1
|
Weber B, Ritchie NE, Hilker S, Chan DCK, Peukert C, Deisinger JP, Ives R, Årdal C, Burrows LL, Brönstrup M, Magolan J, Raivio TL, Brown ED. High-Throughput Discovery of Synthetic Siderophores for Trojan Horse Antibiotics. ACS Infect Dis 2024; 10:3821-3841. [PMID: 39438291 PMCID: PMC11556397 DOI: 10.1021/acsinfecdis.4c00359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
To cause infection, bacterial pathogens must overcome host immune factors and barriers to nutrient acquisition. Reproducing these aspects of host physiology in vitro has shown great promise for antibacterial drug discovery. When used as a bacterial growth medium, human serum replicates several aspects of the host environment, including innate immunity and iron limitation. We previously reported that a high-throughput chemical screen using serum as the growth medium enabled the discovery of novel growth inhibitors overlooked by conventional screens. Here, we report that a subset of compounds from this high-throughput serum screen display an unexpected growth enhancing phenotype and are enriched for synthetic siderophores. We selected 35 compounds of diverse chemical structure and quantified their ability to enhance bacterial growth in human serum. We show that many of these compounds chelate iron, suggesting they were acting as siderophores and providing iron to the bacteria. For two different pharmacophores represented among these synthetic siderophores, conjugation to the β-lactam antibiotic ampicillin imparted iron-dependent enhancement in antibacterial activity. Conjugation of the most potent growth-enhancing synthetic siderophore with the monobactam aztreonam produced MLEB-22043, a broad-spectrum antibiotic with significantly improved activity against Klebsiella pneumoniae, Escherichia coli, Acinetobacter baumannii, and Pseudomonas aeruginosa. This synthetic siderophore-monobactam conjugate uses multiple TonB-dependent transporters for uptake into P. aeruginosa. Like aztreonam, MLEB-22043 demonstrated activity against metallo-β-lactamase expressing bacteria, and, when combined with the β-lactamase inhibitor avibactam, was active against clinical strains coexpressing the NDM-1 metallo-β-lactamase and serine β-lactamases. Our work shows that human serum is an effective bacterial growth medium for the high-throughput discovery of synthetic siderophores, enabling the development of novel Trojan Horse antibiotics.
Collapse
Affiliation(s)
- Brent
S. Weber
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
- Department
of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Nikki E. Ritchie
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| | - Simon Hilker
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Derek C. K. Chan
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| | - Carsten Peukert
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Julia P. Deisinger
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| | - Rowan Ives
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| | - Christine Årdal
- Antimicrobial
Resistance Centre, Norwegian Institute of
Public Health, 0213 Oslo, Norway
| | - Lori L. Burrows
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| | - Mark Brönstrup
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research Inhoffenstraße 7, 38124 Braunschweig, Germany
- German
Center for Infection Research (DZIF), Site
Hannover-Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany
- Institute
for Organic Chemistry (IOC), Leibniz Universität
Hannover, Schneiderberg
1B, 30167 Hannover, Germany
| | - Jakob Magolan
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| | - Tracy L. Raivio
- Department
of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Eric D. Brown
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Michael
G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S
4L8, Canada
| |
Collapse
|
2
|
Li X, Cheng L, Liu X, Wang X, Li R, Fan S, Yan Q, Ma T, Ma Y, Kang J. Dopamine promotes Klebsiella quasivariicola proliferation and inflammatory response in the presence of macrophages. Front Cell Infect Microbiol 2024; 14:1322113. [PMID: 38585654 PMCID: PMC10995343 DOI: 10.3389/fcimb.2024.1322113] [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: 10/15/2023] [Accepted: 03/04/2024] [Indexed: 04/09/2024] Open
Abstract
Background Dopamine, a frequently used therapeutic agent for critically ill patients, has been shown to be implicated in clinical infections recently, however, the precise mechanisms underlying this association remain elusive. Klebsiella quasivariicola, a novel strain belonging to the Klebsiella species, exhibits potential pathogenic attributes. The impact of dopamine on K. quasivariicola infection has aroused our interest. Objective Considering the contribution of host immune factors during infection, this study aimed to investigate the intricate interactions between K. quasivariicola, dopamine, and macrophages were explored. Methods RAW264.7 cells and C57/BL6 mice were infected with K. quasivariicola, and the bacterial growth within macrophage, the production of inflammatory cytokines and the pathological changes in mice lungs were detected, in the absence or presence of dopamine. Results Dopamine inhibited the growth of K. quasivariicola in the medium, but promoted bacterial growth when co-cultured with macrophages. The expression of proinflammatory cytokines increased in RAW 264.7 cells infected with K. quasivariicola, and a significant rise was observed upon the addition of dopamine. The infection of K. quasivariicola in mice induced an inflammatory response and lung injury, which were exacerbated by the administration of dopamine. Conclusions Our findings suggest that dopamine may be one of the potential risk factors associated with K. quasivariicola infection. This empirical insight provides solid references for clinical precision medicine. Furthermore, an in vitro model of microbes-drugs-host immune cells for inhibitor screening was proposed to more accurately replicate the complex in vivo environment. This fundamental work had contributed to the present understanding of the crosstalk between pathogen, dopamine and host immune cells.
Collapse
Affiliation(s)
- Xiang Li
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
- Department of Pathology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Lin Cheng
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xueyang Liu
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoli Wang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Rui Li
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Shao Fan
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Qiulong Yan
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Tonghui Ma
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yufang Ma
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jian Kang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
3
|
Weigert Muñoz A, Zhao W, Sieber SA. Monitoring host-pathogen interactions using chemical proteomics. RSC Chem Biol 2024; 5:73-89. [PMID: 38333198 PMCID: PMC10849124 DOI: 10.1039/d3cb00135k] [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: 07/25/2023] [Accepted: 11/09/2023] [Indexed: 02/10/2024] Open
Abstract
With the rapid emergence and the dissemination of microbial resistance to conventional chemotherapy, the shortage of novel antimicrobial drugs has raised a global health threat. As molecular interactions between microbial pathogens and their mammalian hosts are crucial to establish virulence, pathogenicity, and infectivity, a detailed understanding of these interactions has the potential to reveal novel therapeutic targets and treatment strategies. Bidirectional molecular communication between microbes and eukaryotes is essential for both pathogenic and commensal organisms to colonise their host. In particular, several devastating pathogens exploit host signalling to adjust the expression of energetically costly virulent behaviours. Chemical proteomics has emerged as a powerful tool to interrogate the protein interaction partners of small molecules and has been successfully applied to advance host-pathogen communication studies. Here, we present recent significant progress made by this approach and provide a perspective for future studies.
Collapse
Affiliation(s)
- Angela Weigert Muñoz
- Center for Functional Protein Assemblies, Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich Ernst-Otto-Fischer-Straße 8 D-85748 Garching Germany
| | - Weining Zhao
- College of Pharmacy, Shenzhen Technology University Shenzhen 518118 China
| | - Stephan A Sieber
- Center for Functional Protein Assemblies, Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich Ernst-Otto-Fischer-Straße 8 D-85748 Garching Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Germany
| |
Collapse
|
4
|
Reséndiz-Albor AA, Arciniega-Martínez IM, Montes de Oca AC, Guzmán-Mejía F, Drago-Serrano ME, Estrada-Jiménez T, Abarca-Rojano E. Outcome of stress on G protein-coupled receptors and hypoxia inducible factor-1α. J Med Life 2024; 17:201-204. [PMID: 38813364 PMCID: PMC11131643 DOI: 10.25122/jml-2023-0363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/07/2024] [Indexed: 05/31/2024] Open
Abstract
Stress drives neuroendocrine signals with detrimental effects to the intestinal homeostasis. The aim of this study was to evaluate the effect of stress on intestinal hypoxia response elements, including G protein-coupled receptor 41 (GPR41), GPR43, and hypoxia inducible factor (HIF)-1α. Groups of five BALB/c mice were subjected to acute (2 h per day) and chronic (2 h per day for 4 days) stress induced by restraint, and the results were compared to those of an unstressed control group. Whole mucosal samples from the colon were collected to evaluate the expression of GPR41, GPR43 and HIF-1α using Western blot chemiluminescent analysis. Compared to the control group, in the chronic stress group the expression of GPR43 (P = 0.0092) and HIF-1α (P < 0.0001) were significantly lower and the expression of GPR41 was similar (P = 0.9184); acute stress significantly increased HIF-1α expression (P = 0.0030) and increased GPR41 expression (P = 0.0937), without affecting GPR43 (P = 0.9184). These findings offer insights into the modulation of hypoxia response elements under stress conditions and their pharmacological implications for developing drugs that mitigate the effects of stress on intestinal homeostasis.
Collapse
Affiliation(s)
- Aldo Arturo Reséndiz-Albor
- Laboratorio de Inmunidad de Mucosas, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Ciudad de México, México
| | - Ivonne Maciel Arciniega-Martínez
- Laboratorio de Inmunidad de Mucosas, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Ciudad de México, México
| | - Arturo Contis Montes de Oca
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina Instituto Politecnico Nacional Plan de San Luis y Salvador Diaz Miron, Ciudad de México, México
| | - Fabiola Guzmán-Mejía
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, Ciudad de México, México
| | - Maria Elisa Drago-Serrano
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, Ciudad de México, México
| | - Tania Estrada-Jiménez
- Facultad de Medicina, Decanato de Ciencias Médicas, Universidad Popular Autónoma de Estado de Puebla, Ciudad de México, México
| | - Edgar Abarca-Rojano
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina Instituto Politecnico Nacional Plan de San Luis y Salvador Diaz Miron, Ciudad de México, México
| |
Collapse
|
5
|
Purtov YA, Ozoline ON. Neuromodulators as Interdomain Signaling Molecules Capable of Occupying Effector Binding Sites in Bacterial Transcription Factors. Int J Mol Sci 2023; 24:15863. [PMID: 37958845 PMCID: PMC10647483 DOI: 10.3390/ijms242115863] [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: 09/30/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Hormones and neurotransmitters are important components of inter-kingdom signaling systems that ensure the coexistence of eukaryotes with their microbial community. Their ability to affect bacterial physiology, metabolism, and gene expression was evidenced by various experimental approaches, but direct penetration into bacteria has only recently been reported. This opened the possibility of considering neuromodulators as potential effectors of bacterial ligand-dependent regulatory proteins. Here, we assessed the validity of this assumption for the neurotransmitters epinephrine, dopamine, and norepinephrine and two hormones (melatonin and serotonin). Using flexible molecular docking for transcription factors with ligand-dependent activity, we assessed the ability of neuromodulators to occupy their effector binding sites. For many transcription factors, including the global regulator of carbohydrate metabolism, CRP, and the key regulator of lactose assimilation, LacI, this ability was predicted based on the analysis of several 3D models. By occupying the ligand binding site, neuromodulators can sterically hinder the interaction of the target proteins with the natural effectors or even replace them. The data obtained suggest that the direct modulation of the activity of at least some bacterial transcriptional factors by neuromodulators is possible. Therefore, the natural hormonal background may be a factor that preadapts bacteria to the habitat through direct perception of host signaling molecules.
Collapse
Affiliation(s)
- Yuri A. Purtov
- Department of Functional Genomics of Prokaryotes, Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino 142290, Russia
| | - Olga N. Ozoline
- Department of Functional Genomics of Prokaryotes, Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino 142290, Russia
| |
Collapse
|
6
|
Elfaky MA, Thabit AK, Eljaaly K, Zawawi A, Abdelkhalek AS, Almalki AJ, Ibrahim TS, Hegazy WAH. Controlling of Bacterial Virulence: Evaluation of Anti-Virulence Activities of Prazosin against Salmonella enterica. Antibiotics (Basel) 2022; 11:1585. [PMID: 36358239 PMCID: PMC9686722 DOI: 10.3390/antibiotics11111585] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 08/10/2023] Open
Abstract
Salmonella enterica is a Gram-negative orofecal transmitted pathogen that causes a wide diversity of local and systemic illnesses. Salmonella enterica utilizes several interplayed systems to regulate its invasion and pathogenesis: namely, quorum sensing (QS) and type three secretion system (T3SS). In addition, S. enterica could sense the adrenergic hormones in the surroundings that enhance its virulence. The current study aimed to evaluate the ability of α-adrenoreceptor antagonist prazosin to mitigate the virulence of S. enterica serovar Typhimurium. The prazosin effect on biofilm formation and the expression of sdiA, qseC, qseE, and T3SS-type II encoding genes was evaluated. Furthermore, the prazosin intracellular replication inside macrophage and anti-virulence activity was evaluated in vivo against S. typhimurium. The current finding showed a marked prazosin ability to compete on SdiA and QseC and downregulate their encoding genes. Prazosin significantly downregulated the virulence factors encoding genes and diminished the biofilm formation, intracellular replication inside macrophages, and in vivo protected mice. To sum up, prazosin showed significant inhibitory activities against QS, T3SS, and bacterial espionage, which documents its considered anti-virulence activities.
Collapse
Affiliation(s)
- Mahmoud A. Elfaky
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abrar K. Thabit
- Pharmacy Practice Department, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Khalid Eljaaly
- Pharmacy Practice Department, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ayat Zawawi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ahmed S. Abdelkhalek
- Medicinal Chemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Ahmad J. Almalki
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Tarek S. Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Wael A. H. Hegazy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
- Pharmacy Program, Department of Pharmaceutical Sciences, Oman College of Health Sciences, Muscat 113, Oman
| |
Collapse
|
7
|
Cavalu S, Elbaramawi SS, Eissa AG, Radwan MF, S. Ibrahim T, Khafagy ES, Lopes BS, Ali MAM, Hegazy WAH, Elfaky MA. Characterization of the Anti-Biofilm and Anti-Quorum Sensing Activities of the β-Adrenoreceptor Antagonist Atenolol against Gram-Negative Bacterial Pathogens. Int J Mol Sci 2022; 23:13088. [PMID: 36361877 PMCID: PMC9656717 DOI: 10.3390/ijms232113088] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 08/10/2023] Open
Abstract
The development of bacterial resistance to antibiotics is an increasing public health issue that worsens with the formation of biofilms. Quorum sensing (QS) orchestrates the bacterial virulence and controls the formation of biofilm. Targeting bacterial virulence is promising approach to overcome the resistance increment to antibiotics. In a previous detailed in silico study, the anti-QS activities of twenty-two β-adrenoreceptor blockers were screened supposing atenolol as a promising candidate. The current study aims to evaluate the anti-QS, anti-biofilm and anti-virulence activities of the β-adrenoreceptor blocker atenolol against Gram-negative bacteria Serratia marcescens, Pseudomonas aeruginosa, and Proteus mirabilis. An in silico study was conducted to evaluate the binding affinity of atenolol to S. marcescens SmaR QS receptor, P. aeruginosa QscR QS receptor, and P. mirabilis MrpH adhesin. The atenolol anti-virulence activity was evaluated against the tested strains in vitro and in vivo. The present finding shows considerable ability of atenolol to compete with QS proteins and significantly downregulated the expression of QS- and virulence-encoding genes. Atenolol showed significant reduction in the tested bacterial biofilm formation, virulence enzyme production, and motility. Furthermore, atenolol significantly diminished the bacterial capacity for killing and protected mice. In conclusion, atenolol has potential anti-QS and anti-virulence activities against S. marcescens, P. aeruginosa, and P. mirabilis and can be used as an adjuvant in treatment of aggressive bacterial infections.
Collapse
Affiliation(s)
- Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| | - Samar S. Elbaramawi
- Medicinal Chemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Ahmed G. Eissa
- Medicinal Chemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mohamed F. Radwan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Tarek S. Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - El-Sayed Khafagy
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj 11942, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Bruno Silvester Lopes
- School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BA, UK
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| | - Mohamed A. M. Ali
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia
- Department of Biochemistry, Faculty of Science, Ain Shams University, Abbassia, Cairo 11566, Egypt
| | - Wael A. H. Hegazy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
- Pharmacy Program, Department of Pharmaceutical Sciences, Oman College of Health Sciences, Muscat 113, Oman
| | - Mahmoud A. Elfaky
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
8
|
Laboratory Grown Biofilms of Bacteria Associated with Human Atherosclerotic Carotid Arteries Release Collagenases and Gelatinases during Iron-Induced Dispersion. Microbiol Spectr 2022; 10:e0100121. [PMID: 35543563 PMCID: PMC9241811 DOI: 10.1128/spectrum.01001-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The association of bacteria with arterial plaque lesions in patients with atherosclerosis has been widely reported. However, the role these bacteria play in the progression of atherosclerosis is still unclear. Previous work in our lab has demonstrated that bacteria exist in carotid artery plaques as biofilm deposits. Biofilms are communities of microorganisms enmeshed within a protective, self-produced extracellular matrix and have been shown to contribute to chronic infections in humans. Biofilm communities have the potential to impact surrounding tissues in an infection if they undergo a dispersion response, releasing bacteria into the surrounding environment by enzymatic degradation of the extracellular matrix. One concern relating to these enzymes is that they could cause collateral damage to host tissues. In this study, we present an in vitro multispecies biofilm culturing model used to investigate the potential role of bacterial biofilm dispersion in the progression of atherosclerosis. This work has demonstrated an increase in cell release from mixed-species biofilms formed by bacteria associated with human carotid arterial plaque deposits following treatment with iron or a combination of norepinephrine and transferrin. Greater extracellular lipase, protease, and collagenase/gelatinase activity was also associated with iron-treated biofilms. The results of this work suggest that bacteria in this model undergo iron-induced biofilm dispersion, as evidenced by the increased cell release and higher enzyme activity following treatment. This work demonstrates the potential for multispecies biofilm dispersion to contribute to arterial tissue degradation by bacteria and suggests that in atherosclerotic infections, biofilm dispersion may contribute to thrombogenesis, which can lead to heart attack or stroke. IMPORTANCE Atherosclerosis, or hardening of the arteries, is a leading cause of congestive heart failure, heart attack, and stroke in humans. Mounting evidence, in the literature and from our lab, points to the regular involvement of bacteria within arterial plaque deposits in patients with advanced atherosclerosis. Very little is known about the behavior of these bacteria and whether they may contribute to tissue damage in infected arteries. Tissue damage within the arterial plaque lesion can lead to rupture of the plaque contents into the bloodstream, where a clot may form, resulting in a potential heart attack or stroke. This study shows that plaque-associated bacteria, when cultured as mixed-species biofilms in the laboratory, can release degradative enzymes into their environment as the result of a dispersion response triggered by iron. These degradative enzymes can digest proteins and lipids which are associated with the tissues that separate the plaque lesion from the arterial lumen. Thus, this study demonstrates that if mixed species biofilms are induced to undergo dispersion in an infected atherosclerotic lesion when exposed to an elevated concentration of free iron, they have the potential to contribute to the weakening of arterial tissues, which may contribute to atherosclerotic plaque destabilization.
Collapse
|
9
|
In vivo growth of Staphylococcus lugdunensis is facilitated by the concerted function of heme and non-heme iron acquisition mechanisms. J Biol Chem 2022; 298:101823. [PMID: 35283192 PMCID: PMC9052147 DOI: 10.1016/j.jbc.2022.101823] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus lugdunensis has increasingly been recognized as a pathogen that can cause serious infection indicating this bacterium overcomes host nutritional immunity. Despite this, there exists a significant knowledge gap regarding the iron acquisition mechanisms employed by S. lugdunensis, especially during infection of the mammalian host. Here we show that S. lugdunensis can usurp hydroxamate siderophores and staphyloferrin A and B from Staphylococcus aureus. These transport activities all required a functional FhuC ATPase. Moreover, we show that the acquisition of catechol siderophores and catecholamine stress hormones by S. lugdunensis required the presence of the sst-1 transporter-encoding locus, but not the sst-2 locus. Iron-dependent growth in acidic culture conditions necessitated the ferrous iron transport system encoded by feoAB. Heme iron was acquired via expression of the iron-regulated surface determinant (isd) locus. During systemic infection of mice, we demonstrated that while S. lugdunensis does not cause overt illness, it does colonize and proliferate to high numbers in the kidneys. By combining mutations in the various iron acquisition loci (isd, fhuC, sst-1, and feo), we demonstrate that only a strain deficient for all of these systems was attenuated in its ability to proliferate to high numbers in the murine kidney. We propose the concerted action of heme and non-heme iron acquisition systems also enable S. lugdunensis to cause human infection.
Collapse
|
10
|
Norepinephrine induces growth of Desulfovibrio vulgaris in an iron dependent manner. Anaerobe 2022; 75:102582. [DOI: 10.1016/j.anaerobe.2022.102582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 11/23/2022]
|
11
|
Barroso FD, da Silva LJ, Sá LG, da Silva CR, Neto JB, do Nascimento FB, Queiroz HA, Leitão AC, Cabral VP, Rodrigues DS, Barbosa AD, Cavalcanti BC, Morais MO, Júnior HV. Synergistic activity of dobutamine combined with azoles and its mechanism of action against strains of Candida glabrata. Future Microbiol 2022; 17:437-448. [PMID: 35285249 DOI: 10.2217/fmb-2020-0256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: To evaluate the antifungal effect of dobutamine against Candida glabrata as well as its synergism with azoles and its action on biofilm. Methods: The M27-A3 protocol and flow cytometry were used for elucidation of the possible mechanism of action. Results: The tested isolates presented MICs ranging from 2 to 32 μg/ml for dobutamine, with fungistatic effect. A total of 82% of the strains showed synergism with fluconazole, with 90% showing synergism with itraconazole. The effect on biofilm formation was nonsignificant. Cytometry tests showed that dobutamine induced mitochondrial depolarization. Conclusion: Dobutamine has an antifungal effect on strains of C. glabrata and synergistic activity with azoles. This effect is probably mediated by increased oxidative damage to the membrane.
Collapse
Affiliation(s)
- Fatima Dd Barroso
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Lisandra J da Silva
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Lívia Gav Sá
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil.,Faculty of Biomedicine, Unichristus University Center, Fortaleza, Brazil
| | - Cecília R da Silva
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - João Ba Neto
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil.,Faculty of Biomedicine, Unichristus University Center, Fortaleza, Brazil
| | - Francisca Ba do Nascimento
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Helaine A Queiroz
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Amanda C Leitão
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Vitória Pf Cabral
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Daniel S Rodrigues
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Amanda D Barbosa
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| | - Bruno C Cavalcanti
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 60430-276, Brazil
| | - Manoel O Morais
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 60430-276, Brazil
| | - Hélio Vn Júnior
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
12
|
Eukaryotic catecholamine hormones influence the chemotactic control of Vibrio campbellii by binding to the coupling protein CheW. Proc Natl Acad Sci U S A 2022; 119:e2118227119. [PMID: 35238645 PMCID: PMC8915975 DOI: 10.1073/pnas.2118227119] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Host-emitted stress hormones significantly influence the growth and behavior of various bacterial species; however, their cellular targets have so far remained elusive. Here, we used customized probes and quantitative proteomics to identify the target of epinephrine and the α-adrenoceptor agonist phenylephrine in live cells of the aquatic pathogen Vibrio campbellii. Consequently, we have discovered the coupling protein CheW, which is in the center of the chemotaxis signaling network, as a target of both molecules. We not only demonstrate direct ligand binding to CheW but also elucidate how this affects chemotactic control. These findings are pivotal for further research on hormone-specific effects on bacterial behavior. In addition to their well-known role as stress-associated catecholamine hormones in animals and humans, epinephrine (EPI) and norepinephrine (NE) act as interkingdom signals between eukaryotic hosts and bacteria. However, the molecular basis of their effects on bacteria is not well understood. In initial phenotypic studies utilizing Vibrio campbellii as a model organism, we characterized the bipartite mode of action of catecholamines, which consists of promotion of growth under iron limitation and enhanced colony expansion on soft agar. In order to identify the molecular targets of the hormones, we designed and synthesized tailored probes for chemical proteomic studies. As the catechol group in EPI and NE acts as an iron chelator and is prone to form a reactive quinone moiety, we devised a photoprobe based on the adrenergic agonist phenylephrine (PE), which solely influenced colony expansion. Using this probe, we identified CheW, located at the core of the chemotaxis signaling network, as a major target. In vitro studies confirmed that EPI, NE, PE, and labetalol, a clinically applied antagonist, bind to purified CheW with affinity constants in the submicromolar range. In line with these findings, exposure of V. campbellii to these adrenergic agonists affects the chemotactic control of the bacterium. This study highlights an effect of eukaryotic signaling molecules on bacterial motility.
Collapse
|
13
|
Hamed A, Pullinger G, Stevens M, Farveen F, Freestone P. Characterisation of the E. coli and Salmonella qseC and qseE mutants reveals a metabolic rather than adrenergic receptor role. FEMS Microbiol Lett 2022; 369:6524176. [PMID: 35137015 PMCID: PMC8897314 DOI: 10.1093/femsle/fnac012] [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: 08/26/2021] [Revised: 12/31/2021] [Accepted: 02/04/2022] [Indexed: 11/14/2022] Open
Abstract
Catecholamine stress hormones (norepinephrine, epinephrine, and dopamine) are signals that have been shown to be used as environmental cues, which affect the growth and virulence of normal microbiota as well as pathogenic bacteria. It has been reported that Escherichia coli and Salmonella use the two-component system proteins QseC and QseE to recognise catecholamines and so act as bacterial adrenergic receptors. In this study, we mutated the E. coli O157:H7 and Salmonella enterica serovar Typhimurium genes encoding QseC and QseE and found that this did not block stress hormone responsiveness in either species. Motility, biofilm formation, and analysis of virulence of the mutants using two infection models were similar to the wild-type strains. The main differences in phenotypes of the qseC and qseE mutants were responses to changes in temperature and growth in different media particularly with respect to salt, carbon, and nitrogen salt sources. In this physiological respect, it was also found that the phenotypes of the qseC and qseE mutants differed between E. coli and Salmonella. These findings collectively suggest that QseC and QseE are not essential for E. coli and Salmonella to respond to stress hormones and that the proteins may be playing a role in regulating metabolism.
Collapse
Affiliation(s)
- Abdalla Hamed
- Department of Microbiology and Immunology, Faculty of Medicine, University of Zawia, Zawiya QP7X+536, Libya
| | - Gillian Pullinger
- Division of Microbiology, Institute for Animal Health, Compton, Newbury RG20 7NN, United Kingdom
| | - Mark Stevens
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, United Kingdom
| | - Fathima Farveen
- Department of Respiratory Sciences, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Primrose Freestone
- Corresponding author: Department of Respiratory Sciences, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom. Tel: +44 (0)116 2525656; Fax: +44 (0)116 2525030; E-mail:
| |
Collapse
|
14
|
Sharma VK, Akavaram S, Bayles DO. Genomewide transcriptional response of Escherichia coli O157:H7 to norepinephrine. BMC Genomics 2022; 23:107. [PMID: 35135480 PMCID: PMC8822769 DOI: 10.1186/s12864-021-08167-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/10/2021] [Indexed: 01/18/2023] Open
Abstract
Background Chemical signaling between a mammalian host and intestinal microbes is health and maintenance of ‘healthy’ intestinal microbiota. Escherichia coli O157:H7 can hijack host- and microbiota-produced chemical signals for survival in a harsh and nutritionally competitive gastrointestinal environment and for intestinal colonization. Norepinephrine (NE) produced by sympathetic neurons of the enteric nervous system has been shown in vitro to induce expression of genes controlling E. coli O157:H7 swimming motility, acid resistance, and adherence to epithelial cells. A previous study used a microarray approach to identify differentially expressed genes in E. coli O157:H7 strain EDL933 in response to NE. To elucidate a comprehensive transcriptional response to NE, we performed RNA-Seq on rRNA-depleted RNA of E. coli O157:H7 strain NADC 6564, an isolate of a foodborne E. coli O157:H7 strain 86–24. The reads generated by RNA-Seq were mapped to NADC 6564 genome using HiSat2. The mapped reads were quantified by htseq-count against the genome of strain NADC 6564. The differentially expressed genes were identified by analyzing quantified reads by DESeq2. Results Of the 585 differentially expressed genes (≥ 2.0-fold; p < 0.05), many encoded pathways promoting ability of E. coli O157:H7 strain NADC 6564 to colonize intestines of carrier animals and to produce disease in an incidental human host through increased adherence to epithelial cells and production of Shiga toxins. In addition, NE exposure also induced the expression of genes encoding pathways conferring prolonged survival at extreme acidity, controlling influx/efflux of specific nutrients/metabolites, and modulating tolerance to various stressors. A correlation was also observed between the EvgS/EvgA signal transduction system and the ability of bacterial cells to survive exposure to high acidity for several hours. Many genes involved in nitrogen, sulfur, and amino acid uptake were upregulated while genes linked to iron (Fe3+) acquisition and transport were downregulated. Conclusion The availability of physiological levels of NE in gastrointestinal tract could serve as an important cue for E. coli O157:H7 to engineer its virulence, stress, and metabolic pathways for colonization in reservoir animals, such as cattle, causing illness in humans, and surviving outside of a host. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08167-z.
Collapse
Affiliation(s)
- Vijay K Sharma
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, ARS-USDA, Ames, IA, 50010, USA.
| | - Suryatej Akavaram
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, ARS-USDA, Ames, IA, 50010, USA.,Current address: 4302 TX-332, Freeport, TX, 77541, USA
| | - Darrell O Bayles
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, ARS-USDA, Ames, IA, 50010, USA
| |
Collapse
|
15
|
Computational and Biological Evaluation of β-Adrenoreceptor Blockers as Promising Bacterial Anti-Virulence Agents. Pharmaceuticals (Basel) 2022; 15:ph15020110. [PMID: 35215223 PMCID: PMC8877484 DOI: 10.3390/ph15020110] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 02/04/2023] Open
Abstract
Bacterial resistance to antibiotics is an increasing public health threat as it has the potential to affect people at any stage of life, as well as veterinary. Various approaches have been proposed to counteract the bacterial resistance development. Tackling bacterial virulence is one of the most promising approaches that confer several merits. The bacterial virulence is mainly regulated by a communication system known as quorum sensing (QS) system. Meanwhile, bacteria can sense the adrenergic hormones and eavesdrops on the host cells to establish their infection, adrenergic hormones were shown to enhance the bacterial virulence. In this study, β-adrenoreceptor blockers were proposed not only to stop bacterial espionage on our cells but also as inhibitors to the bacterial QS systems. In this context, a detailed in silico study has been conducted to evaluate the affinities of twenty-two β-blockers to compete on different structural QS receptors. Among the best docked and thermodynamically stable β-blockers; atenolol, esmolol, and metoprolol were subjected to further in vitro and in vivo investigation to evaluate their anti-QS activities against Chromobacterium violaceum, Pseudomonas aeruginosa and Salmonella typhimurium. The three tested β-blockers decreased the production of QS-controlled C. violaceum, and the formation of biofilm by P. aeruginosa and S. typhimurium. Additionally, the tested β-blockers down-regulated the P. aeruginosa QS-encoding genes and S. typhimurium sensor kinase encoding genes. Furthermore, metoprolol protected mice against P. aeruginosa and S. typhimurium. Conclusively, these investigated β-blockers are promising anti-virulence agents antagonizing adrenergic hormones induced virulence, preventing bacterial espionage, and blocking bacterial QS systems.
Collapse
|
16
|
Alghofaili F, Najmuldeen H, Kareem BO, Shlla B, Fernandes VE, Danielsen M, Ketley JM, Freestone P, Yesilkaya H. Host Stress Signals Stimulate Pneumococcal Transition from Colonization to Dissemination into the Lungs. mBio 2021; 12:e0256921. [PMID: 34696596 PMCID: PMC8546540 DOI: 10.1128/mbio.02569-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/24/2021] [Indexed: 11/20/2022] Open
Abstract
Streptococcus pneumoniae is an asymptomatic colonizer of the nasopharynx, but it is also one of the most important bacterial pathogens of humans, causing a wide range of mild to life-threatening diseases. The basis of the pneumococcal transition from a commensal to a parasitic lifestyle is not fully understood. We hypothesize that exposure to host catecholamine stress hormones is important for this transition. In this study, we demonstrated that pneumococci preexposed to a hormone released during stress, norepinephrine (NE), have an increased capacity to translocate from the nasopharynx into the lungs compared to untreated pneumococci. Examination of NE-treated pneumococci revealed major alterations in metabolic profiles, cell associations, capsule synthesis, and cell size. By systemically mutating all 12 two-component and 1 orphan regulatory systems, we also identified a unique genetic regulatory circuit involved in pneumococcal recognition and responsiveness to human stress hormones. IMPORTANCE Microbes acquire unique lifestyles under different environmental conditions. Although this is a widespread occurrence, our knowledge of the importance of various host signals and their impact on microbial behavior is not clear despite the therapeutic value of this knowledge. We discovered that catecholamine stress hormones are the host signals that trigger the passage of Streptococcus pneumoniae from a commensal to a parasitic state. We identify that stress hormone treatment of this microbe leads to reductions in cell size and capsule synthesis and renders it more able to migrate from the nasopharynx into the lungs in a mouse model of infection. The microbe requires the TCS09 protein for the recognition and processing of stress hormone signals. Our work has particular clinical significance as catecholamines are abundant in upper respiratory fluids as well as being administered therapeutically to reduce inflammation in ventilated patients, which may explain why intubation in the critically ill is a recognized risk factor for the development of pneumococcal pneumonia.
Collapse
Affiliation(s)
- Fayez Alghofaili
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
- Department of Biology, College of Science, Majmaah University, Majmaah, Saudi Arabia
| | - Hastyar Najmuldeen
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
- Department of Biology, College of Science, University of Sulaimani, Sulaymaniyah, Iraq
| | - Banaz O. Kareem
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| | - Bushra Shlla
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
- Department of Biology, College of Science, University of Mosul, Mosul, Iraq
| | - Vitor E. Fernandes
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| | | | - Julian M. Ketley
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Primrose Freestone
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| | - Hasan Yesilkaya
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
17
|
Boukerb AM, Cambronel M, Rodrigues S, Mesguida O, Knowlton R, Feuilloley MGJ, Zommiti M, Connil N. Inter-Kingdom Signaling of Stress Hormones: Sensing, Transport and Modulation of Bacterial Physiology. Front Microbiol 2021; 12:690942. [PMID: 34690943 PMCID: PMC8526972 DOI: 10.3389/fmicb.2021.690942] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 08/06/2021] [Indexed: 12/29/2022] Open
Abstract
Prokaryotes and eukaryotes have coexisted for millions of years. The hormonal communication between microorganisms and their hosts, dubbed inter-kingdom signaling, is a recent field of research. Eukaryotic signals such as hormones, neurotransmitters or immune system molecules have been shown to modulate bacterial physiology. Among them, catecholamines hormones epinephrine/norepinephrine, released during stress and physical effort, or used therapeutically as inotropes have been described to affect bacterial behaviors (i.e., motility, biofilm formation, virulence) of various Gram-negative bacteria (e.g., Escherichia coli, Salmonella enterica serovar Typhimurium, Pseudomonas aeruginosa, Vibrio sp.). More recently, these molecules were also shown to influence the physiology of some Gram-positive bacteria like Enterococcus faecalis. In E. coli and S. enterica, the stress-associated mammalian hormones epinephrine and norepinephrine trigger a signaling cascade by interacting with the QseC histidine sensor kinase protein. No catecholamine sensors have been well described yet in other bacteria. This review aims to provide an up to date report on catecholamine sensors in eukaryotes and prokaryotes, their transport, and known effects on bacteria.
Collapse
Affiliation(s)
- Amine Mohamed Boukerb
- Laboratoire de Microbiologie Signaux et Microenvironnement EA 4312, Université de Rouen, Normandie Université, Évreux, France
| | - Melyssa Cambronel
- Laboratoire de Microbiologie Signaux et Microenvironnement EA 4312, Université de Rouen, Normandie Université, Évreux, France
| | - Sophie Rodrigues
- EA 3884, LBCM, IUEM, Université de Bretagne-Sud, Lorient, France
| | - Ouiza Mesguida
- Laboratoire de Microbiologie Signaux et Microenvironnement EA 4312, Université de Rouen, Normandie Université, Évreux, France
| | - Rikki Knowlton
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Marc G J Feuilloley
- Laboratoire de Microbiologie Signaux et Microenvironnement EA 4312, Université de Rouen, Normandie Université, Évreux, France
| | - Mohamed Zommiti
- Laboratoire de Microbiologie Signaux et Microenvironnement EA 4312, Université de Rouen, Normandie Université, Évreux, France
| | - Nathalie Connil
- Laboratoire de Microbiologie Signaux et Microenvironnement EA 4312, Université de Rouen, Normandie Université, Évreux, France
| |
Collapse
|
18
|
Ozuna H, Uriarte SM, Demuth DR. The Hunger Games: Aggregatibacter actinomycetemcomitans Exploits Human Neutrophils As an Epinephrine Source for Survival. Front Immunol 2021; 12:707096. [PMID: 34456916 PMCID: PMC8387626 DOI: 10.3389/fimmu.2021.707096] [Citation(s) in RCA: 10] [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: 05/08/2021] [Accepted: 07/21/2021] [Indexed: 11/13/2022] Open
Abstract
Aggregatibacter actinomycetemcomitans is a gram-negative facultative anaerobe and an opportunistic oral pathogen, strongly associated with periodontitis and other inflammatory diseases. Periodontitis is a chronic inflammation of the periodontium resulting from the inflammatory response of the host towards the dysbiotic microbial community present at the gingival crevice. Previously, our group identified catecholamines and iron as the signals that activate the QseBC two-component system in A. actinomycetemcomitans, necessary for the organism to acquire iron as a nutrient to survive in the anaerobic environment. However, the source of catecholamines has not been identified. It has been reported that mouse neutrophils can release catecholamines. In periodontitis, large infiltration of neutrophils is found at the subgingival pocket; hence, we wanted to test the hypothesis that A. actinomycetemcomitans exploits human neutrophils as a source for catecholamines. In the present study, we showed that human neutrophils synthesize, store, and release epinephrine, one of the three main types of catecholamines. Human neutrophil challenge with A. actinomycetemcomitans induced exocytosis of neutrophil granule subtypes: secretory vesicles, specific granules, gelatinase granules, and azurophilic granules. In addition, by selectively inhibiting granule exocytosis, we present the first evidence that epinephrine is stored in azurophilic granules. Using QseC mutants, we showed that the periplasmic domain of the QseC sensor kinase is required for the interaction between A. actinomycetemcomitans and epinephrine. Finally, epinephrine-containing supernatants collected from human neutrophils promoted A. actinomycetemcomitans growth and induced the expression of the qseBC operon under anaerobic conditions. Based on our findings, we propose that A. actinomycetemcomitans promotes azurophilic granule exocytosis by neutrophils as an epinephrine source to promote bacterial survival.
Collapse
Affiliation(s)
- Hazel Ozuna
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, United States
| | - Silvia M. Uriarte
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, United States
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, United States
| | - Donald R. Demuth
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, United States
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, United States
| |
Collapse
|
19
|
Kim HS, Hashimoto T, Fischer K, Bernigaud C, Chosidow O, Yosipovitch G. Scabies itch: an update on neuroimmune interactions and novel targets. J Eur Acad Dermatol Venereol 2021; 35:1765-1776. [PMID: 33960033 DOI: 10.1111/jdv.17334] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022]
Abstract
Frequently described as 'the worst itch' one can ever experience scabies itch is the hallmark of Sarcoptes scabiei mite infestation. Notably, the itchiness often persists for weeks despite scabicides therapy. The mechanism of scabies itch is not yet fully understood, and effective treatment modalities are still missing which can severely affect the quality of life. The aim of this review is to provide an overview of the scope of itch in scabies and highlight candidate mechanisms underlying this itch. We herein discuss scabies itch, with a focus on the nature, candidate underlying mechanisms and treatment options. We also synthesize this information with current understanding of the mechanisms contributing to non-histaminergic itch in other conditions. Itch is a major problem in scabies and can lead to grave consequences. We provide the latest insights on host-mite interaction, secondary microbial infection and neural sensitization with special emphasis on keratinocytes and mast cells to better understand the mechanism of itch in scabies. Also, the most relevant current modalities remaining under investigation that possess promising perspectives for scabies itch (i.e. protease-activated receptor-2 (PAR-2) inhibitor, Mas-related G protein-coupled receptor X2 (MRGPRX2) antagonist) are discussed. Greater understanding of these diverse mechanisms may provide a rational basis for the development of improved and targeted approaches to control itch in individuals with scabies.
Collapse
Affiliation(s)
- H S Kim
- Dr Philip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, Miller School of Medicine, University of Miami, Miami, FL, USA.,Department of Dermatology, Incheon St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - T Hashimoto
- Department of Dermatology, National Defense Medical College, Saitama, Japan
| | - K Fischer
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - C Bernigaud
- Faculté de Santé de Créteil et Service de Dermatologie, APHP, Hôpital Henri-Mondor, Université Paris-Est, Créteil, France.,Research Group Dynamic, EA7380, Faculté de Santé de Créteil, Ecole Nationale Vétérinaire d'Alfort, USC ANSES, Université Paris-Est Créteil, Créteil, France
| | - O Chosidow
- Faculté de Santé de Créteil et Service de Dermatologie, APHP, Hôpital Henri-Mondor, Université Paris-Est, Créteil, France.,Research Group Dynamic, EA7380, Faculté de Santé de Créteil, Ecole Nationale Vétérinaire d'Alfort, USC ANSES, Université Paris-Est Créteil, Créteil, France
| | - G Yosipovitch
- Dr Philip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
20
|
Klebba PE, Newton SMC, Six DA, Kumar A, Yang T, Nairn BL, Munger C, Chakravorty S. Iron Acquisition Systems of Gram-negative Bacterial Pathogens Define TonB-Dependent Pathways to Novel Antibiotics. Chem Rev 2021; 121:5193-5239. [PMID: 33724814 PMCID: PMC8687107 DOI: 10.1021/acs.chemrev.0c01005] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Iron is an indispensable metabolic cofactor in both pro- and eukaryotes, which engenders a natural competition for the metal between bacterial pathogens and their human or animal hosts. Bacteria secrete siderophores that extract Fe3+ from tissues, fluids, cells, and proteins; the ligand gated porins of the Gram-negative bacterial outer membrane actively acquire the resulting ferric siderophores, as well as other iron-containing molecules like heme. Conversely, eukaryotic hosts combat bacterial iron scavenging by sequestering Fe3+ in binding proteins and ferritin. The variety of iron uptake systems in Gram-negative bacterial pathogens illustrates a range of chemical and biochemical mechanisms that facilitate microbial pathogenesis. This document attempts to summarize and understand these processes, to guide discovery of immunological or chemical interventions that may thwart infectious disease.
Collapse
Affiliation(s)
- Phillip E Klebba
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506, United States
| | - Salete M C Newton
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506, United States
| | - David A Six
- Venatorx Pharmaceuticals, Inc., 30 Spring Mill Drive, Malvern, Pennsylvania 19355, United States
| | - Ashish Kumar
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506, United States
| | - Taihao Yang
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506, United States
| | - Brittany L Nairn
- Department of Biological Sciences, Bethel University, 3900 Bethel Drive, St. Paul, Minnesota 55112, United States
| | - Colton Munger
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506, United States
| | - Somnath Chakravorty
- Jacobs School of Medicine and Biomedical Sciences, SUNY Buffalo, Buffalo, New York 14203, United States
| |
Collapse
|
21
|
The Ambivalent Role of Skin Microbiota and Adrenaline in Wound Healing and the Interplay between Them. Int J Mol Sci 2021; 22:ijms22094996. [PMID: 34066786 PMCID: PMC8125934 DOI: 10.3390/ijms22094996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023] Open
Abstract
After skin injury, wound healing sets into motion a dynamic process to repair and replace devitalized tissues. The healing process can be divided into four overlapping phases: hemostasis, inflammation, proliferation, and maturation. Skin microbiota has been reported to participate in orchestrating the wound healing both in negative and positive ways. Many studies reported that skin microbiota can impose negative and positive effects on the wound. Recent findings have shown that many bacterial species on human skin are able to convert aromatic amino acids into so-called trace amines (TAs) and convert corresponding precursors into dopamine and serotonin, which are all released into the environment. As a stress reaction, wounded epithelial cells release the hormone adrenaline (epinephrine), which activates the β2-adrenergic receptor (β2-AR), impairing the migration ability of keratinocytes and thus re-epithelization. This is where TAs come into play, as they act as antagonists of β2-AR and thus attenuate the effects of adrenaline. The result is that not only TAs but also TA-producing skin bacteria accelerate wound healing. Adrenergic receptors (ARs) play a key role in many physiological and disease-related processes and are expressed in numerous cell types. In this review, we describe the role of ARs in relation to wound healing in keratinocytes, immune cells, fibroblasts, and blood vessels and the possible role of the skin microbiota in wound healing.
Collapse
|
22
|
Perraud Q, Kuhn L, Fritsch S, Graulier G, Gasser V, Normant V, Hammann P, Schalk IJ. Opportunistic use of catecholamine neurotransmitters as siderophores to access iron by Pseudomonas aeruginosa. Environ Microbiol 2020; 24:878-893. [PMID: 33350053 DOI: 10.1111/1462-2920.15372] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 10/22/2022]
Abstract
Iron is an essential nutrient for bacterial growth and the cause of a fierce battle between the pathogen and host during infection. Bacteria have developed several strategies to access iron from the host, the most common being the production of siderophores, small iron-chelating molecules secreted into the bacterial environment. The opportunist pathogen Pseudomonas aeruginosa produces two siderophores, pyoverdine and pyochelin, and is also able to use a wide panoply of xenosiderophores, siderophores produced by other microorganisms. Here, we demonstrate that catecholamine neurotransmitters (dopamine, l-DOPA, epinephrine and norepinephrine) are able to chelate iron and efficiently bring iron into P. aeruginosa cells via TonB-dependent transporters (TBDTs). Bacterial growth assays under strong iron-restricted conditions and with numerous mutants showed that the TBDTs involved are PiuA and PirA. PiuA exhibited more pronounced specificity for dopamine uptake than for norepinephrine, epinephrine and l-DOPA, whereas PirA specificity appeared to be higher for l-DOPA and norepinephrine. Proteomic and qRT-PCR approaches showed pirA transcription and expression to be induced in the presence of all four catecholamines. Finally, the oxidative properties of catecholamines enable them to reduce iron, and we observed ferrous iron uptake via the FeoABC system in the presence of l-DOPA.
Collapse
Affiliation(s)
- Quentin Perraud
- Université de Strasbourg, InnoVec, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France.,CNRS, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France
| | - Lauriane Kuhn
- Plateforme Proteomique Strasbourg - Esplanade, Institut de Biologie Moléculaire et Cellulaire, CNRS, FR1589, 15 rue Descartes, Strasbourg Cedex, F-67084, France
| | - Sarah Fritsch
- Université de Strasbourg, InnoVec, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France.,CNRS, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France
| | - Gwenaëlle Graulier
- Université de Strasbourg, InnoVec, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France.,CNRS, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France
| | - Véronique Gasser
- Université de Strasbourg, InnoVec, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France.,CNRS, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France
| | - Vincent Normant
- Université de Strasbourg, InnoVec, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France.,CNRS, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France
| | - Philippe Hammann
- Plateforme Proteomique Strasbourg - Esplanade, Institut de Biologie Moléculaire et Cellulaire, CNRS, FR1589, 15 rue Descartes, Strasbourg Cedex, F-67084, France
| | - Isabelle J Schalk
- Université de Strasbourg, InnoVec, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France.,CNRS, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France
| |
Collapse
|
23
|
Liu M, Lyte M. Pyruvate is required for catecholamine-stimulated growth of different strains of Campylobacter jejuni. PeerJ 2020; 8:e10011. [PMID: 33062434 PMCID: PMC7528810 DOI: 10.7717/peerj.10011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 08/31/2020] [Indexed: 01/10/2023] Open
Abstract
Humans and food-producing animals are constantly exposed to and affected by stress. As a consequence of stress, the release of stress-related catecholamines, such as norepinephrine (NE) and dopamine (DA), from nerve terminals in the gastrointestinal tract potentiates both the growth and the virulence of pathogenic bacteria. This may lead to the enhancement of gastrointestinal infections in humans or food-producing animals. Compared with foodborne bacterial pathogens such as Escherichia coli and Salmonella spp., less is known about the effect of stress catecholamines on Campylobacter jejuni subsp. jejuni. The present study focuses on the effect(s) of stress catecholamines DA and NE in iron-restricted media and how they affect the growth of different C. jejuni strains NCTC 11168, 81-176, and ML2126. Results demonstrated that DA- and NE-enhanced growth of C. jejuni in iron-restricted media may involve different mechanisms that cannot be explained by current understanding which relies on catecholamine-mediated iron delivery. Specifically, we found that DA-enhanced growth requires pyruvate, whereas NE-enhanced growth does not. We further report significant strain-specific dependence of C. jejuni growth on various catecholamines in the presence or absence of pyruvate. These data provide novel insights into the effect(s) of stress catecholamines on the in vitro growth of C. jejuni in iron-restricted environments, such as the intestinal tract. They suggest a mechanism by which stress-related catecholamines affect the growth of C. jejuni in the intestinal tract of food-producing animals, which in turn may influence colonization and transmission to humans.
Collapse
Affiliation(s)
- Meicen Liu
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States of America
| | - Mark Lyte
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, United States of America
| |
Collapse
|
24
|
Bistoletti M, Bosi A, Banfi D, Giaroni C, Baj A. The microbiota-gut-brain axis: Focus on the fundamental communication pathways. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 176:43-110. [PMID: 33814115 DOI: 10.1016/bs.pmbts.2020.08.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Davide Banfi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy.
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
25
|
Li J, Ma X, Zhao L, Li Y, Zhou Q, Du X. Extended Contact Lens Wear Promotes Corneal Norepinephrine Secretion and Pseudomonas aeruginosa Infection in Mice. Invest Ophthalmol Vis Sci 2020; 61:17. [PMID: 32298434 PMCID: PMC7401850 DOI: 10.1167/iovs.61.4.17] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose Extended contact lens (CL) wear predisposes the wearer to Pseudomonas aeruginosa infection of the cornea, but the mechanism involved remains incompletely understood. The purpose of this study was to investigate the role of the stress hormone norepinephrine (NE) in the pathogenesis of CL-induced P. aeruginosa keratitis. Methods A total 195 adult C57BL/6 mice were used in this study. Corneal NE content was measured after 48 hours of sterile CL wear in mice. The effect of NE on P. aeruginosa adhesion and biofilm formation on the CL surface was examined in vitro. Moreover, mouse eyes were covered with P. aeruginosa-contaminated CLs, and either 500-µM NE was topically applied or the eyes were subconjunctivally injected with 100 µg of N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) to deplete local NE. Clinical scores, neutrophil infiltration, proinflammatory cytokine levels, and bacterial load on the corneas and CLs were evaluated. Results Corneal NE content was elevated with extended CL wear in mice. In vitro, NE promoted the adhesion and biofilm formation of P. aeruginosa on the CL surface. In mice, topical application of NE aggravated P. aeruginosa infection, accompanied with increased clinical scores, neutrophil infiltration, proinflammatory cytokine expression, and bacterial burden on the corneas and CLs. However, pre-depletion of local NE with DSP-4 significantly alleviated the severity of P. aeruginosa keratitis. Conclusions Extended CL wear elevates corneal NE content, which promotes the pathogenesis of CL-induced P. aeruginosa keratitis in mice. Targeting NE may provide a potential strategy for the treatment of CL-related corneal infection caused by P. aeruginosa.
Collapse
|
26
|
Zhang Y, Edmonds KA, Raines DJ, Murphy BA, Wu H, Guo C, Nolan EM, VanNieuwenhze MS, Duhme-Klair AK, Giedroc DP. The Pneumococcal Iron Uptake Protein A (PiuA) Specifically Recognizes Tetradentate Fe IIIbis- and Mono-Catechol Complexes. J Mol Biol 2020; 432:5390-5410. [PMID: 32795535 DOI: 10.1016/j.jmb.2020.08.005] [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: 06/24/2020] [Revised: 08/02/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022]
Abstract
Streptococcus pneumoniae (Spn) is an important Gram-positive human pathogen that causes millions of infections worldwide with an increasing occurrence of antibiotic resistance. Fe acquisition is a crucial virulence determinant in Spn; further, Spn relies on exogenous FeIII-siderophore scavenging to meet nutritional Fe needs. Recent studies suggest that the human catecholamine stress hormone, norepinephrine (NE), facilitates Fe acquisition in Spn under conditions of transferrin-mediated Fe starvation. Here we show that the solute binding lipoprotein PiuA from the piu Fe acquisition ABC transporter PiuBCDA, previously described as an Fe-hemin binding protein, binds tetradentate catechol FeIII complexes, including NE and the hydrolysis products of enterobactin. Two protein-derived ligands (H238, Y300) create a coordinately saturated FeIII complex, which parallel recent studies in the Gram-negative intestinal pathogen Campylobacter jejuni. Our in vitro studies using NMR spectroscopy and 54Fe LC-ICP-MS confirm the FeIII can move from transferrin to apo-PiuA in an NE-dependent manner. Structural analysis of PiuA FeIII-bis-catechol and GaIII-bis-catechol and GaIII-(NE)2 complexes by NMR spectroscopy reveals only localized structural perturbations in PiuA upon ligand binding, largely consistent with recent descriptions of other solute binding proteins of type II ABC transporters. We speculate that tetradentate FeIII complexes formed by mono- and bis-catechol species are important Fe sources in Gram-positive human pathogens, since PiuA functions in the same way as SstD from Staphylococcus aureus.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA; Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405, USA
| | - Katherine A Edmonds
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA
| | - Daniel J Raines
- Department of Chemistry, University of York, Heslington, York YO10 5DD, United Kingdom
| | - Brennan A Murphy
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA
| | - Hongwei Wu
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA
| | - Chuchu Guo
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Elizabeth M Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Anne-K Duhme-Klair
- Department of Chemistry, University of York, Heslington, York YO10 5DD, United Kingdom
| | - David P Giedroc
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA; Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405, USA.
| |
Collapse
|
27
|
Zhang Y, Sen S, Giedroc DP. Iron Acquisition by Bacterial Pathogens: Beyond Tris-Catecholate Complexes. Chembiochem 2020; 21:1955-1967. [PMID: 32180318 PMCID: PMC7367709 DOI: 10.1002/cbic.201900778] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/06/2020] [Indexed: 12/11/2022]
Abstract
Sequestration of the essential nutrient iron from bacterial invaders that colonize the vertebrate host is a central feature of nutritional immunity and the "fight over transition metals" at the host-pathogen interface. The iron quota for many bacterial pathogens is large, as iron enzymes often make up a significant share of the metalloproteome. Iron enzymes play critical roles in respiration, energy metabolism, and other cellular processes by catalyzing a wide range of oxidation-reduction, electron transfer, and oxygen activation reactions. In this Concept article, we discuss recent insights into the diverse ways that bacterial pathogens acquire this essential nutrient, beyond the well-characterized tris-catecholate FeIII complexes, in competition and cooperation with significant host efforts to cripple these processes. We also discuss pathogen strategies to adapt their metabolism to less-than-optimal iron concentrations, and briefly speculate on what might be an integrated adaptive response to the concurrent limitation of both iron and zinc in the infected host.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405-7102, USA
| | - Sambuddha Sen
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA
| | - David P Giedroc
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405-7102, USA
| |
Collapse
|
28
|
Le TH, Lee HJ, Kim JH, Park SJ. Detection of Ferric Ions and Catecholamine Neurotransmitters via Highly Fluorescent Heteroatom Co-Doped Carbon Dots. SENSORS (BASEL, SWITZERLAND) 2020; 20:E3470. [PMID: 32575578 PMCID: PMC7349486 DOI: 10.3390/s20123470] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/03/2020] [Accepted: 06/16/2020] [Indexed: 12/23/2022]
Abstract
Carbon dots (CDs) demonstrate very poor fluorescence quantum yield (QY). In this study, with the help of a hydrothermal method, we combined CDs with nitrogen and phosphorus elements belonging to the VA group (in the periodic table) to form heteroatom co-doped CDs, i.e., nitrogen and phosphorus co-doped carbon dots (NPCDs). These displayed a significant improvement in the QY (up to 84%), which was as much as four times than that of CDs synthesized by the same method. The as-prepared NPCDs could be used as an "off-on" fluorescence detector for the rapid and effective sensing of ferric ions (Fe3+) and catecholamine neurotransmitters (CNs) such as dopamine (DA), adrenaline (AD), and noradrenaline (NAD). The fluorescence of NPCDs was "turned off" and the emission wavelength was slightly red-shifted upon increasing the Fe3+ concentration. However, when CNs were incorporated, the fluorescence of NPCDs was recovered in a short response time; this indicated that CN concentration could be monitored, relying on enhancing the fluorescence signal of NPCDs. As a result, NPCDs are considered as a potential fluorescent bi-sensor for Fe3+ and CN detection. Particularly, in this research, we selected DA as the representative neurotransmitter of the CN group along with Fe3+ to study the sensing system based on NPCDs. The results exhibited good linear ranges with a limit of detection (LOD) of 0.2 and 0.1 µM for Fe3+ and DA, respectively.
Collapse
Affiliation(s)
| | | | | | - Sang Joon Park
- Department of Chemical and Biological Engineering, Gachon University, Seongnam 13120, Korea; (T.H.L.); (H.J.L.); (J.H.K.)
| |
Collapse
|
29
|
Sung Kim H, Yosipovitch G. THE SKIN MICROBIOTA AND ITCH: Is There a Link? THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2020; 13:S39-S46. [PMID: 33282109 PMCID: PMC7710288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Itch is an unpleasant sensation that emanates primarily from the skin. The chemical mediators that drive neuronal activity originate from a complex interaction between keratinocytes, inflammatory cells, nerve endings, and the skin microbiota, relaying itch signals to the brain. Stress also exacerbates itch via the skin-brain axis. Recently, the microbiota has surfaced as a major player to regulate this axis, notably during stress settings aroused by actual or perceived homeostatic challenge. The routes of communication between the microbiota and brain are slowly being unraveled and involve neurochemicals (i.e., acetylcholine, histamine, catecholamines, and corticotropin) that originate from the microbiota itself. By focusing on itch biology and by referring to the more established field of pain research, this review examines the possible means by which the skin microbiota contributes to itch.
Collapse
Affiliation(s)
- Hei Sung Kim
- Dr. Kim is with the Department of Dermatology and Cutaneous Surgery at Miami Itch Center, Miller School of Medicine at University of Miami in Miami, Florida, the Department of Dermatology at Incheon St. Mary's Hospital, The Catholic University of Korea in Seoul, Korea, and the Department of Biomedicine and Health Sciences, at The Catholic University of Korea in Seoul, Korea
- Dr. Yosipovitch is with the Department of Dermatology and Cutaneous Surgery at Miami Itch Center, Miller School of Medicine at the University of Miami in Miami, Florida
| | - Gil Yosipovitch
- Dr. Kim is with the Department of Dermatology and Cutaneous Surgery at Miami Itch Center, Miller School of Medicine at University of Miami in Miami, Florida, the Department of Dermatology at Incheon St. Mary's Hospital, The Catholic University of Korea in Seoul, Korea, and the Department of Biomedicine and Health Sciences, at The Catholic University of Korea in Seoul, Korea
- Dr. Yosipovitch is with the Department of Dermatology and Cutaneous Surgery at Miami Itch Center, Miller School of Medicine at the University of Miami in Miami, Florida
| |
Collapse
|
30
|
Ma X, Wang Q, Song F, Li Y, Li J, Dou S, Xie L, Zhou Q. Corneal epithelial injury-induced norepinephrine promotes Pseudomonas aeruginosa keratitis. Exp Eye Res 2020; 195:108048. [PMID: 32376471 DOI: 10.1016/j.exer.2020.108048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/17/2020] [Accepted: 04/27/2020] [Indexed: 12/17/2022]
Abstract
Tissue injury causes the secretion of stress hormone catecholamine and increases susceptibility to opportunistic infection. Pseudomonas aeruginosa (P. aeruginosa) is an opportunistic pathogen that is a leading cause of microbial keratitis usually associated with ocular injury or contact lens wear. However, the effect of catecholamine on P. aeruginosa induced corneal infection is unknown. Here, we test if norepinephrine (NE) would promote the progression of P. aeruginosa keratitis in mice. Adult C57BL/6 mouse corneas were scarified and then inoculated with P. aeruginosa. The content of NE was elevated in corneas after scarification and inoculation with P. aeruginosa. Then, exogenous NE was applied to the infected corneas at 24 h after inoculation; control eyes were treated with sterile saline. Topical application of NE aggravated the severity of P. aeruginosa keratitis, accompanied with the increase of clinical score, bacterial load, pathological changes, neutrophils infiltration, bacterial virulence factors and proinflammatory factors levels. In order to further verify the role of NE, N-(2-Chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride (DSP-4), a neurotoxin selected to deplete NE, was injected subconjunctivally 12 h before scarification. Pre-depletion of local NE by DSP-4 significantly alleviated the severity of corneal infection. Moreover, NE was also confirmed to increase the bacterial growth and the expression of virulence factors gene in vitro. Together, these data showed that increased corneal NE content facilitated the progression of P. aeruginosa keratitis in mice by amplifying host excessive inflammatory response and bacterial virulence. Therefore, targeting NE may provide a potential strategy for the treatment of P. aeruginosa keratitis.
Collapse
Affiliation(s)
- Xiubin Ma
- Medical College, Qingdao University, Qingdao, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Qun Wang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Fangying Song
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Ya Li
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Jing Li
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Shengqian Dou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Lixin Xie
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.
| |
Collapse
|
31
|
The Skin Microbiota and Itch: Is There a Link? J Clin Med 2020; 9:jcm9041190. [PMID: 32331207 PMCID: PMC7230651 DOI: 10.3390/jcm9041190] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
Itch is an unpleasant sensation that emanates primarily from the skin. The chemical mediators that drive neuronal activity originate from a complex interaction between keratinocytes, inflammatory cells, nerve endings and the skin microbiota, relaying itch signals to the brain. Stress also exacerbates itch via the skin–brain axis. Recently, the microbiota has surfaced as a major player to regulate this axis, notably during stress settings aroused by actual or perceived homeostatic challenge. The routes of communication between the microbiota and brain are slowly being unraveled and involve neurochemicals (i.e., acetylcholine, histamine, catecholamines, corticotropin) that originate from the microbiota itself. By focusing on itch biology and by referring to the more established field of pain research, this review examines the possible means by which the skin microbiota contributes to itch.
Collapse
|
32
|
Marchetti M, De Bei O, Bettati S, Campanini B, Kovachka S, Gianquinto E, Spyrakis F, Ronda L. Iron Metabolism at the Interface between Host and Pathogen: From Nutritional Immunity to Antibacterial Development. Int J Mol Sci 2020; 21:E2145. [PMID: 32245010 PMCID: PMC7139808 DOI: 10.3390/ijms21062145] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 02/08/2023] Open
Abstract
Nutritional immunity is a form of innate immunity widespread in both vertebrates and invertebrates. The term refers to a rich repertoire of mechanisms set up by the host to inhibit bacterial proliferation by sequestering trace minerals (mainly iron, but also zinc and manganese). This strategy, selected by evolution, represents an effective front-line defense against pathogens and has thus inspired the exploitation of iron restriction in the development of innovative antimicrobials or enhancers of antimicrobial therapy. This review focuses on the mechanisms of nutritional immunity, the strategies adopted by opportunistic human pathogen Staphylococcus aureus to circumvent it, and the impact of deletion mutants on the fitness, infectivity, and persistence inside the host. This information finally converges in an overview of the current development of inhibitors targeting the different stages of iron uptake, an as-yet unexploited target in the field of antistaphylococcal drug discovery.
Collapse
Affiliation(s)
- Marialaura Marchetti
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
| | - Omar De Bei
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (O.D.B.); (B.C.)
| | - Stefano Bettati
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Institute of Biophysics, National Research Council, 56124 Pisa, Italy
- National Institute of Biostructures and Biosystems, 00136 Rome, Italy
| | - Barbara Campanini
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (O.D.B.); (B.C.)
| | - Sandra Kovachka
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Luca Ronda
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Institute of Biophysics, National Research Council, 56124 Pisa, Italy
| |
Collapse
|
33
|
Abstract
In an oxygenic environment, poorly soluble Fe3+ must be reduced to meet the cellular Fe2+ demand. This study demonstrates that elevated CO2/HCO3− levels accelerate chemical Fe3+ reduction through phenolic compounds, thus increasing intracellular Fe2+ availability. A number of biological environments are characterized by the presence of phenolic compounds and elevated HCO3− levels and include soil habitats and the human body. Fe2+ availability is of particular interest in the latter, as it controls the infectiousness of pathogens. Since the effect postulated here is abiotic, it generally affects the Fe2+ distribution in nature. Iron is a vital mineral for almost all living organisms and has a pivotal role in central metabolism. Despite its great abundance on earth, the accessibility for microorganisms is often limited, because poorly soluble ferric iron (Fe3+) is the predominant oxidation state in an aerobic environment. Hence, the reduction of Fe3+ is of essential importance to meet the cellular demand of ferrous iron (Fe2+) but might become detrimental as excessive amounts of intracellular Fe2+ tend to undergo the cytotoxic Fenton reaction in the presence of hydrogen peroxide. We demonstrate that the complex formation rate of Fe3+ and phenolic compounds like protocatechuic acid was increased by 46% in the presence of HCO3− and thus accelerated the subsequent redox reaction, yielding reduced Fe2+. Consequently, elevated CO2/HCO3− levels increased the intracellular Fe2+ availability, which resulted in at least 50% higher biomass-specific fluorescence of a DtxR-based Corynebacterium glutamicum reporter strain, and stimulated growth. Since the increased Fe2+ availability was attributed to the interaction of HCO3− and chemical iron reduction, the abiotic effect postulated in this study is of general relevance in geochemical and biological environments.
Collapse
|
34
|
Inbaraj S, Sejian V, Ramasamy S. Role of environmental stressor-host immune system–pathogen interactions in development of infectious disease in farm animals. BIOL RHYTHM RES 2019. [DOI: 10.1080/09291016.2019.1695084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Sophia Inbaraj
- Division of Bacteriology and Mycology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Veerasamy Sejian
- Animal Physiology Division, ICAR-National Institute Animal Nutrition and Physiology, Bengaluru, India
| | - Santhamani Ramasamy
- Department of microbiology and immunology, Post-doctoral research fellow, Albert Einstein College of Medicine, New York, NY, USA
| |
Collapse
|
35
|
Machorro-Rojas N, Sainz-Espuñes T, Godínez-Victoria M, Castañeda-Sánchez JI, Campos-Rodríguez R, Pacheco-Yepez J, Drago-Serrano ME. Impact of chronic immobilization stress on parameters of colonic homeostasis in BALB/c mice. Mol Med Rep 2019; 20:2083-2090. [PMID: 31257542 PMCID: PMC6691234 DOI: 10.3892/mmr.2019.10437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 03/07/2019] [Indexed: 12/24/2022] Open
Abstract
The intestinal epithelium is a monolayer of cells arranged side‑by‑side and connected by tight junction (TJ) proteins expressed at the apical extreme of the paracellular membrane. This layer prevents stress‑induced inflammatory responses, thus helping to maintain gut barrier function and gut homeostasis. The aim of the present study was to evaluate the effects of chronic immobilization stress on the colonic expression of various parameters of homeostasis. A total of two groups of female BALB/c mice (n=6) were included: A stressed group (short‑term immobilization for 2 h/day for 4 consecutive days) and an unstressed (control) group. Colon samples were obtained to detect neutrophils and goblet cells by optical microscopy, TJ protein expression (occludin, and claudin ‑2, ‑4, ‑7, ‑12 and ‑15) by western blotting, mRNA levels of TJ genes and proinflammatory cytokines [tumor necrosis factor (TNF)‑α, interleukin (IL)‑1β, ‑6 and ‑8] by reverse transcription‑quantitative PCR, fecal lactoferrin by ELISA and the number of colony‑forming units of aerobic bacteria. Compared with goblet cells in control mice, goblet cells were enlarged and reduced in number in stressed mice, whereas neutrophil cellularity was unaltered. Stressed mice exhibited reduced mRNA expression for all evaluated TJ mRNAs, with the exception of claudin‑7, which was upregulated. Protein levels of occludin and all claudins (with the exception of claudin‑12) were decreased in stressed mice. Fecal lactoferrin, proinflammatory cytokine mRNA levels and aerobic bacterial counts were all increased in the stressed group. These results indicated that immobilization stress induced proinflammatory and potential remodeling effects in the colon by decreasing TJ protein expression. The present study may be a useful reference for therapies aiming to regulate the effects of stress on intestinal inflammatory dysfunction.
Collapse
Affiliation(s)
- Nancy Machorro-Rojas
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, Mexico City 04960, Mexico
| | - Teresita Sainz-Espuñes
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, Mexico City 04960, Mexico
| | - Marycarmen Godínez-Victoria
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | | | - Rafael Campos-Rodríguez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Judith Pacheco-Yepez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Maria Elisa Drago-Serrano
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, Mexico City 04960, Mexico
| |
Collapse
|
36
|
Aldriwesh M, Al-Dayan N, Barratt J, Freestone P. The Iron Biology Status of Peritoneal Dialysis Patients May Be a Risk Factor for Development of Infectious Peritonitis. Perit Dial Int 2019; 39:362-374. [PMID: 31123076 DOI: 10.3747/pdi.2018.00052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 01/18/2019] [Indexed: 01/09/2023] Open
Abstract
Background:Infectious peritonitis is a clinically important condition contributing to the significant mortality and morbidity rates observed in peritoneal dialysis (PD) patients. Although some of the socioeconomic risk factors for PD-associated peritonitis have been identified, it is still unclear why certain patients are more susceptible than others to infection.Methods:We examined the molecular components of human peritoneal dialysate (HPD) in an attempt to identify factors that might increase patient susceptibility to infection. Characterization studies were performed on initial and follow-up dialysate samples collected from 9 renal failure patients on PD.Results:Our in vitro data showed that peritonitis-causing bacteria grew differently in the patient dialysates. Proteomic analysis identified an association between transferrin presence and infection risk, as peritoneal transferrin was discovered to be iron-saturated, which was in marked contrast to transferrin in blood. Further, use of radioactive iron-labeled transferrin showed peritoneal transferrin could act as a direct iron source for the growth of peritonitis-causing bacteria. We also found catecholamine stress hormones noradrenaline and adrenaline were present in the dialysates and were apparently involved in enhancing the growth of the bacteria via transferrin iron provision. This suggests the iron biology status of the PD patient may be a risk factor for development of infectious peritonitisConclusions:Collectively, our study suggests transferrin and catecholamines within peritoneal dialysate may be indicators of the potential for bacterial growth in HPD and, as infection risk factors, represent possible future targets for therapeutic manipulation.
Collapse
Affiliation(s)
- Marwh Aldriwesh
- College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Noura Al-Dayan
- Department of Medical Laboratory, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Jonathan Barratt
- John Walls Renal Unit, Leicester General Hospital, Leicester, UK.,Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Primrose Freestone
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| |
Collapse
|
37
|
Influences of stress hormones on microbial infections. Microb Pathog 2019; 131:270-276. [PMID: 30981718 DOI: 10.1016/j.micpath.2019.04.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 04/08/2019] [Accepted: 04/08/2019] [Indexed: 02/08/2023]
Abstract
Stress hormones have been recently suggested to influence the pathogenicity of bacteria significantly. Stress has been identified as part of the factors causing an outbreak of infections in the aquaculture industry. The most studied neuroendocrine hormonal family from a microbial endocrinology perspective is the catecholamine comprising of norepinephrine, epinephrine, and dopamine. It is of importance that catecholamine affects the growth and virulence of bacteria. The influence of stress on bacterial infections is attributed to the ability of catecholamines to suppress the immune system as the mode of action for increased bacterial growth. Catecholamines have increased the growth of bacteria, virulence-associated factors, adhesions, and biofilm formation and consequently influence the outcome of infections by these bacteria in many hosts. The siderophores and the ferric iron transport system plays a vital role in the mechanism by which catecholamines stimulates growth and exposure of genes to stress hormones enhances the expression of genes involved in bacterial virulence. In recent years, it has been discovered that intestinal microflora takes part in bidirectional communication between the gut and brain. The rapidly growing field of microbiome research, understanding the communities of bacteria living within our bodies and the genes they contain is yielding new perspectives. This review reveals catecholamines effects on the growth and virulence of bacteria and the latest trends in microbial endocrinology.
Collapse
|
38
|
Chawla LS, Beers-Mulroy B, Tidmarsh GF. Therapeutic Opportunities for Hepcidin in Acute Care Medicine. Crit Care Clin 2019; 35:357-374. [DOI: 10.1016/j.ccc.2018.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
39
|
Conroy BS, Grigg JC, Kolesnikov M, Morales LD, Murphy MEP. Staphylococcus aureus heme and siderophore-iron acquisition pathways. Biometals 2019; 32:409-424. [PMID: 30911924 DOI: 10.1007/s10534-019-00188-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/18/2019] [Indexed: 11/24/2022]
Abstract
Staphylococcus aureus is a versatile opportunistic human pathogen. Infection by this bacterium requires uptake of iron from the human host, but iron is highly restricted in this environment. Staphylococcus aureus iron sufficiency is achieved primarily through uptake of heme and high-affinity iron chelators, known as siderophores. Two siderophores (staphyloferrins) are produced and secreted by S. aureus into the extracellular environment to capture iron. Staphylococcus aureus expresses specific uptake systems for staphyloferrins and more general uptake systems for siderophores produced by other microorganisms. The S. aureus heme uptake system uses highly-specific cell surface receptors to extract heme from hemoglobin and hemoglobin-haptoglobin complexes for transport into the cytoplasm where it is degraded to liberate iron. Initially thought to be independent systems, recent findings indicate that these iron uptake pathways intersect. IruO is a reductase that releases iron from heme and some ferric-siderophores. Moreover, multifunctional SbnI produces a precursor for staphyloferrin B biosynthesis, and also binds heme to regulate expression of the staphyloferrin B biosynthesis pathway. Intersection of the S. aureus iron uptake pathways is hypothesized to be important for rapid adaptation to available iron sources. Components of the heme and siderophore uptake systems are currently being targeted in the development of therapeutics against S. aureus.
Collapse
Affiliation(s)
- Brigid S Conroy
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada
| | - Jason C Grigg
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada
| | - Maxim Kolesnikov
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada
| | - L Daniela Morales
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada
| | - Michael E P Murphy
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada.
| |
Collapse
|
40
|
Dopamine Is a Siderophore-Like Iron Chelator That Promotes Salmonella enterica Serovar Typhimurium Virulence in Mice. mBio 2019; 10:mBio.02624-18. [PMID: 30723125 PMCID: PMC6428752 DOI: 10.1128/mbio.02624-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We have recently shown that the catecholamine dopamine regulates cellular iron homeostasis in macrophages. As iron is an essential nutrient for microbes, and intracellular iron availability affects the growth of intracellular bacteria, we studied whether dopamine administration impacts the course of Salmonella infections. Dopamine was found to promote the growth of Salmonella both in culture and within bone marrow-derived macrophages, which was dependent on increased bacterial iron acquisition. Dopamine administration to mice infected with Salmonella enterica serovar Typhimurium resulted in significantly increased bacterial burdens in liver and spleen, as well as reduced survival. The promotion of bacterial growth by dopamine was independent of the siderophore-binding host peptide lipocalin-2. Rather, dopamine enhancement of iron uptake requires both the histidine sensor kinase QseC and bacterial iron transporters, in particular SitABCD, and may also involve the increased expression of bacterial iron uptake genes. Deletion or pharmacological blockade of QseC reduced but did not abolish the growth-promoting effects of dopamine. Dopamine also modulated systemic iron homeostasis by increasing hepcidin expression and depleting macrophages of the iron exporter ferroportin, which enhanced intracellular bacterial growth. Salmonella lacking all central iron uptake pathways failed to benefit from dopamine treatment. These observations are potentially relevant to critically ill patients, in whom the pharmacological administration of catecholamines to improve circulatory performance may exacerbate the course of infection with siderophilic bacteria.IMPORTANCE Here we show that dopamine increases bacterial iron incorporation and promotes Salmonella Typhimurium growth both in vitro and in vivo These observations suggest the potential hazards of pharmacological catecholamine administration in patients with bacterial sepsis but also suggest that the inhibition of bacterial iron acquisition might provide a useful approach to antimicrobial therapy.
Collapse
|
41
|
Xu D, Zhou S, Sun L. RNA-seq based transcriptional analysis reveals dynamic genes expression profiles and immune-associated regulation under heat stress in Apostichopus japonicus. FISH & SHELLFISH IMMUNOLOGY 2018; 78:169-176. [PMID: 29684611 DOI: 10.1016/j.fsi.2018.04.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/08/2018] [Accepted: 04/18/2018] [Indexed: 06/08/2023]
Abstract
In this study, we explored the gene expression profiles in Apostichopus japonicus under continuous heat stress (6 h, 48 h and 192 h) by applying RNA-seq technique. A total of 676, 1010 and 1083 differentially expressed genes were detected at three heat stress groups respectively, which suggested complex regulation of various biological processes. Then we focused on the changing of immune system under HS in sea cucumbers. Key immune-associated genes were involved in heat stress response, which were classified into six groups: heat shock proteins, transferrin superfamily members, effector genes, proteases, complement system, and pattern recognition receptors and signaling. Moreover, the mRNA expression of the immune-associated genes were validated by the real time PCR. Our results showed that an immunological strategy in this species was developed to confront abrupt elevated temperatures in the environment.
Collapse
Affiliation(s)
- Dongxue Xu
- College of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China.
| | - Shun Zhou
- College of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Lina Sun
- Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| |
Collapse
|
42
|
Boother EJ, Brownlow S, Tighe HC, Bamford KB, Jackson JE, Shovlin CL. Cerebral Abscess Associated With Odontogenic Bacteremias, Hypoxemia, and Iron Loading in Immunocompetent Patients With Right-to-Left Shunting Through Pulmonary Arteriovenous Malformations. Clin Infect Dis 2018; 65:595-603. [PMID: 28430880 PMCID: PMC5849101 DOI: 10.1093/cid/cix373] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/17/2017] [Indexed: 12/18/2022] Open
Abstract
Background Cerebral abscess is a recognized complication of pulmonary arteriovenous malformations (PAVMs) that allow systemic venous blood to bypass the pulmonary capillary bed through anatomic right-to-left shunts. Broader implications and mechanisms remain poorly explored. Methods Between June 2005 and December 2016, at a single institution, 445 consecutive adult patients with computed tomography–confirmed PAVMs (including 403 [90.5%] with hereditary hemorrhagic telangiectasia) were recruited to a prospective series. Multivariate logistic regression was performed and detailed periabscess histories were evaluated to identify potential associations with cerebral abscess. Rates were compared to an earlier nonoverlapping series. Results Thirty-seven of the 445 (8.3%) patients experienced a cerebral abscess at a median age of 50 years (range, 19–76 years). The rate adjusted for ascertainment bias was 27 of 435 (6.2%). Twenty-nine of 37 (78.4%) patients with abscess had no PAVM diagnosis prior to their abscess, a rate unchanged from earlier UK series. Twenty-one of 37 (56.7%) suffered residual neurological deficits (most commonly memory/cognition impairment), hemiparesis, and visual defects. Isolation of periodontal microbes, and precipitating dental and other interventional events, emphasized potential sources of endovascular inoculations. In multivariate logistic regression, cerebral abscess was associated with low oxygen saturation (indicating greater right-to-left shunting); higher transferrin iron saturation index; intravenous iron use for anemia (adjusted odds ratio, 5.4 [95% confidence interval, 1.4–21.1]); male sex; and venous thromboemboli. There were no relationships with anatomic attributes of PAVMs, or red cell indices often increased due to secondary polycythemia. Conclusions Greater appreciation of the risk of cerebral abscess in undiagnosed PAVMs is required. Lower oxygen saturation and intravenous iron may be modifiable risk factors.
Collapse
Affiliation(s)
- Emily J Boother
- NHLI Cardiovascular Sciences, Imperial College London, London, UK.,Imperial College School of Medicine, Imperial College London
| | - Sheila Brownlow
- Respiratory Medicine, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Hannah C Tighe
- Respiratory Medicine, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Kathleen B Bamford
- Department of Microbiology, Hammersmith Campus, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - James E Jackson
- Department of Imaging, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Claire L Shovlin
- NHLI Cardiovascular Sciences, Imperial College London, London, UK.,Respiratory Medicine, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
43
|
Lyte M, Villageliú DN, Crooker BA, Brown DR. Symposium review: Microbial endocrinology-Why the integration of microbes, epithelial cells, and neurochemical signals in the digestive tract matters to ruminant health. J Dairy Sci 2018; 101:5619-5628. [PMID: 29550113 DOI: 10.3168/jds.2017-13589] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 02/07/2018] [Indexed: 12/14/2022]
Abstract
The union of microbiology and neurobiology, which has been termed microbial endocrinology, is defined as the study of the ability of microorganisms to produce and respond to neurochemicals that originate either within the microorganisms themselves or within the host they inhabit. It serves as the basis for an evolutionarily derived method of communication between a host and its microbiota. Mechanisms elucidated by microbial endocrinology give new insight into the ways the microbiota can affect host stress, metabolic efficiency, resistance to disease, and other factors that may prove relevant to the dairy industry.
Collapse
Affiliation(s)
- Mark Lyte
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames 50011.
| | - Daniel N Villageliú
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames 50011
| | - Brian A Crooker
- Department of Animal Science, University of Minnesota, St. Paul 55108
| | - David R Brown
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul 55108
| |
Collapse
|
44
|
Abstract
Iron is an essential micronutrient for both microbes and humans alike. For well over half a century we have known that this element, in particular, plays a pivotal role in health and disease and, most especially, in shaping host-pathogen interactions. Intracellular iron concentrations serve as a critical signal in regulating the expression not only of high-affinity iron acquisition systems in bacteria, but also of toxins and other noted virulence factors produced by some major human pathogens. While we now are aware of many strategies that the host has devised to sequester iron from invading microbes, there are as many if not more sophisticated mechanisms by which successful pathogens overcome nutritional immunity imposed by the host. This review discusses some of the essential components of iron sequestration and scavenging mechanisms of the host, as well as representative Gram-negative and Gram-positive pathogens, and highlights recent advances in the field. Last, we address how the iron acquisition strategies of pathogenic bacteria may be exploited for the development of novel prophylactics or antimicrobials.
Collapse
|
45
|
Daily propranolol administration reduces persistent injury-associated anemia after severe trauma and chronic stress. J Trauma Acute Care Surg 2017; 82:714-721. [PMID: 28099381 DOI: 10.1097/ta.0000000000001374] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND After severe trauma, patients develop a norepinephrine-mediated persistent, injury-associated anemia. This anemia is associated with suppression of bone marrow (BM) erythroid colony growth, along with decreased iron levels, and elevated erythropoietin (EPO) levels, which are insufficient to promote effective erythropoiesis. The impact of norepinephrine on iron regulators, such as ferroportin, transferrin, and transferrin receptor-1 (TFR-1), is unknown. Using a clinically relevant rodent model of lung contusion (LC), hemorrhagic shock (HS), and chronic stress (CS), we hypothesize that daily propranolol (BB), a nonselective β blocker, restores BM function and improves iron homeostasis. METHODS Male Sprague-Dawley rats were subjected to LCHS ± BB and LCHS/CS ± BB. BB was achieved with propranolol (10 mg/kg) daily until the day of sacrifice. Hemoglobin, plasma EPO, plasma hepcidin, BM cellularity and BM erythroid colony growth were assessed. RNA was isolated to measure transferrin, TFR-1 and ferroportin expression. Data are presented as mean ± SD; *p < 0.05 versus untreated counterpart by t test. RESULTS The addition of CS to LCHS leads to persistent anemia on posttrauma day 7, while the addition of BB improved hemoglobin levels (LCHS/CS: 10.6 ± 0.8 vs. LCHS/CS + BB: 13.9 ± 0.4* g/dL). Daily BB use after LCHS/CS improved BM cellularity, colony-forming units granulocyte, erythrocyte, monocyte megakaryocyte, burst-forming unit erythroid and colony-forming unit erythroid cell colony growth. LCHS/CS + BB significantly reduced plasma EPO levels and increased plasma hepcidin levels on day 7. The addition of CS to LCHS resulted in decreased liver ferroportin expression as well as decreased BM transferrin and TFR-1 expression, thus, blocking iron supply to erythroid cells. However, daily BB after LCHS/CS improved expression of all iron regulators. CONCLUSION Daily propranolol administration after LCHS/CS restored BM function and improved anemia after severe trauma. In addition, iron regulators are significantly reduced after LCHS/CS, which may contribute to iron restriction after injury. However, daily propranolol administration after LCHS/CS improved iron homeostasis.
Collapse
|
46
|
Hu W, Yu X, Liu Z, Sun Y, Chen X, Yang X, Li X, Lam WK, Duan Y, Cao X, Steller H, Liu K, Huang P. The complex of TRIP-Br1 and XIAP ubiquitinates and degrades multiple adenylyl cyclase isoforms. eLife 2017; 6. [PMID: 28656888 PMCID: PMC5503512 DOI: 10.7554/elife.28021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 06/28/2017] [Indexed: 12/03/2022] Open
Abstract
Adenylyl cyclases (ACs) generate cAMP, a second messenger of utmost importance that regulates a vast array of biological processes in all kingdoms of life. However, almost nothing is known about how AC activity is regulated through protein degradation mediated by ubiquitination or other mechanisms. Here, we show that transcriptional regulator interacting with the PHD-bromodomain 1 (TRIP-Br1, Sertad1), a newly identified protein with poorly characterized functions, acts as an adaptor that bridges the interaction of multiple AC isoforms with X-linked inhibitor of apoptosis protein (XIAP), a RING-domain E3 ubiquitin ligase. XIAP ubiquitinates a highly conserved Lys residue in AC isoforms and thereby accelerates the endocytosis and degradation of multiple AC isoforms in human cell lines and mice. XIAP/TRIP-Br1-mediated degradation of ACs forms part of a negative-feedback loop that controls the homeostasis of cAMP signaling in mice. Our findings reveal a previously unrecognized mechanism for degrading multiple AC isoforms and modulating the homeostasis of cAMP signaling. DOI:http://dx.doi.org/10.7554/eLife.28021.001
Collapse
Affiliation(s)
- Wenbao Hu
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Xiaojie Yu
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Zhengzhao Liu
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Ying Sun
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Xibing Chen
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Xin Yang
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Xiaofen Li
- Division of Biomedical Engineering, Hong Kong University of Science and Technology, Hong Kong, China
| | - Wai Kwan Lam
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Yuanyuan Duan
- Division of Biomedical Engineering, Hong Kong University of Science and Technology, Hong Kong, China
| | - Xu Cao
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Hermann Steller
- Strang Laboratory of Apoptosis and Cancer Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Kai Liu
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China.,State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Pingbo Huang
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China.,Division of Biomedical Engineering, Hong Kong University of Science and Technology, Hong Kong, China.,State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
47
|
Mackos AR, Maltz R, Bailey MT. The role of the commensal microbiota in adaptive and maladaptive stressor-induced immunomodulation. Horm Behav 2017; 88:70-78. [PMID: 27760302 PMCID: PMC5303636 DOI: 10.1016/j.yhbeh.2016.10.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/13/2016] [Accepted: 10/14/2016] [Indexed: 02/06/2023]
Abstract
Over the past decade, it has become increasingly evident that there are extensive bidirectional interactions between the body and its microbiota. These interactions are evident during stressful periods, where it is recognized that commensal microbiota community structure is significantly changed. Many different stressors, ranging from early life stressors to stressors administered during adulthood, lead to significant, community-wide differences in the microbiota. The mechanisms through which this occurs are not yet known, but it is known that commensal microbes can recognize, and respond to, mammalian hormones and neurotransmitters, including those that are involved with the physiological response to stressful stimuli. In addition, the physiological stress response also changes many aspects of gastrointestinal physiology that can impact microbial community composition. Thus, there are many routes through which microbial community composition might be disrupted during stressful periods. The implications of these disruptions in commensal microbial communities for host health are still not well understood, but the commensal microbiota have been linked to stressor-induced immunopotentiation. The role of the microbiota in stressor-induced immunopotentiation can be adaptive, such as when these microbes stimulate innate defenses against bacterial infection. However, the commensal microbiota can also lead to maladaptive immune responses during stressor-exposure. This is evident in animal models of colonic inflammation where stressor exposure increases the inflammation through mechanisms involving the microbiota. It is likely that during stressor exposure, immune cell functioning is regulated by combined effects of both neurotransmitters/hormones and commensal microbes. Defining this regulation should be a focus of future studies.
Collapse
Affiliation(s)
- Amy R Mackos
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, United States.
| | - Ross Maltz
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, United States; Department of Gastroenterology, Nationwide Children's Hospital, Columbus, OH 43205, United States
| | - Michael T Bailey
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, United States; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, United States
| |
Collapse
|
48
|
Abstract
Transfusion of rbc is a routine, often lifesaving procedure that depends on a stored supply of blood. In the US, 42 days is the maximum duration allowed for rbc storage; however, several lines of evidence indicate that patients that receive blood at the upper end of this storage limit are at a higher risk of morbidity and mortality. In this issue of the JCI, Rapido and colleagues evaluated the effects of transfusing one unit of blood close to the storage limit into healthy adults. Compared to those that received rbc stored for five weeks or less, those that received blood stored for six weeks showed several effects associated with increased harm, including disruption in iron handling, increased extravascular hemolysis, and the formation of circulating non-transferrin-bound iron. Together, the results of this study suggest that current maximum storage durations should be carefully reevaluated.
Collapse
|
49
|
Pretorius E, Akeredolu OO, Soma P, Kell DB. Major involvement of bacterial components in rheumatoid arthritis and its accompanying oxidative stress, systemic inflammation and hypercoagulability. Exp Biol Med (Maywood) 2016; 242:355-373. [PMID: 27889698 PMCID: PMC5298544 DOI: 10.1177/1535370216681549] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We review the evidence that infectious agents, including those that become dormant within the host, have a major role to play in much of the etiology of rheumatoid arthritis and the inflammation that is its hallmark. This occurs in particular because they can produce cross-reactive (auto-)antigens, as well as potent inflammagens such as lipopolysaccharide that can themselves catalyze further inflammagenesis, including via β-amyloid formation. A series of observables coexist in many chronic, inflammatory diseases as well as rheumatoid arthritis. They include iron dysregulation, hypercoagulability, anomalous morphologies of host erythrocytes, and microparticle formation. Iron dysregulation may be responsible for the periodic regrowth and resuscitation of the dormant bacteria, with concomitant inflammagen production. The present systems biology analysis benefits from the philosophical idea of "coherence," that reflects the principle that if a series of ostensibly unrelated findings are brought together into a self-consistent narrative, that narrative is thereby strengthened. As such, we provide a coherent and testable narrative for the major involvement of (often dormant) bacteria in rheumatoid arthritis.
Collapse
Affiliation(s)
- Etheresia Pretorius
- 1 Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, Pretoria 0007, South Africa
| | - Oore-Ofe Akeredolu
- 1 Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, Pretoria 0007, South Africa
| | - Prashilla Soma
- 1 Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, Pretoria 0007, South Africa
| | - Douglas B Kell
- 2 School of Chemistry, The University of Manchester, Manchester, M13 9PL, UK.,3 The Manchester Institute of Biotechnology, The University of Manchester, Manchester, M1 7DN, UK.,4 Centre for Synthetic Biology of Fine and Speciality Chemicals, The University of Manchester, Manchester, M1 7DN, UK
| |
Collapse
|
50
|
Gart EV, Suchodolski JS, Welsh TH, Alaniz RC, Randel RD, Lawhon SD. Salmonella Typhimurium and Multidirectional Communication in the Gut. Front Microbiol 2016; 7:1827. [PMID: 27920756 PMCID: PMC5118420 DOI: 10.3389/fmicb.2016.01827] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 10/31/2016] [Indexed: 12/20/2022] Open
Abstract
The mammalian digestive tract is home to trillions of microbes, including bacteria, archaea, protozoa, fungi, and viruses. In monogastric mammals the stomach and small intestine harbor diverse bacterial populations but are typically less populated than the colon. The gut bacterial community (microbiota hereafter) varies widely among different host species and individuals within a species. It is influenced by season of the year, age of the host, stress and disease. Ideally, the host and microbiota benefit each other. The host provides nutrients to the microbiota and the microbiota assists the host with digestion and nutrient metabolism. The resident microbiota competes with pathogens for space and nutrients and, through this competition, protects the host in a phenomenon called colonization resistance. The microbiota participates in development of the host immune system, particularly regulation of autoimmunity and mucosal immune response. The microbiota also shapes gut–brain communication and host responses to stress; and, indeed, the microbiota is a newly recognized endocrine organ within mammalian hosts. Salmonella enterica serovar Typhimurium (S. Typhimurium hereafter) is a food-borne pathogen which adapts to and alters the gastrointestinal (GI) environment. In the GI tract, S. Typhimurium competes with the microbiota for nutrients and overcomes colonization resistance to establish infection. To do this, S. Typhimurium uses multiple defense mechanisms to resist environmental stressors, like the acidic pH of the stomach, and virulence mechanisms which allow it to invade the intestinal epithelium and disseminate throughout the host. To coordinate gene expression and disrupt signaling within the microbiota and between host and microbiota, S. Typhimurium employs its own chemical signaling and may regulate host hormone metabolism. This review will discuss the multidirectional interaction between S. Typhimurium, host and microbiota as well as mechanisms that allow S. Typhimurium to succeed in the gut.
Collapse
Affiliation(s)
- Elena V Gart
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station TX, USA
| | - Jan S Suchodolski
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Texas A&M University, College Station TX, USA
| | - Thomas H Welsh
- Department of Animal Science, College of Agriculture and Life Sciences, Texas A&M University, College Station TX, USA
| | - Robert C Alaniz
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, College Station TX, USA
| | | | - Sara D Lawhon
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station TX, USA
| |
Collapse
|