be: 0 (encapsulated nano-selenium), IV (selenites of polysaccharides), or II (selenoglycosides or selenium built into the sugar ring to replace oxygen). The great interest in Se-polysaccharides results from the expected synergy between selenium and polysaccharides. Several plant- and mushroom-derived polysaccharides are potent macromolecules with antitumor, immunomodulatory, antioxidant, and other biological properties. Selenium, a trace element of fundamental importance to human health, has been shown to possess several analogous functions. The mechanism by which selenium exerts anticancer and immunomodulatory activity differs from that of polysaccharide fractions, but a similar pharmacological effect suggests a possible synergy of these two agents. Various functions of Se-polysaccharides have been explored, including antitumor, immune-enhancement, antioxidant, antidiabetic, anti-inflammatory, hepatoprotective, and neuroprotective activities. Due to being non-toxic or much less toxic than inorganic selenium compounds, Se-polysaccharides are potential dietary supplements that could be used, e.g., in chemoprevention.
Collapse
|
25
|
Silva AJD, de Macêdo LS, Leal LRS, de Jesus ALS, Freitas AC. Yeasts as a promising delivery platform for DNA and RNA vaccines. FEMS Yeast Res 2021; 21:foab018. [PMID: 33837785 DOI: 10.1093/femsyr/foab018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/19/2021] [Indexed: 12/13/2022] Open
Abstract
Yeasts are considered a useful system for the development of vaccines for human and veterinary health. Species such as Saccharomyces cerevisiae and Pichia pastoris have been used successfully as host organisms for the production of subunit vaccines. These organisms have been also explored as vaccine vehicles enabling the delivery of antigens such as proteins and nucleic acids. The employed species possess a GRAS status (Generally Recognized as Safe) for the production of therapeutic proteins, besides promoting immunostimulation due to the properties of their wall cell composition. This strategy allows the administration of nucleic acids orally and a specific delivery to professional antigen-presenting cells (APCs). In this review, we seek to outline the development of whole yeast vaccines (WYV) carrying nucleic acids in different approaches in the medical field, as well as the immunological aspects of this vaccine strategy. The data presented here reveal the application of this platform in promoting effective immune responses in the context of prophylactic and therapeutic approaches.
Collapse
Affiliation(s)
- Anna Jéssica Duarte Silva
- Laboratório de Estudos Moleculares e Terapia Experimental, Department of Genetics, Federal University of Pernambuco, Av. Prof. Moraes Rêgo, 1235, Cidade Universitaria, Recife, Pernambuco, Brazil
| | - Larissa Silva de Macêdo
- Laboratório de Estudos Moleculares e Terapia Experimental, Department of Genetics, Federal University of Pernambuco, Av. Prof. Moraes Rêgo, 1235, Cidade Universitaria, Recife, Pernambuco, Brazil
| | - Lígia Rosa Sales Leal
- Laboratório de Estudos Moleculares e Terapia Experimental, Department of Genetics, Federal University of Pernambuco, Av. Prof. Moraes Rêgo, 1235, Cidade Universitaria, Recife, Pernambuco, Brazil
| | - André Luiz Santos de Jesus
- Laboratório de Estudos Moleculares e Terapia Experimental, Department of Genetics, Federal University of Pernambuco, Av. Prof. Moraes Rêgo, 1235, Cidade Universitaria, Recife, Pernambuco, Brazil
| | - Antonio Carlos Freitas
- Laboratório de Estudos Moleculares e Terapia Experimental, Department of Genetics, Federal University of Pernambuco, Av. Prof. Moraes Rêgo, 1235, Cidade Universitaria, Recife, Pernambuco, Brazil
| |
Collapse
|
26
|
Aggarwal SD, Lloyd AJ, Yerneni SS, Narciso AR, Shepherd J, Roper DI, Dowson CG, Filipe SR, Hiller NL. A molecular link between cell wall biosynthesis, translation fidelity, and stringent response in Streptococcus pneumoniae. Proc Natl Acad Sci U S A 2021; 118:e2018089118. [PMID: 33785594 PMCID: PMC8040666 DOI: 10.1073/pnas.2018089118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Survival in the human host requires bacteria to respond to unfavorable conditions. In the important Gram-positive pathogen Streptococcus pneumoniae, cell wall biosynthesis proteins MurM and MurN are tRNA-dependent amino acyl transferases which lead to the production of branched muropeptides. We demonstrate that wild-type cells experience optimal growth under mildly acidic stressed conditions, but ΔmurMN strain displays growth arrest and extensive lysis. Furthermore, these stress conditions compromise the efficiency with which alanyl-tRNAAla synthetase can avoid noncognate mischarging of tRNAAla with serine, which is toxic to cells. The observed growth defects are rescued by inhibition of the stringent response pathway or by overexpression of the editing domain of alanyl-tRNAAla synthetase that enables detoxification of tRNA misacylation. Furthermore, MurM can incorporate seryl groups from mischarged Seryl-tRNAAlaUGC into cell wall precursors with exquisite specificity. We conclude that MurM contributes to the fidelity of translation control and modulates the stress response by decreasing the pool of mischarged tRNAs. Finally, we show that enhanced lysis of ΔmurMN pneumococci is caused by LytA, and the murMN operon influences macrophage phagocytosis in a LytA-dependent manner. Thus, MurMN attenuates stress responses with consequences for host-pathogen interactions. Our data suggest a causal link between misaminoacylated tRNA accumulation and activation of the stringent response. In order to prevent potential corruption of translation, consumption of seryl-tRNAAla by MurM may represent a first line of defense. When this mechanism is overwhelmed or absent (ΔmurMN), the stringent response shuts down translation to avoid toxic generation of mistranslated/misfolded proteins.
Collapse
Affiliation(s)
- Surya D Aggarwal
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Adrian J Lloyd
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom;
| | | | - Ana Rita Narciso
- Laboratory of Bacterial Cell Surfaces and Pathogenesis, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 1099-085 Oeiras, Portugal
- Unidade de Ciências Biomoleculares Aplicadas (UCIBIO), Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2825-149 Caparica, Portugal
| | - Jennifer Shepherd
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - David I Roper
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Christopher G Dowson
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Sergio R Filipe
- Laboratory of Bacterial Cell Surfaces and Pathogenesis, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 1099-085 Oeiras, Portugal;
- Unidade de Ciências Biomoleculares Aplicadas (UCIBIO), Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2825-149 Caparica, Portugal
| | - N Luisa Hiller
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213;
| |
Collapse
|
27
|
Mohammad M, Na M, Hu Z, Nguyen MT, Kopparapu PK, Jarneborn A, Karlsson A, Ali A, Pullerits R, Götz F, Jin T. Staphylococcus aureus lipoproteins promote abscess formation in mice, shielding bacteria from immune killing. Commun Biol 2021; 4:432. [PMID: 33785850 PMCID: PMC8010101 DOI: 10.1038/s42003-021-01947-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [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: 03/02/2021] [Indexed: 12/27/2022] Open
Abstract
Despite being a major bacterial factor in alerting the human immune system, the role of Staphylococcus aureus (S. aureus) lipoproteins (Lpp) in skin infections remains largely unknown. Here, we demonstrated that subcutaneous injection of S. aureus Lpp led to infiltration of neutrophils and monocytes/macrophages and induced skin lesions in mice. Lipid-moiety of S. aureus Lpp and host TLR2 was responsible for such effect. Lpp-deficient S. aureus strains exhibited smaller lesion size and reduced bacterial loads than their parental strains; the altered phenotype in bacterial loads was TLR2-independent. Lpp expression in skin infections contributed to imbalanced local hemostasis toward hypercoagulable state. Depletion of leukocytes or fibrinogen abrogated the effects induced by Lpp in terms of skin lesions and bacterial burden. Our data suggest that S. aureus Lpp induce skin inflammation and promote abscess formation that protects bacteria from innate immune killing. This suggests an intriguing bacterial immune evasion mechanism.
Collapse
Affiliation(s)
- Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Manli Na
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Zhicheng Hu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Microbiology and Immunology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Minh-Thu Nguyen
- Department of Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
- Section of Medical and Geographical Infectiology, Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Pradeep Kumar Kopparapu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Jarneborn
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anna Karlsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Abukar Ali
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rille Pullerits
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Friedrich Götz
- Department of Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
28
|
Burger E. Paracoccidioidomycosis Protective Immunity. J Fungi (Basel) 2021; 7:jof7020137. [PMID: 33668671 PMCID: PMC7918802 DOI: 10.3390/jof7020137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 12/26/2022] Open
Abstract
Protective immunity against Paracoccidioides consists of a stepwise activation of numerous effector mechanisms that comprise many cellular and soluble components. At the initial phase of non-specific innate immunity, resistance against Paracoccidioides comes from phagocytic polymorphonuclear neutrophils, natural killer (NK) cells and monocytes, supplemented by soluble factors such as cytokines and complement system components. Invariant receptors (Toll-like receptors (TLRs), Dectins) which are present in cells of the immune system, detect patterns present in Paracoccidioides (but not in the host) informing the hosts cells that there is an infection in progress, and that the acquired immunity must be activated. The role of components involved in the innate immunity of paracoccidioidomycosis is herein presented. Humoral immunity, represented by specific antibodies which control the fungi in the blood and body fluids, and its role in paracoccidioidomycosis (which was previously considered controversial) is also discussed. The protective mechanisms (involving various components) of cellular immunity are also discussed, covering topics such as: lysis by activated macrophages and cytotoxic T lymphocytes, the participation of lytic products, and the role of cytokines secreted by T helper lymphocytes in increasing the efficiency of Paracoccidioides, lysis.
Collapse
Affiliation(s)
- Eva Burger
- Department of Microbiology and Immunology, Universidade Federal de Alfenas, Alfenas 37130-001, Brazil
| |
Collapse
|
29
|
Baranov MV, Kumar M, Sacanna S, Thutupalli S, van den Bogaart G. Modulation of Immune Responses by Particle Size and Shape. Front Immunol 2021; 11:607945. [PMID: 33679696 PMCID: PMC7927956 DOI: 10.3389/fimmu.2020.607945] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022] Open
Abstract
The immune system has to cope with a wide range of irregularly shaped pathogens that can actively move (e.g., by flagella) and also dynamically remodel their shape (e.g., transition from yeast-shaped to hyphal fungi). The goal of this review is to draw general conclusions of how the size and geometry of a pathogen affect its uptake and processing by phagocytes of the immune system. We compared both theoretical and experimental studies with different cells, model particles, and pathogenic microbes (particularly fungi) showing that particle size, shape, rigidity, and surface roughness are important parameters for cellular uptake and subsequent immune responses, particularly inflammasome activation and T cell activation. Understanding how the physical properties of particles affect immune responses can aid the design of better vaccines.
Collapse
Affiliation(s)
- Maksim V. Baranov
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| | - Manoj Kumar
- Simons Center for the Study of Living Machines, National Centre for Biological Sciences, Tata Institute for Fundamental Research, Bangalore, India
| | - Stefano Sacanna
- Molecular Design Institute, Department of Chemistry, New York University, New York, NY, United States
| | - Shashi Thutupalli
- Simons Center for the Study of Living Machines, National Centre for Biological Sciences, Tata Institute for Fundamental Research, Bangalore, India
- International Centre for Theoretical Sciences, Tata Institute for Fundamental Research, Bangalore, India
| | - Geert van den Bogaart
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| |
Collapse
|
30
|
Jayakumar J, Kumar VA, Biswas L, Biswas R. Therapeutic applications of lysostaphin against Staphylococcus aureus. J Appl Microbiol 2021; 131:1072-1082. [PMID: 33382154 DOI: 10.1111/jam.14985] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/11/2020] [Accepted: 12/28/2020] [Indexed: 12/14/2022]
Abstract
Staphylococcus aureus, an opportunistic pathogen, causes diverse community and nosocomial-acquired human infections, including folliculitis, impetigo, sepsis, septic arthritis, endocarditis, osteomyelitis, implant-associated biofilm infections and contagious mastitis in cattle. In recent days, both methicillin-sensitive and methicillin-resistant S. aureus infections have increased. Highly effective anti-staphylococcal agents are urgently required. Lysostaphin is a 27 kDa zinc metallo antimicrobial lytic enzyme that is produced by Staphylococcus simulans biovar staphylolyticus and was first discovered in the 1960s. Lysostaphin is highly active against S. aureus strains irrespective of their drug-resistant patterns with a minimum inhibitory concentration of ranges between 0·001 and 0·064 μg ml-1 . Lysostaphin has activity against both dividing and non-dividing S. aureus cells; and can seep through the extracellular matrix to kill the biofilm embedded S. aureus. In spite of having excellent anti-staphylococcal activity, its clinical application is hindered because of its immunogenicity and reduced bio-availability. Extensive research with lysostaphin lead to the development of several engineered lysostaphin derivatives with reduced immunogenicity and increased serum half-life. Therapeutic efficacy of both native and engineered lysostaphin derivatives was studied by several research groups. This review provides an overview of the therapeutic applications of native and engineered lysostaphin derivatives developed to eradicate S. aureus infections.
Collapse
Affiliation(s)
- J Jayakumar
- Center for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - V A Kumar
- Department of Microbiology, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - L Biswas
- Center for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - R Biswas
- Center for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| |
Collapse
|
31
|
Vangroenweghe F, Poulsen K, Thas O. Supplementation of a β-mannanase enzyme reduces post-weaning diarrhea and antibiotic use in piglets on an alternative diet with additional soybean meal. Porcine Health Manag 2021; 7:8. [PMID: 33431048 PMCID: PMC7798280 DOI: 10.1186/s40813-021-00191-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/06/2021] [Indexed: 12/02/2022] Open
Abstract
Enzyme supplementation with a β-mannanase to degrade β-mannan fibers present in the diet has been shown to restore and improve performance in swine. The current study was conducted on a farm which had historical episodes of post-weaning diarrhea. In total, 896 newly weaned piglets were enrolled in two consecutive trials. Each trial consisted of 32 pens of 14 piglets housed in one large post-weaning compartment. Piglets at the same feeder were randomly assigned to the two treatment groups. The study compared the performance of post-weaned piglets fed either a commercial 3-phase nursery diet (Control) or an adapted diet supplemented with a β-mannanase (Hemicell HT; Elanco) (Enzyme), with some of the more expensive proteins replaced by soy bean meal in phase 1 and 2, and net energy (NE) content reduced by 65 kcal/kg in phase 3. All data analyses were performed using R version 3.6.3 (R Core Team, 2020). All tests were performed at the 5% level of significance. When multiple testing was involved, the nominal 5% Familywise Error Rate (FWER) was used. The study showed similar performance on the alternative diet with β-mannanase and the common commercial diets (P > 0.05). However, the Enzyme treated group had a significantly better general clinical score. Moreover, the number of individual treatments was a factor exp(0.69441) or 2 (CI 95% [1.46; 2.74]) higher (P < 0.001) in the Control group as compared to the Enzyme treated group. The number of treated animals was a factor exp(0.62861) or 1.87 (CI 95% [1.43; 2.53]) higher (P < 0.001) and the number of pigs with a repeated treatment was a factor exp(0.9293) or 2.53 (CI 95% [1.26; 5.09]) higher (P = 0.009) in the Control group as compared to the Enzyme treated group. In total, 7 (1.56%) piglets died in the Control group, whereas only 2 (0.45%) piglets died in the Enzyme treated group. The hazard ratio for mortality in the Control group relative to the Enzyme treated group was and estimated as 1.74 (CI 95% [0.51; 5.96]). Thus, the Control group had a non-significantly (P = 0.375) increased mortality. In conclusion, the results suggest that the use of an exogenous heat-tolerant β-mannanase allowed reduced levels of expensive protein sources to be used in the first two diets fed post-weaning, and 65 kcal/kg lower net energy content to be used in the third diet without adverse effects on intestinal health or overall performance. In fact, the occurrence of PWD and number of individual treatments during the post-weaning period were significantly reduced on the β-mannanase supplemented diets.
Collapse
Affiliation(s)
- Frédéric Vangroenweghe
- Elanco, BU Food Animals, Elanco Benelux, Plantijn en Moretuslei 1 - 3rd floor, 2018, Antwerpen, Belgium. .,Faculty of Veterinary Medicine, Unit of Porcine Health Management, Ghent University, Merelbeke, Belgium.
| | - Karl Poulsen
- Elanco, BU Nutritional Health, Plantijn en Moretuslei 1 - 3rd floor, 2018, Antwerpen, Belgium
| | - Olivier Thas
- I-BioStat, Data Science Institute, Hasselt University, Campus Diepenbeek, Agoralaan gebouw D, 3590, Diepenbeek, Belgium.,Department of Data Analysis and Mathematical Modelling, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium.,National Institute of Applied Statistics Research Australia (NIASRA), University of Wollongong, Northfields Ave, Wollongong, NSW, 2522, Australia
| |
Collapse
|
32
|
Moreno-Córdova EN, Islas-Osuna MA, Contreras-Vergara CA, López-Zavala AA, Ruiz-Bustos E, Reséndiz-Sandoval MG, Castillo-Yañez FJ, Criscitiello MF, Arvizu-Flores AA. Molecular characterization and expression analysis of the chicken-type and goose-type lysozymes from totoaba (Totoaba macdonaldi). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 113:103807. [PMID: 32735961 DOI: 10.1016/j.dci.2020.103807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/23/2020] [Accepted: 07/23/2020] [Indexed: 06/11/2023]
Abstract
Lysozymes play a key role in innate immune response to bacterial pathogens, catalyzing the hydrolysis of the peptidoglycan layer of bacterial cell walls. In this study, the genes encoding the c-type (TmLyzc) and g-type (TmLyzg) lysozymes from Totoaba macdonaldi were cloned and characterized. The cDNA sequences of TmLyzg and TmLyzc were 582 and 432 bp, encoding polypeptides of 193 and 143 amino acids, respectively. Amino acid sequences of these lysozymes shared high identity (60-90%) with their counterparts of other teleosts and showed conserved functional-structural signatures of the lysozyme superfamily. Phylogenetic analysis indicated a close relationship with their vertebrate homologues but distinct evolutionary paths for each lysozyme. Expression analysis by qRT-PCR revealed that TmLyzc was expressed in stomach and pyloric caeca, while TmLyzg was highly expressed in stomach and heart. These results suggest that both lysozymes play important roles in defense of totoaba against bacterial infections or as digestive enzyme.
Collapse
Affiliation(s)
- Elena N Moreno-Córdova
- Departamento de Ciencias Químico Biológicas, Universidad de Sonora. Blvd. Rosales S/N, Centro. Hermosillo, SON, CP, 83000, Mexico
| | - María A Islas-Osuna
- Departamento de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo, Gustavo Enrique Astiazarán Rosas, N0. 46. Hermosillo, SON, CP, 83304, Mexico
| | - Carmen A Contreras-Vergara
- Departamento de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo, Gustavo Enrique Astiazarán Rosas, N0. 46. Hermosillo, SON, CP, 83304, Mexico
| | - Alonso A López-Zavala
- Departamento de Ciencias Químico Biológicas, Universidad de Sonora. Blvd. Rosales S/N, Centro. Hermosillo, SON, CP, 83000, Mexico
| | - Eduardo Ruiz-Bustos
- Departamento de Ciencias Químico Biológicas, Universidad de Sonora. Blvd. Rosales S/N, Centro. Hermosillo, SON, CP, 83000, Mexico
| | - Mónica G Reséndiz-Sandoval
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, Gustavo Enrique Astiazarán Rosas, N0. 46. Hermosillo, SON, CP, 83304, Mexico
| | - Francisco J Castillo-Yañez
- Departamento de Ciencias Químico Biológicas, Universidad de Sonora. Blvd. Rosales S/N, Centro. Hermosillo, SON, CP, 83000, Mexico
| | - Michael F Criscitiello
- Comparative Immunogenetics Laboratory, Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA; Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University. Bryan, TX, 77807, USA.
| | - Aldo A Arvizu-Flores
- Departamento de Ciencias Químico Biológicas, Universidad de Sonora. Blvd. Rosales S/N, Centro. Hermosillo, SON, CP, 83000, Mexico.
| |
Collapse
|
33
|
Zhao J, Zhu Y, Han J, Lin YW, Aichem M, Wang J, Chen K, Velkov T, Schreiber F, Li J. Genome-Scale Metabolic Modeling Reveals Metabolic Alterations of Multidrug-Resistant Acinetobacter baumannii in a Murine Bloodstream Infection Model. Microorganisms 2020; 8:microorganisms8111793. [PMID: 33207684 PMCID: PMC7696501 DOI: 10.3390/microorganisms8111793] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 01/22/2023] Open
Abstract
Multidrug-resistant (MDR) Acinetobacter baumannii is a critical threat to human health globally. We constructed a genome-scale metabolic model iAB5075 for the hypervirulent, MDR A. baumannii strain AB5075. Predictions of nutrient utilization and gene essentiality were validated using Biolog assay and a transposon mutant library. In vivo transcriptomics data were integrated with iAB5075 to elucidate bacterial metabolic responses to the host environment. iAB5075 contains 1530 metabolites, 2229 reactions, and 1015 genes, and demonstrated high accuracies in predicting nutrient utilization and gene essentiality. At 4 h post-infection, a total of 146 metabolic fluxes were increased and 52 were decreased compared to 2 h post-infection; these included enhanced fluxes through peptidoglycan and lipopolysaccharide biosynthesis, tricarboxylic cycle, gluconeogenesis, nucleotide and fatty acid biosynthesis, and altered fluxes in amino acid metabolism. These flux changes indicate that the induced central metabolism, energy production, and cell membrane biogenesis played key roles in establishing and enhancing A. baumannii bloodstream infection. This study is the first to employ genome-scale metabolic modeling to investigate A. baumannii infection in vivo. Our findings provide important mechanistic insights into the adaption of A. baumannii to the host environment and thus will contribute to the development of new therapeutic agents against this problematic pathogen.
Collapse
Affiliation(s)
- Jinxin Zhao
- Infection and Immunity Program, Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (J.Z.); (Y.-W.L.); (J.W.); (K.C.)
| | - Yan Zhu
- Infection and Immunity Program, Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (J.Z.); (Y.-W.L.); (J.W.); (K.C.)
- Correspondence: (Y.Z.); (J.L.); Tel.: +61-3-99029178 (Y.Z.); +61-3-99039172 (J.L.); Fax: +61-3-99056450 (J.L.)
| | - Jiru Han
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia;
| | - Yu-Wei Lin
- Infection and Immunity Program, Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (J.Z.); (Y.-W.L.); (J.W.); (K.C.)
| | - Michael Aichem
- Department of Computer and Information Science, University of Konstanz, 78457 Konstanz, Germany; (M.A.); (F.S.)
| | - Jiping Wang
- Infection and Immunity Program, Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (J.Z.); (Y.-W.L.); (J.W.); (K.C.)
| | - Ke Chen
- Infection and Immunity Program, Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (J.Z.); (Y.-W.L.); (J.W.); (K.C.)
| | - Tony Velkov
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Falk Schreiber
- Department of Computer and Information Science, University of Konstanz, 78457 Konstanz, Germany; (M.A.); (F.S.)
| | - Jian Li
- Infection and Immunity Program, Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (J.Z.); (Y.-W.L.); (J.W.); (K.C.)
- Correspondence: (Y.Z.); (J.L.); Tel.: +61-3-99029178 (Y.Z.); +61-3-99039172 (J.L.); Fax: +61-3-99056450 (J.L.)
| |
Collapse
|
34
|
Galactomannan Produced by Aspergillus fumigatus: An Update on the Structure, Biosynthesis and Biological Functions of an Emblematic Fungal Biomarker. J Fungi (Basel) 2020; 6:jof6040283. [PMID: 33198419 PMCID: PMC7712326 DOI: 10.3390/jof6040283] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/03/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022] Open
Abstract
The galactomannan (GM) that is produced by the human fungal pathogen Aspergillus fumigatus is an emblematic biomarker in medical mycology. The GM is composed of two monosaccharides: mannose and galactofuranose. The furanic configuration of galactose residues, absent in mammals, is responsible for the antigenicity of the GM and has favoured the development of ELISA tests to diagnose aspergillosis in immunocompromised patients. The GM that is produced by A. fumigatus is a unique fungal polysaccharide containing a tetramannoside repeat unit and having three different forms: (i) membrane bound through a glycosylphosphatidylinositol (GPI)-anchor, (ii) covalently linked to β-1,3-glucans in the cell wall, or (iii) released in the culture medium as a free polymer. Recent studies have revealed the crucial role of the GM during vegetative and polarized fungal growth. This review highlights these recent data on its biosynthetic pathway and its biological functions during the saprophytic and pathogenic life of this opportunistic human fungal pathogen.
Collapse
|
35
|
Huang J, Li J, Li Q, Li L, Zhu N, Xiong X, Li G. Peptidoglycan derived from Lactobacillus rhamnosus MLGA up-regulates the expression of chicken β-defensin 9 without triggering an inflammatory response. Innate Immun 2020; 26:733-745. [PMID: 32847443 PMCID: PMC7787553 DOI: 10.1177/1753425920949917] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Defensins are critical components of the innate immune system and play an important role in the integration of innate and adaptive immune responses. Although information on the immunomodulatory properties of peptidoglycan from bacteria is abundant, little is known about the β-defensin induction effect of peptidoglycan from the probiotic Lactobacillus. This study investigated the effect of intact peptidoglycan from L. rhamnosus MLGA on the induction of avian β-defensin 9 in chicken immune cells and intestinal explants. Peptidoglycan from Lactobacillus rhamnosus MLGA dose dependently promoted avian β-defensin 9 mRNA expression in chicken PBMCs, splenocytes, thymocytes, hepatocytes, and chicken embryo jejunum, ileum, and cecum explants and increased the capacity of PBMC or splenocyte lysates to inhibit the growth of Salmonella Enteritidis. In contrast to the effect of L. rhamnosus MLGA-derived peptidoglycan, peptidoglycan derived from pathogenic Staphylococcus aureus reduced avian β-defensin 9 mRNA expression in chicken PBMCs and splenocytes. The inducible effect of peptidoglycan from L. rhamnosus MLGA on avian β-defensin 9 expression in PBMCs and splenocytes was observed without activation of the expression of associated pro-inflammatory cytokines IL-1β, IL-8, and IL-12p40, whereas these cytokine expressions were suppressed by peptidoglycan hydrolysate obtained by lysozyme digestion. The results of the present study show the capability of peptidoglycan derived from L. rhamnosus MLGA to induce the antimicrobial peptide defensin while simultaneously avoiding the deleterious risks of an inflammatory response.
Collapse
Affiliation(s)
- Juan Huang
- Jiangxi Province Key Laboratory of Animal Nutrition, 91595Jiangxi Agricultural University, China
| | - Junhui Li
- Jiangxi Province Key Laboratory of Animal Nutrition, 91595Jiangxi Agricultural University, China
| | - Qiufen Li
- Jiangxi Province Key Laboratory of Animal Nutrition, 91595Jiangxi Agricultural University, China
| | - Lin Li
- Jiangxi Province Key Laboratory of Animal Nutrition, 91595Jiangxi Agricultural University, China
| | - Nianhua Zhu
- Jiangxi Province Key Laboratory of Animal Nutrition, 91595Jiangxi Agricultural University, China
| | - Xiaowen Xiong
- Jiangxi Province Key Laboratory of Animal Nutrition, 91595Jiangxi Agricultural University, China
| | - Guanhong Li
- Jiangxi Province Key Laboratory of Animal Nutrition, 91595Jiangxi Agricultural University, China
| |
Collapse
|
36
|
Baral B, Mozafari MR. Strategic Moves of "Superbugs" Against Available Chemical Scaffolds: Signaling, Regulation, and Challenges. ACS Pharmacol Transl Sci 2020; 3:373-400. [PMID: 32566906 PMCID: PMC7296549 DOI: 10.1021/acsptsci.0c00005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Indexed: 12/12/2022]
Abstract
Superbugs' resistivity against available natural products has become an alarming global threat, causing a rapid deterioration in public health and claiming tens of thousands of lives yearly. Although the rapid discovery of small molecules from plant and microbial origin with enhanced bioactivity has provided us with some hope, a rapid hike in the resistivity of superbugs has proven to be the biggest therapeutic hurdle of all times. Moreover, several distinct mechanisms endowed by these notorious superbugs make them immune to these antibiotics subsequently causing our antibiotic wardrobe to be obsolete. In this unfortunate situation, though the time frame for discovering novel "hit molecules" down the line remains largely unknown, our small hope and untiring efforts injected in hunting novel chemical scaffolds with unique molecular targets using high-throughput technologies may safeguard us against these life-threatening challenges to some extent. Amid this crisis, the current comprehensive review highlights the present status of knowledge, our search for bacteria Achilles' heel, distinct molecular signaling that an opportunistic pathogen bestows to trespass the toxicity of antibiotics, and facile strategies and appealing therapeutic targets of novel drugs. Herein, we also discuss multidimensional strategies to combat antimicrobial resistance.
Collapse
Affiliation(s)
- Bikash Baral
- Department
of Biochemistry, University of Turku, Tykistökatu 6, Turku, Finland
| | - M. R. Mozafari
- Australasian
Nanoscience and Nanotechnology Initiative, 8054 Monash University LPO, Clayton, Victoria 3168, Australia
| |
Collapse
|
37
|
Mycobacterium tuberculosis Rv3717 enhances the survival of Mycolicibacterium smegmatis by inhibiting host innate immune and caspase-dependent apoptosis. INFECTION GENETICS AND EVOLUTION 2020; 84:104412. [PMID: 32531516 DOI: 10.1016/j.meegid.2020.104412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 02/03/2023]
Abstract
Tuberculosis caused by Mycobacterium tuberculosis (M. tuberculosis) infection remains a serious public threat despite decades of creative endeavors. There are few reports on the roles of M. tuberculosis enzymes involved in cell envelope biosynthesis in pathogen survival and persistence. M. tuberculosis Rv3717 encodes N-acetylmuramoyl-l-alanine amidase, a cell-wall hydrolase that hydrolyzes the bond between N-acetylmuramic acid and l-alanine in cell-wall peptidoglycan. In this paper, we demonstrated the Rv3717 promoted the survival of Mycolicibacterium smegmatis(M. smegmatis) within macrophages. More importantly, we demonstrated that this effect is because MS_Rv3717 reduces the release of host pro-inflammatory cytokines such as IL-1β, IL-6, IL-12 p40, TNF-α, and increased transcription of anti-inflammatory cytokine IL-10. At the same time, MS_Rv3717 inhibits apoptosis by inhibiting the activation of Caspase-3/9, reducing the host's elimination of M. smegmatis. Finally, from a bacterial perspective, we found Rv3717 decreased the survival of M. smegmatis under stresses such as SDS and low pH. This is the first report of the involvement of Mycobacterium cell envelope biosynthetic enzyme in host-pathogen interaction.
Collapse
|
38
|
In Vitro Effects of Streptococcus oralis Biofilm on Peri-Implant Soft Tissue Cells. Cells 2020; 9:cells9051226. [PMID: 32429151 PMCID: PMC7290395 DOI: 10.3390/cells9051226] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Human gingival epithelial cells (HGEps) and fibroblasts (HGFs) are the main cell types in peri-implant soft tissue. HGEps are constantly exposed to bacteria, but HGFs are protected by connective tissue as long as the mucosa-implant seal is intact. Streptococcus oralis is one of the commensal bacteria, is highly abundant at healthy implant sites, and might modulate soft tissue cells-as has been described for other streptococci. We have therefore investigated the effects of the S. oralis biofilm on HGEps and HGFs. HGEps or HGFs were grown separately on titanium disks and responded to challenge with S. oralis biofilm. HGFs were severely damaged after 4 h, exhibiting transcriptional inflammatory and stress responses. In contrast, challenge with S. oralis only induced a mild transcriptional inflammatory response in HGEps, without cellular damage. HGFs were more susceptible to the S. oralis biofilm than HGEps. The pro-inflammatory interleukin 6 (IL-6) was attenuated in HGFs, as was interleukin 8 (CXCL8) in HGEps. This indicates that S. oralis can actively protect tissue. In conclusion, commensal biofilms can promote homeostatic tissue protection, but only if the implant-mucosa interface is intact and HGFs are not directly exposed.
Collapse
|
39
|
Abstract
Polysaccharides that contain many sugar monomers include starch and non-starch polysaccharides (NSPs) together with resistant starch (RS). Dietary polysaccharides are well known to have a wide range of biological benefits for bowel health. Gut microbiota and their fermentative products, short chain fatty acids (SCFA), which have recently been highlighted as metabolic regulators, are thought to mediate the function of dietary complex carbohydrates and bowel health. We discuss the influence of various polysaccharides on human bowel health and the mechanisms underlying these effects. We also describe their biological effects on intestinal health and the mechanisms underlying their activity; the polysaccharides were divided into three categories: dietary, microbial, and host-derived polysaccharides. Physiological impacts of non-starch polysaccharides (NSPs) and resistant starch (RS), both of which pass through the small intestine nearly intact and can be fermented by gut microbiota in the large intestine, are similar to each other. They exert a wide range of beneficial effects including anti-inflammation, gut epithelial barrier protection, and immune modulation through both microbiota-dependent and -independent mechanisms. Bacterial polysaccharides usually found in the cell wall generally act as immune modulators, and host-derived polysaccharides not only protect host cells from pathogenic microbial neighbors but also affect overall intestinal health via interactions with gut microbes. Considering these observations, further studies on polysaccharides will be important for bowel health.
Collapse
Affiliation(s)
- Moon Ho Do
- Research Division of Food Functionality, Korea Food Research Institute, Jeollabuk-do, Republic of Korea
| | - Ye Seul Seo
- Research Division of Food Functionality, Korea Food Research Institute, Jeollabuk-do, Republic of Korea
| | - Ho-Young Park
- Research Division of Food Functionality, Korea Food Research Institute, Jeollabuk-do, Republic of Korea
| |
Collapse
|
40
|
Liu Y, Duan Y, Li Y. Integrated Gene Expression Profiling Analysis Reveals Probable Molecular Mechanism and Candidate Biomarker in Anti-TNFα Non-Response IBD Patients. J Inflamm Res 2020; 13:81-95. [PMID: 32104045 PMCID: PMC7024800 DOI: 10.2147/jir.s236262] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
Purpose To explore the molecular mechanism and search for candidate biomarkers in the gene expression profile of IBD patients associated with the response to anti-TNFα agents. Methods Differentially expressed genes (DEGs) of response vs non-response IBD patients in datasets GSE12251, GSE16879, and GSE23597 were integrated using NetworkAnalyst. We conducted functional enrichment analysis of Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and extracted hub genes from the protein–protein interaction network. The proportion of immune cell types was estimated via CIBERSORT. ROC curve analysis and binomial Lasso regression were applied to assess the expression level of hub genes in datasets GSE12251, GSE16879, and GSE23597, and another two datasets GSE107865 and GSE42296. Results A total of 287 DEGs were obtained from the integrated dataset. They were enriched in 14 Gene Ontology terms and 11 KEGG pathways. Polarization from M2 to M1 macrophages was relatively high in non-response individuals. We found nine hub genes (TLR4, TLR1, TLR8, CCR1, CD86, CCL4, HCK, and FCGR2A), mainly related to the interaction between Toll-like Receptor (TLR) pathway and FcγR signaling in non-response anti-TNFα individuals. FCGR2A, HCK, TLR1, TLR4, TLR8, and CCL4 show great value for prediction in intestinal tissue. Besides, FCGR2A, HCK, and TLR8 might be candidate blood biomarkers of anti-TNFα non-response IBD patients. Conclusion Over-activated interaction between FcγR-TLR axis in the innate immune cells of IBD patients might be used to identify non-response individuals and increased our understanding of resistance to anti-TNFα therapy.
Collapse
Affiliation(s)
- Yifan Liu
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, People's Republic of China
| | - Yantao Duan
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, People's Republic of China
| | - Yousheng Li
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, People's Republic of China
| |
Collapse
|
41
|
Yuba E. Development of functional liposomes by modification of stimuli-responsive materials and their biomedical applications. J Mater Chem B 2020; 8:1093-1107. [PMID: 31960007 DOI: 10.1039/c9tb02470k] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Liposomes are a promising nanocarrier for drug delivery because of their biocompatibility and the encapsulation capacity of drugs. Liposomes can be functionalized easily by introduction of functional materials such as stimulus-responsive materials. Temperature-responsive liposomes and pH-responsive liposomes are representative stimulus-responsive liposomes that can deliver drugs to locally heated target tissues and intracellular organelles. Here, temperature-responsive liposomes for the selective release of cargo and pH-responsive liposomes for the induction of antigen-specific immunity are overviewed. Temperature-responsive polymer-modified liposomes immediately released drugs in response to heating, which achieved selective drug release at a tumour after topical heating of tumour-bearing mice. Introduction of MR-detectable molecules enabled the tracing of liposome accumulation into target sites to optimize the heating timing. These liposomes can also be combined with magnetic nanoparticles or carbon nanomaterials to attain magnetic field-responsive, electric field-responsive and light-responsive properties to support on-demand drug release or control of biological reactions using these external stimuli. pH-Responsive liposomes were produced by modification of poly(carboxylic acid) derivatives or by pH-responsive amphiphiles. These liposomes delivered antigenic proteins into the cytosol of antigen presenting cells, which induced cross-presentation and antigen-specific cellular immunity. Adjuvant molecules or bioactive polysaccharide-based pH-responsive polymers improved their immunity-inducing effect further, leading to tumour regression in tumour-bearing mice. Precise design and control of the structures of stimulus-responsive materials and combination with functional materials are expected to create novel methodologies to control biological functions and to produce highly potent liposomal drugs that can achieve selective release of bioactive molecules.
Collapse
Affiliation(s)
- Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan.
| |
Collapse
|
42
|
Campanero-Rhodes MA, Palma AS, Menéndez M, Solís D. Microarray Strategies for Exploring Bacterial Surface Glycans and Their Interactions With Glycan-Binding Proteins. Front Microbiol 2020; 10:2909. [PMID: 32010066 PMCID: PMC6972965 DOI: 10.3389/fmicb.2019.02909] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/03/2019] [Indexed: 12/14/2022] Open
Abstract
Bacterial surfaces are decorated with distinct carbohydrate structures that may substantially differ among species and strains. These structures can be recognized by a variety of glycan-binding proteins, playing an important role in the bacteria cross-talk with the host and invading bacteriophages, and also in the formation of bacterial microcolonies and biofilms. In recent years, different microarray approaches for exploring bacterial surface glycans and their recognition by proteins have been developed. A main advantage of the microarray format is the inherent miniaturization of the method, which allows sensitive and high-throughput analyses with very small amounts of sample. Antibody and lectin microarrays have been used for examining bacterial glycosignatures, enabling bacteria identification and differentiation among strains. In addition, microarrays incorporating bacterial carbohydrate structures have served to evaluate their recognition by diverse host/phage/bacterial glycan-binding proteins, such as lectins, effectors of the immune system, or bacterial and phagic cell wall lysins, and to identify antigenic determinants for vaccine development. The list of samples printed in the arrays includes polysaccharides, lipopoly/lipooligosaccharides, (lipo)teichoic acids, and peptidoglycans, as well as sequence-defined oligosaccharide fragments. Moreover, microarrays of cell wall fragments and entire bacterial cells have been developed, which also allow to study bacterial glycosylation patterns. In this review, examples of the different microarray platforms and applications are presented with a view to give the current state-of-the-art and future prospects in this field.
Collapse
Affiliation(s)
- María Asunción Campanero-Rhodes
- Instituto de Química Física Rocasolano, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Angelina Sa Palma
- UCIBIO, Department of Chemistry, Faculty of Science and Technology, NOVA University of Lisbon, Lisbon, Portugal
| | - Margarita Menéndez
- Instituto de Química Física Rocasolano, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Dolores Solís
- Instituto de Química Física Rocasolano, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
43
|
Del Cornò M, Gessani S, Conti L. Shaping the Innate Immune Response by Dietary Glucans: Any Role in the Control of Cancer? Cancers (Basel) 2020; 12:cancers12010155. [PMID: 31936360 PMCID: PMC7016572 DOI: 10.3390/cancers12010155] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
β-glucans represent a heterogeneous group of naturally occurring and biologically active polysaccharides found in many kinds of edible mushrooms, baker’s yeast, cereals and seaweeds, whose health-promoting effects have been known since ancient times. These compounds can be taken orally as food supplements or as part of daily diets, and are safe to use, nonimmunogenic and well tolerated. A main feature of β-glucans is their capacity to function as biological response modifiers, exerting regulatory effects on inflammation and shaping the effector functions of different innate and adaptive immunity cell populations. The potential to interfere with processes involved in the development or control of cancer makes β-glucans interesting candidates as adjuvants in antitumor therapies as well as in cancer prevention strategies. Here, the regulatory effects of dietary β-glucans on human innate immunity cells are reviewed and their potential role in cancer control is discussed.
Collapse
|
44
|
Souza ACO, Favali C, Soares NC, Tavares NM, Jerônimo MS, Veloso Junior PH, Marina CL, Santos C, Brodskyn C, Bocca AL. New Role of P. brasiliensis α-Glucan: Differentiation of Non-conventional Dendritic Cells. Front Microbiol 2019; 10:2445. [PMID: 31736892 PMCID: PMC6833476 DOI: 10.3389/fmicb.2019.02445] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/11/2019] [Indexed: 01/22/2023] Open
Abstract
The cell wall has a critical role in the host immune response to fungal pathogens. In this study, we investigated the influence of two cell wall fractions of the dimorphic fungi Paracoccidioides brasiliensis (Pb) in the in vitro generation of monocyte-derived dendritic cells (MoDCs). Monocytes were purified from the peripheral blood of healthy donors and cultivated for 7 days in medium supplemented with IL-4 and GM-CSF in the presence of Pb cell wall fractions: the alkali-insoluble F1, constituted by β-1,3-glucans, chitin and proteins, and the alkali-soluble F2, mainly constituted by α-glucan. MoDCs phenotypes were evaluated regarding cell surface expression of CD1a, DC-SIGN, HLA-DR, CD80, and CD83 and production of cytokines. The α-glucan-rich cell wall fraction downregulated the differentiation of CD1a+ MoDCs, a dendritic cell subset that stimulate Th1 responses. The presence of both cell fractions inhibited DC-SIGN and HLA-DR expression, while the expression of maturation markers was differentially induced in CD1a– MoDCs. Differentiation upon F1 and F2 stimulation induced mixed profile of inflammatory cytokines. Altogether, these data demonstrate that Pb cell wall fractions differentially induce a dysregulation in DCs differentiation. Moreover, our results suggest that cell wall α-glucan promote the differentiation of CD1a– DCs, potentially favoring Th2 polarization and contributing to pathogen persistence.
Collapse
Affiliation(s)
| | - Cecília Favali
- Departamento de Biologia Celular, Universidade de Brasília, Brasília, Brazil
| | | | | | | | | | - Clara Luna Marina
- Departamento de Biologia Celular, Universidade de Brasília, Brasília, Brazil
| | - Claire Santos
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Cláudia Brodskyn
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | | |
Collapse
|
45
|
van Thiel IAM, Botschuijver S, de Jonge WJ, Seppen J. Painful interactions: Microbial compounds and visceral pain. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165534. [PMID: 31634534 DOI: 10.1016/j.bbadis.2019.165534] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 12/18/2022]
Abstract
Visceral pain, characterized by abdominal discomfort, originates from organs in the abdominal cavity and is a characteristic symptom in patients suffering from irritable bowel syndrome, vulvodynia or interstitial cystitis. Most organs in which visceral pain originates are in contact with the external milieu and continuously exposed to microbes. In order to maintain homeostasis and prevent infections, the immune- and nervous system in these organs cooperate to sense and eliminate (harmful) microbes. Recognition of microbial components or products by receptors expressed on cells from the immune and nervous system can activate immune responses but may also cause pain. We review the microbial compounds and their receptors that could be involved in visceral pain development.
Collapse
Affiliation(s)
- I A M van Thiel
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, location AMC, Meibergdreef 69, 1105 BK Amsterdam, the Netherlands
| | - S Botschuijver
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, location AMC, Meibergdreef 69, 1105 BK Amsterdam, the Netherlands
| | - W J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, location AMC, Meibergdreef 69, 1105 BK Amsterdam, the Netherlands
| | - J Seppen
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, location AMC, Meibergdreef 69, 1105 BK Amsterdam, the Netherlands.
| |
Collapse
|
46
|
Torrens G, Escobar-Salom M, Pol-Pol E, Camps-Munar C, Cabot G, López-Causapé C, Rojo-Molinero E, Oliver A, Juan C. Comparative Analysis of Peptidoglycans From Pseudomonas aeruginosa Isolates Recovered From Chronic and Acute Infections. Front Microbiol 2019; 10:1868. [PMID: 31507543 PMCID: PMC6719521 DOI: 10.3389/fmicb.2019.01868] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 07/29/2019] [Indexed: 12/20/2022] Open
Abstract
Pseudomonas aeruginosa is one of the first causes of acute nosocomial and chronic infections in patients with underlying respiratory pathologies such as cystic fibrosis (CF). It has been proposed that P. aeruginosa accumulates mutations driving to peptidoglycan modifications throughout the development of the CF-associated infection, as a strategy to lower the immune detection hence ameliorating the chronic persistence. As well, some studies dealing with peptidoglycan modifications driving to a better survival within the host have been published in other gram-negatives. According to these facts, the gram-negative peptidoglycan could be considered as a pathogen-associated molecular pattern with very important implications regarding the host’s detection-response, worthy to dissect in detail. For this reason, in this work we characterized for the first time the peptidoglycans of three large collections [early CF, late CF and acute infection (bloodstream) P. aeruginosa strains] from qualitative (HPLC), quantitative and inflammatory capacity-related perspectives. The final goal was to identify composition trends potentially supporting the cited strategy of evasion/resistance to the immune system and providing information regarding the differential intrinsic adaptation depending on the type of infection. Although we found several punctual strain-specific particularities, our results indicated a high degree of inter-collection uniformity in the peptidoglycan-related features and the absence of trends amongst the strains studied here. These results suggest that the peptidoglycan of P. aeruginosa is a notably conserved structure in natural isolates regardless of transitory changes that some external conditions could force. Finally, the inverse correlation between the relative amount of stem pentapeptides within the murein sacculus and the resistance to immune lytic attacks against the peptidoglycan was also suggested by our results. Altogether, this work is a major step ahead to understand the biology of peptidoglycan from P. aeruginosa natural strains, hopefully useful in future for therapeutic alternatives design.
Collapse
Affiliation(s)
- Gabriel Torrens
- Servicio de Microbiología-Unidad de Investigación, Hospital Universitari Son Espases-Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - María Escobar-Salom
- Servicio de Microbiología-Unidad de Investigación, Hospital Universitari Son Espases-Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Elisabet Pol-Pol
- Servicio de Microbiología-Unidad de Investigación, Hospital Universitari Son Espases-Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Cristina Camps-Munar
- Servicio de Microbiología-Unidad de Investigación, Hospital Universitari Son Espases-Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Gabriel Cabot
- Servicio de Microbiología-Unidad de Investigación, Hospital Universitari Son Espases-Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Carla López-Causapé
- Servicio de Microbiología-Unidad de Investigación, Hospital Universitari Son Espases-Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Estrella Rojo-Molinero
- Servicio de Microbiología-Unidad de Investigación, Hospital Universitari Son Espases-Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Antonio Oliver
- Servicio de Microbiología-Unidad de Investigación, Hospital Universitari Son Espases-Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Carlos Juan
- Servicio de Microbiología-Unidad de Investigación, Hospital Universitari Son Espases-Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| |
Collapse
|
47
|
Brott AS, Clarke AJ. Peptidoglycan O-Acetylation as a Virulence Factor: Its Effect on Lysozyme in the Innate Immune System. Antibiotics (Basel) 2019; 8:E94. [PMID: 31323733 PMCID: PMC6783866 DOI: 10.3390/antibiotics8030094] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/11/2019] [Accepted: 07/13/2019] [Indexed: 11/16/2022] Open
Abstract
The peptidoglycan sacculus of both Gram-positive and Gram-negative bacteria acts as a protective mesh and provides structural support around the entirety of the cell. The integrity of this structure is of utmost importance for cell viability and so naturally is the first target for attack by the host immune system during bacterial infection. Lysozyme, a muramidase and the first line of defense of the innate immune system, targets the peptidoglycan sacculus hydrolyzing the β-(1→4) linkage between repeating glycan units, causing lysis and the death of the invading bacterium. The O-acetylation of N-acetylmuramoyl residues within peptidoglycan precludes the productive binding of lysozyme, and in doing so renders it inactive. This modification has been shown to be an important virulence factor in pathogens such as Staphylococcus aureus and Neisseria gonorrhoeae and is currently being investigated as a novel target for anti-virulence therapies. This article reviews interactions made between peptidoglycan and the host immune system, specifically with respect to lysozyme, and how the O-acetylation of the peptidoglycan interrupts these interactions, leading to increased pathogenicity.
Collapse
Affiliation(s)
- Ashley S Brott
- Department of Molecular & Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Anthony J Clarke
- Department of Molecular & Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
48
|
Morse DJ, Smith A, Wilson MJ, Marsh L, White L, Posso R, Bradshaw DJ, Wei X, Lewis MAO, Williams DW. Molecular community profiling of the bacterial microbiota associated with denture-related stomatitis. Sci Rep 2019; 9:10228. [PMID: 31308427 PMCID: PMC6629705 DOI: 10.1038/s41598-019-46494-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/27/2019] [Indexed: 12/12/2022] Open
Abstract
Denture-associated stomatitis (DS) affects over two-thirds of denture-wearers. DS presents as erythema of the palatal mucosa in areas where denture-surface associated polymicrobial biofilms containing the fungus Candida albicans exist. The contribution of the oral bacterial microbiota toward the infection is unknown. Therefore, this study characterised the bacterial microbiota of sites within the oral cavity to identify potential associations with occurrence of DS. Denture-wearing patients were recruited (denture stomatitis (DS) n = 8; non-denture stomatitis (NoDS) n = 11) and the oral bacterial microbiota of the tongue, palate and denture-fitting surface was characterised using next-generation sequencing. Operational taxonomic units (OTUs) were identified to bacterial genera and species, and presence/absence and relative abundances were examined. A significant (P = 0.007) decrease in the number of OTUs and thus, diversity of the microbiota was observed in tongue samples of DS patients (vs non-DS). The microbiota of denture-fitting surfaces and palatal mucosae were similar. Large differences in the abundance of bacterial genera and species were observed at each sample site, and unique presence/absence of bacteria was noted. Presence/absence and relative abundance of specific bacteria associated with DS warrants further in vitro and in vivo evaluation, particularly as our previous work has shown C. albicans virulence factor modulation by oral bacteria.
Collapse
Affiliation(s)
- Daniel J Morse
- Microbiomes, Microbes and Informatics Group, School of Biosciences, Cardiff University, Cardiff, UK.
| | - Ann Smith
- School of Medicine, Cardiff University, Cardiff, UK
| | - Melanie J Wilson
- Oral and Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff, UK
| | - Lucy Marsh
- School of Medicine, Cardiff University, Cardiff, UK
| | - Lewis White
- UKCMN Regional Mycology Reference Laboratory, Public Health Wales, Microbiology Cardiff, Cardiff, UK
| | - Raquel Posso
- UKCMN Regional Mycology Reference Laboratory, Public Health Wales, Microbiology Cardiff, Cardiff, UK
| | | | - Xiaoqing Wei
- Oral and Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff, UK
| | - Michael A O Lewis
- Oral and Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff, UK
| | - David W Williams
- Oral and Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff, UK
| |
Collapse
|
49
|
Abstract
The evolutionary separated Gram-negative Chlamydiales show a biphasic life cycle and replicate exclusively within eukaryotic host cells. Members of the genus Chlamydia are responsible for many acute and chronic diseases in humans, and Chlamydia-related bacteria are emerging pathogens. We revisit past efforts to detect cell wall material in Chlamydia and Chlamydia-related bacteria in the context of recent breakthroughs in elucidating the underlying cellular and molecular mechanisms of the chlamydial cell wall biosynthesis. In this review, we also discuss the role of cell wall biosynthesis in chlamydial FtsZ-independent cell division and immune modulation. In the past, penicillin susceptibility of an invisible wall was referred to as the "chlamydial anomaly." In light of new mechanistic insights, chlamydiae may now emerge as model systems to understand how a minimal and modified cell wall biosynthetic machine supports bacterial cell division and how cell wall-targeting beta-lactam antibiotics can also act bacteriostatically rather than bactericidal. On the heels of these discussions, we also delve into the effects of other cell wall antibiotics in individual chlamydial lineages.
Collapse
|
50
|
Brott AS, Jones CS, Clarke AJ. Development of a High Throughput Screen for the Identification of Inhibitors of Peptidoglycan O-Acetyltransferases, New Potential Antibacterial Targets. Antibiotics (Basel) 2019; 8:E65. [PMID: 31137799 PMCID: PMC6627197 DOI: 10.3390/antibiotics8020065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/22/2019] [Accepted: 05/22/2019] [Indexed: 12/02/2022] Open
Abstract
The O-acetylation of peptidoglycan occurs in many Gram-negative and most Gram-positive pathogens and this modification to the essential wall polymer controls the lytic activity of the autolysins, particularly the lytic transglycosylases, and inhibits that of the lysozymes of innate immunity systems. As such, the peptidoglycan O-acetyltransferases PatA/B and OatA are recognized as virulence factors. In this study, we present the high throughput screening of small compound libraries to identify the first known inhibitors of these enzymes. The fluorometric screening assay developed involved monitoring the respective O-acetyltransferases as esterases using 4-methylumbelliferylacetate as substrate. Pilot screens of 3921 compounds validated the usefulness of the HTS protocol. A number of potential inhibitors were identified amongst a total of 145,000 low molecular-weight compounds, some of which were common to both enzymes, while others were unique to each. After eliminating a number of false positives in secondary screens, dose response curves confirmed the apparent specificity of a benzothiazolyl-pyrazolo-pyridine as an inhibitor of Neisseria gonorrhoeae PatB, and several coumarin-based compounds as inhibitors of both this PatB and OatA from Staphylococcus aureus. The benzothiazolyl-pyrazolo-pyridine was determined to be a non-competitive inhibitor of PatB with a Ki of 126 µM. At 177 µg/mL and close to its solubility limit, this compound caused a 90% reduction in growth of N. gonorrhoeae, while growth of Escherichia coli, a bacterium that lacks PatB and, hence, does not produce O-acetylated peptidoglycan, was unaffected. These data provide preliminary proof of concept that peptidoglycan O-acetyltransferases would serve as useful antibacterial targets.
Collapse
Affiliation(s)
- Ashley S Brott
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Carys S Jones
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Anthony J Clarke
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|