1
|
Russell C, Amankwaa AO. The persistence of bloodstains on buried fabrics and robustness of the leucomalachite green test in detecting aged bloodstains on buried fabrics. Forensic Sci Int 2025; 370:112463. [PMID: 40220723 DOI: 10.1016/j.forsciint.2025.112463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/05/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Understanding the persistence and detection of bloodstains in different casework scenarios can inform the development of appropriate examination strategies, prioritisation of exhibits for DNA analysis, interpretation of test results, and evidence evaluation. This study investigated how soil burial impacts the persistence and detection of bloodstains on cotton, polyester, and wool fabrics over 9 weeks. Bloodstained fabric samples were buried in topsoil and evaluated weekly using the Leucomalachite Green (LMG) test, rated on a scale from 1 to 5 for colour intensity. Surface-level fabrics were compared to buried fabrics to assess burial effects on persistence of bloodstains and performance of the LMG test. Bloodstains on fabrics showed significant degradation and loss over a 9-week burial period, with wool being the most persistent, followed by polyester and cotton. Kruskal-Wallis H tests revealed significant differences in colour intensity scores by fabric type and burial duration (p < 0.001). Mann-Whitney U tests indicated significant differences between fabric types (p < 0.001) and notable degradation and loss from week 1 onward (p < 0.001). These findings are vital as it showed that the LMG test is not robust in detecting aged bloodstains from buried cotton and polyester textiles. Further, in combination with other blood markers (such as DNA or RNA), the persistence pattern observed using the LMG test could potentially inform the estimation of time since deposition of stains. The data generated can inform best practices for the prioritisation of buried exhibits for DNA analysis and storage of recovered buried clothing.
Collapse
Affiliation(s)
- Cenya Russell
- Forensic Science Research Group, Department of Applied Sciences, Northumbria University, Ellison Building, Newcastle upon Tyne, England NE1 8ST, United Kingdom
| | - Aaron Opoku Amankwaa
- Forensic Science Research Group, Department of Applied Sciences, Northumbria University, Ellison Building, Newcastle upon Tyne, England NE1 8ST, United Kingdom.
| |
Collapse
|
2
|
Zhang W, Ma Y, Xie Y, Liu X, Tan L, Zhao J, Ni Y, Wang Z, Li C, Xu B. Interaction and cross-contamination potential of prepared beef steak isolates Pseudomonas weihenstephanensis and Macrococcus caseolyticus in biofilms of dual-species. Food Microbiol 2025; 127:104685. [PMID: 39667856 DOI: 10.1016/j.fm.2024.104685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/04/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024]
Abstract
This study evaluated the interactions between single or dual-species biofilms formed by dominant spoilage bacteria P. weihenstephanensis and M. caseolyticus isolated from refrigerated, spoilage prepared beef steaks at 4 °C and elucidated the interactive behavior of biofilm development in dual species. In addition, the relationship between biofilm formation capacity and cross-contamination was analyzed by simulating surface to food contact transfer. The results showed that the two species exhibited synergism as biofilms developed, which was the main mode of interaction observed. Under aerobic conditions, Pseudomonas weihenstephanensis and Macrococcus caseolyticus co-cultured for 96 h showed obvious biofilm formation ability, resulting in greater cross-contamination. Scanning electron microscopy and Confocal laser scanning microscopy showed the formation of flattened dense biofilms in the co-culture. The significant increase in Fe content and decrease in siderophore content of the dual-species biofilm as determined by ICP-MS was attributed to respiratory inhibition resulting in a decrease in the transcription of genes regulating the two-component regulatory system of Macrococcus tyrolyticus SrrAB and an increase in the expression of cytoplasmic hydrolase leading to the rupture of the release of hemoglobin to provide a source of iron for P. weihenstephanensis. The increase of heme content in the supernatant of dual-species and the results of RT-qPCR showed that the gene expression of the heme transport system of P. weihenstephanensis was significantly up-regulated and the siderophore gene expression was decreased, which further revealed that P. weihenstephanensis preferentially uses the heme uptake system to take up the iron source provided by M. caseolyticus for P. weihenstephanensis. Overall, our results provide insight into the complex dynamics of biofilms formed by P. weihenstephanensis and M. caseolyticus, emphasizing that the iron reaction pathway may be a key factor influencing the growth of P. weihenstephanensis biofilms, and that these results will provide a theoretical basis for the control of spoilage of refrigerated foods.
Collapse
Affiliation(s)
- Wendi Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230601, China
| | - Yunhao Ma
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230601, China; College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yong Xie
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230601, China
| | - Xiaoyan Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230601, China
| | - Lijun Tan
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230601, China
| | - Jinsong Zhao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230601, China
| | - Yongsheng Ni
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230601, China
| | - Zhaoming Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230601, China
| | - Cong Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230601, China
| | - Baocai Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230601, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230601, China.
| |
Collapse
|
3
|
Kim SH, Kwak M, Hwang JK, Keum J, Jin HY, Lee CY, Tanpure RS, Kim YJ, Hoh JK, Park JY, Chung W, Jeon BH, Park HK. Altered heme metabolism and hemoglobin concentration due to empirical antibiotics-induced gut dysbiosis in preterm infants. Comput Struct Biotechnol J 2025; 27:937-945. [PMID: 40123796 PMCID: PMC11930222 DOI: 10.1016/j.csbj.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/04/2025] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
Background High-risk infants are usually treated with empirical antibiotics after birth, regardless of the evidence of infection; however, their gut microbiome and metabolome have seldom been studied. This study investigated the influence of antibiotic exposure on the gut microbiome and associated metabolic pathways in term and preterm infants. Methods Thirty-six infants within 10 days of birth who were admitted to a neonatal intensive care unit/newborn nursery unit were divided into four groups based on maturity (gestational age) and use of empirical antibiotics. Genomic DNA was extracted from the fecal samples and underwent high-throughput 16S rRNA amplicon sequencing using the Illumina platforms. Taxonomic classification, diversity analysis, and metagenomic function prediction were performed. Results Preterm infants with empirical antibiotics showed a significantly decreased population of Firmicutes (p = 0.003) and an increased population of Proteobacteria (p < 0.001) compared to other groups. At the genus level, the populations of Raoultella (p = 0.065) and Escherichia (p = 0.052) showed an increased trend. The change in microbial composition was correlated with increased heme biosynthesis and decreased hemoglobin levels. Conclusion Collectively, our finding suggested that empirical antibiotic exposure in preterm infants alters the gut microbiome, potentially leading to adverse health outcomes. This dysbiosis may affect heme metabolism, increasing the risk of anemia in these vulnerable infants. Therefore, antibiotic use should be carefully tailored to minimize potential harm.
Collapse
Affiliation(s)
- Seung Hyun Kim
- Department of Pediatrics, Hanyang University College of Medicine, Seoul 04763, South Korea
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Min‑Jin Kwak
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32611, USA
| | - Jae Kyoon Hwang
- Department of Pediatrics, Hanyang University College of Medicine, Seoul 04763, South Korea
| | - Jihyun Keum
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Seoul 04763, South Korea
| | - Hee Yeon Jin
- Division of Microbiome, Int-Gen Company, Seoul 04799, South Korea
| | - Chan-Yeong Lee
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul 04763, South Korea
| | - Rahul Sadashiv Tanpure
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul 04763, South Korea
| | - Yong Joo Kim
- Department of Pediatrics, Hanyang University College of Medicine, Seoul 04763, South Korea
| | - Jeong-Kyu Hoh
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Seoul 04763, South Korea
| | - Jae Yong Park
- Division of Microbiome, Int-Gen Company, Seoul 04799, South Korea
| | - Woojin Chung
- Department of Civil & Energy System Engineering, Kyonggi University, Suwon 16227, South Korea
| | - Byong-Hun Jeon
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul 04763, South Korea
| | - Hyun-Kyung Park
- Department of Pediatrics, Hanyang University College of Medicine, Seoul 04763, South Korea
| |
Collapse
|
4
|
Olson EG, Bodie AR, Tarcin HA, Rubinelli PM, Applegate SF, Stephens TP, Rothrock MJ, Ricke SC. Comparison of Media for the Detection of Campylobacter jejuni Using a Commercial RT-PCR System. Pathogens 2025; 14:166. [PMID: 40005541 PMCID: PMC11858337 DOI: 10.3390/pathogens14020166] [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: 01/09/2025] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
The accurate quantification of Campylobacter jejuni in poultry samples is critical for ensuring food safety and compliance with regulatory standards. This study evaluated the performance of three enrichment media-Mueller-Hinton Broth (MHB), Bolton's Blood-Free Broth 2x (BFBB2x), and Buffered Peptone Water (BPW)-in supporting C. jejuni detection and quantification using the BAX® Q7-RT PCR system and traditional plate count methods. Results demonstrated high reliability across all media types, with BFBB2x and MHB showing the strongest correlations (R2 = 0.99) for the BAX® system. BFBB2x exhibited the lowest RMSE (0.13), while MHB balanced precision (RMSE = 0.4) with sensitivity. For plate counts, MHB and BPW achieved the highest correlations (R2 = 0.99) and precision (RMSE = 0.26), with MHB demonstrating the lowest detection limit (2.56 log10 CFU/mL) compared to BFBB2x (2.93 log10 CFU/mL) and BPW (3.31 log10 CFU/mL). The findings underscore MHB's robustness as an enrichment medium, offering consistent performance across both molecular and culture-based methods. The current study supports MHB as the more effective medium for the reliable and precise quantification of C. jejuni in poultry-associated matrices, highlighting its utility in minimizing contamination risks and enhancing food safety. Future research should explore its applicability in diverse poultry products and production environments.
Collapse
Affiliation(s)
- Elena G. Olson
- Meat Science and Animal Biologics Discovery Program, Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI 53706, USA
| | - Aaron R. Bodie
- Poultry Science Department, University of Georgia, 120 D W Brooks Dr., Athens, GA 30602, USA
| | - Haley A. Tarcin
- Meat Science and Animal Biologics Discovery Program, Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI 53706, USA
| | - Peter M. Rubinelli
- Department of Food Science, University of Arkansas, 2650 Young Avenue, Fayetteville, AR 72704, USA
| | | | | | - Michael J. Rothrock
- United States Department of Agriculture, Agricultural Research Service, Athens, GA 30605, USA
| | - Steven C. Ricke
- Meat Science and Animal Biologics Discovery Program, Department of Animal and Dairy Sciences, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
5
|
Lee M, Armstrong CM, Smith AT. Characterization of intact FeoB in a lipid bilayer using styrene-maleic acid (SMA) copolymers. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2025; 1867:184404. [PMID: 39694085 PMCID: PMC11725443 DOI: 10.1016/j.bbamem.2024.184404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/12/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
The acquisition of ferrous iron (Fe2+) is crucial for the survival of many pathogenic bacteria living within acidic and/or anoxic conditions such as Vibrio cholerae, the causative agent of the disease cholera. Bacterial pathogens utilize iron as a cofactor to drive essential metabolic processes, and the primary prokaryotic Fe2+ acquisition mechanism is the ferrous iron transport (Feo) system. In V. cholerae, the Feo system comprises two cytosolic proteins (FeoA, FeoC) and a complex, polytopic transmembrane protein (FeoB) that is regulated by an N-terminal soluble domain (NFeoB) with promiscuous NTPase activity. While the soluble components of the Feo system have been frequently studied, very few reports exist on the intact membrane protein FeoB. Moreover, FeoB has been characterize almost exclusively in detergent micelles that can cause protein misfolding, disrupt protein oligomerization, and even dramatically alter protein function. As many of these characteristics of FeoB remain unclear, there is a critical need to characterize FeoB in a more native-like lipid environment. To address this unmet need, we employ styrene-maleic acid (SMA) copolymers to isolate and to characterize V. cholerae FeoB (VcFeoB) encapsulated by a styrene-maleic acid lipid particle (SMALP). In this work, we describe the development of a workflow for the expression and the purification of VcFeoB in a SMALP. Leveraging mass photometry, we explore the oligomerization of FeoB in a lipid bilayer and show that the VcFeoB-SMALP is mostly monomeric, consistent with our previous oligomerization observations in surfo. Finally, we characterize the NTPase activity of VcFeoB in the SMALP and in a detergent (DDM), revealing higher NTPase activity in the presence of the lipid bilayer. When taken together, this report represents the first characterization of any FeoB in a native-like lipid bilayer and provides a viable approach for the future structural characterization of FeoB.
Collapse
Affiliation(s)
- Mark Lee
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Candice M Armstrong
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Aaron T Smith
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD 21250, USA.
| |
Collapse
|
6
|
Roe K. Preeclampsia and eclampsia: the role of hemolytic protozoan iron. Adv Clin Chem 2025; 125:169-194. [PMID: 39988406 DOI: 10.1016/bs.acc.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Organisms as well as pathogens require several transition metals including iron, copper, zinc, manganese, nickel and cobalt, for genetic replication and other cellular functions. Of these, iron is vital and plays a key role in DNA replication, transcription, synthesis of cofactors and other essential enzymes. During infection, iron deprivation, particularly sequestration thereof, represents a unique response against pathogen attack. The host sequesters ferrous (Fe2+) and ferric (Fe3+) iron via lactoferrin binding at mucosal surfaces, transferrin in blood and tissue and ferritin in blood and cytoplasm. Despite this protective mechanism, pathogens can be resilient in obtaining iron. For example, hemolytic protozoan parasites can obtain iron from heme by rupturing red blood cells. Furthermore, earlier pathogens, driven from active to inactive infections by iron deprivation, could now acquire sufficient iron to enable reactivation resulting in chronic inflammation, oxidative stress to organs and/or circulatory hypertension potentially leading to death. This review discusses the impact of hemolytic protozoan parasite infection in reactivation of latent iron-deprived pathogen infections thus explaining two puzzling pregnancy disorders, pre-eclampsia (PE) and eclampsia. The unknown causations of both disorders have created centuries of confusion and killed millions of women worldwide. Furthermore, reduction-oxidation reactions with iron promote additional oxidative stress damage to vital organs, particularly the kidneys, a common symptom in PE and eclampsia.
Collapse
Affiliation(s)
- Kevin Roe
- United States Patent and Trademark Office, San Jose, California, United States.
| |
Collapse
|
7
|
McGregor AK, Wolthers KR. HutZ from Aliivibrio fischeri Inhibits HutW-Mediated Anaerobilin Formation by Sequestering Heme. Biochemistry 2024; 63:3357-3368. [PMID: 39642050 DOI: 10.1021/acs.biochem.4c00624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2024]
Abstract
Anaerobilin synthase catalyzes the decyclization of the heme protoporphyrin ring, an O2-independent reaction that liberates iron and produces the linear tetrapyrrole, anaerobilin. The marine bacterium Aliivibrio fischeri, the enteric pathogen Escherichia coli O157:H7, and the opportunistic oral pathogen Fusobacterium nucleatum encode anaerobilin synthase as part of their heme uptake/utilization operons, designated chu (E. coli O157:H7), hmu (F. nucleatum), and hut (A. fischeri). F. nucleatum and E. coli O157:H7 contain accessory proteins (ChuS, ChuY, and HmuF) encoded in their respective operons that mitigate against the cytotoxicity of labile heme and anaerobilin by functioning in heme trafficking and anaerobilin reduction. However, the hut operon of A. fischeri and other members of the Vibrionaceae family including the enteric pathogen Vibrio cholerae do not contain homologues to these accessory proteins, raising questions as to how members of this family mitigate against anaerobilin and heme toxicity. Herein, we show that HutW (anaerobilin synthase) from A. fischeri produces anaerobilin, but that HutX and HutZ, encoded downstream of HutW, do not catalyze anaerobilin reduction in the presence of excess NAD(P)H, FAD, and FMN. However, we show that HutZ prevents labile heme and anaerobilin cytotoxicity by binding tightly to heme, sequestering it from HutW, and preventing anaerobilin formation. Thus, A. fischeri is seemingly unable to extract iron from heme using the hutWXZ gene products. Our results further suggest that the structurally distinct chu, hmu, and hut operons have functionally converged to protect the cell from anaerobilin accumulation and heme cytotoxicity.
Collapse
Affiliation(s)
- Alexandra K McGregor
- Department of Chemistry, University of British Columbia, Okanagan Campus, 3247 University Way, Kelowna V1V 1V7, Canada
| | - Kirsten R Wolthers
- Department of Chemistry, University of British Columbia, Okanagan Campus, 3247 University Way, Kelowna V1V 1V7, Canada
| |
Collapse
|
8
|
Yu F, Wang Z, Zhang Z, Zhou J, Li J, Chen J, Du G, Zhao X. Biosynthesis, acquisition, regulation, and upcycling of heme: recent advances. Crit Rev Biotechnol 2024; 44:1422-1438. [PMID: 38228501 DOI: 10.1080/07388551.2023.2291339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/10/2023] [Accepted: 11/25/2023] [Indexed: 01/18/2024]
Abstract
Heme, an iron-containing tetrapyrrole in hemoproteins, including: hemoglobin, myoglobin, catalase, cytochrome c, and cytochrome P450, plays critical physiological roles in different organisms. Heme-derived chemicals, such as biliverdin, bilirubin, and phycocyanobilin, are known for their antioxidant and anti-inflammatory properties and have shown great potential in fighting viruses and diseases. Therefore, more and more attention has been paid to the biosynthesis of hemoproteins and heme derivatives, which depends on the adequate heme supply in various microbial cell factories. The enhancement of endogenous biosynthesis and exogenous uptake can improve the intracellular heme supply, but the excess free heme is toxic to the cells. Therefore, based on the heme-responsive regulators, several sensitive biosensors were developed to fine-tune the intracellular levels of heme. In this review, recent advances in the: biosynthesis, acquisition, regulation, and upcycling of heme were summarized to provide a solid foundation for the efficient production and application of high-value-added hemoproteins and heme derivatives.
Collapse
Affiliation(s)
- Fei Yu
- Science Center for Future Foods, Jiangnan University, Wuxi, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
| | - Ziwei Wang
- Science Center for Future Foods, Jiangnan University, Wuxi, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
| | - Zihan Zhang
- Science Center for Future Foods, Jiangnan University, Wuxi, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
| | - Jingwen Zhou
- Science Center for Future Foods, Jiangnan University, Wuxi, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
| | - Jianghua Li
- Science Center for Future Foods, Jiangnan University, Wuxi, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
| | - Jian Chen
- Science Center for Future Foods, Jiangnan University, Wuxi, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
| | - Guocheng Du
- Science Center for Future Foods, Jiangnan University, Wuxi, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi, China
| | - Xinrui Zhao
- Science Center for Future Foods, Jiangnan University, Wuxi, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, Wuxi, China
| |
Collapse
|
9
|
Imelio JA, Trajtenberg F, Mondino S, Zarantonelli L, Vitrenko I, Lemée L, Cokelaer T, Picardeau M, Buschiazzo A. Signal-sensing triggers the shutdown of HemKR, regulating heme and iron metabolism in the spirochete Leptospira biflexa. PLoS One 2024; 19:e0311040. [PMID: 39325783 PMCID: PMC11426443 DOI: 10.1371/journal.pone.0311040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Heme and iron metabolic pathways are highly intertwined, both compounds being essential for key biological processes, yet becoming toxic if overabundant. Their concentrations are exquisitely regulated, including via dedicated two-component systems (TCSs) that sense signals and regulate adaptive responses. HemKR is a TCS present in both saprophytic and pathogenic Leptospira species, involved in the control of heme metabolism. However, the molecular means by which HemKR is switched on/off in a signal-dependent way, are still unknown. Moreover, a comprehensive list of HemKR-regulated genes, potentially overlapped with iron-responsive targets, is also missing. Using the saprophytic species Leptospira biflexa as a model, we now show that 5-aminolevulinic acid (ALA) triggers the shutdown of the HemKR pathway in live cells, and does so by stimulating the phosphatase activity of HemK towards phosphorylated HemR. Phospho~HemR dephosphorylation leads to differential expression of multiple genes, including of heme metabolism and transport systems. Besides the heme-biosynthetic genes hemA and the catabolic hmuO, which we had previously reported as phospho~HemR targets, we now extend the regulon identifying additional genes. Finally, we discover that HemR inactivation brings about an iron-deficit tolerant phenotype, synergistically with iron-responsive signaling systems. Future studies with pathogenic Leptospira will be able to confirm whether such tolerance to iron deprivation is conserved among Leptospira spp., in which case HemKR could play a vital role during infection where available iron is scarce. In sum, HemKR responds to abundance of porphyrin metabolites by shutting down and controlling heme homeostasis, while also contributing to integrate the regulation of heme and iron metabolism in the L. biflexa spirochete model.
Collapse
Affiliation(s)
- Juan Andrés Imelio
- Laboratory of Molecular & Structural Microbiology, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Felipe Trajtenberg
- Laboratory of Molecular & Structural Microbiology, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Sonia Mondino
- Laboratory of Molecular & Structural Microbiology, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | | | - Iakov Vitrenko
- Plateforme Technologique Biomics, C2RT, Institut Pasteur, Université Paris Cité, Paris, France
| | - Laure Lemée
- Plateforme Technologique Biomics, C2RT, Institut Pasteur, Université Paris Cité, Paris, France
| | - Thomas Cokelaer
- Plateforme Technologique Biomics, C2RT, Institut Pasteur, Université Paris Cité, Paris, France
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Paris, France
| | - Mathieu Picardeau
- Biology of Spirochetes Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Alejandro Buschiazzo
- Laboratory of Molecular & Structural Microbiology, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Dept of Microbiology, Institut Pasteur, Université Paris Cité, Paris, France
| |
Collapse
|
10
|
Choi SR, Hassan MA, Britigan BE, Narayanasamy P. Antimicrobial Activity of Gallium(III) Compounds: Pathogen-Dependent Targeting of Multiple Iron/Heme-Dependent Biological Processes. Curr Issues Mol Biol 2024; 46:9149-9161. [PMID: 39194758 DOI: 10.3390/cimb46080541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
Metals play vital roles in biological systems, with iron/heme being essential for cellular and metabolic functions necessary for survival and/or virulence in many bacterial pathogens. Given the rise of bacterial resistance to current antibiotics, there is an urgent need for the development of non-toxic and novel antibiotics that do not contribute to resistance to other antibiotics. Gallium, which mimics iron, has emerged as a promising antimicrobial agent, offering a novel approach to combat bacterial infections. Gallium does not have any known functions in biological systems. Gallium exerts its effects primarily by replacing iron in redox enzymes, effectively inhibiting bacterial growth by targeting multiple iron/heme-dependent biological processes and suppressing the development of drug resistance. The aim of this review is to highlight recent findings on the mechanisms of action of gallium and provide further insights into the development of gallium-based compounds. Understanding the mechanisms underlying gallium's biological activities is crucial for designing drugs that enhance their therapeutic therapies while minimizing side effects, offering promising avenues for the treatment of infectious diseases.
Collapse
Affiliation(s)
- Seoung-Ryoung Choi
- Department of Pathology, Microbiology and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mohammed A Hassan
- Department of Pathology, Microbiology and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bradley E Britigan
- Department of Pathology, Microbiology and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Prabagaran Narayanasamy
- Department of Pathology, Microbiology and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
11
|
Shilpha J, Lee J, Kwon JS, Lee HA, Nam JY, Jang H, Kang WH. An improved bacterial mRNA enrichment strategy in dual RNA sequencing to unveil the dynamics of plant-bacterial interactions. PLANT METHODS 2024; 20:99. [PMID: 38951818 PMCID: PMC11218159 DOI: 10.1186/s13007-024-01227-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 06/22/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Dual RNA sequencing is a powerful tool that enables a comprehensive understanding of the molecular dynamics underlying plant-microbe interactions. RNA sequencing (RNA-seq) poses technical hurdles in the transcriptional analysis of plant-bacterial interactions, especially in bacterial transcriptomics, owing to the presence of abundant ribosomal RNA (rRNA), which potentially limits the coverage of essential transcripts. Therefore, to achieve cost-effective and comprehensive sequencing of the bacterial transcriptome, it is imperative to devise efficient methods for eliminating rRNA and enhancing the proportion of bacterial mRNA. In this study, we modified a strand-specific dual RNA-seq method with the goal of enriching the proportion of bacterial mRNA in the bacteria-infected plant samples. The enriched method involved the sequential separation of plant mRNA by poly A selection and rRNA removal for bacterial mRNA enrichment followed by strand specific RNA-seq library preparation steps. We assessed the efficiency of the enriched method in comparison to the conventional method by employing various plant-bacterial interactions, including both host and non-host resistance interactions with pathogenic bacteria, as well as an interaction with a beneficial rhizosphere associated bacteria using pepper and tomato plants respectively. RESULTS In all cases of plant-bacterial interactions examined, an increase in mapping efficiency was observed with the enriched method although it produced a lower read count. Especially in the compatible interaction with Xanthmonas campestris pv. Vesicatoria race 3 (Xcv3), the enriched method enhanced the mapping ratio of Xcv3-infected pepper samples to its own genome (15.09%; 1.45-fold increase) and the CDS (8.92%; 1.49-fold increase). The enriched method consistently displayed a greater number of differentially expressed genes (DEGs) than the conventional RNA-seq method at all fold change threshold levels investigated, notably during the early stages of Xcv3 infection in peppers. The Gene Ontology (GO) enrichment analysis revealed that the DEGs were predominantly enriched in proteolysis, kinase, serine type endopeptidase and heme binding activities. CONCLUSION The enriched method demonstrated in this study will serve as a suitable alternative to the existing RNA-seq method to enrich bacterial mRNA and provide novel insights into the intricate transcriptomic alterations within the plant-bacterial interplay.
Collapse
Affiliation(s)
- Jayabalan Shilpha
- Department of Horticulture, Division of Applied Life Science (BK21 Four Program), Institute of Agriculture & Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Junesung Lee
- Department of Horticulture, Division of Applied Life Science (BK21 Four Program), Institute of Agriculture & Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Ji-Su Kwon
- Department of Horticulture, Division of Applied Life Science (BK21 Four Program), Institute of Agriculture & Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Hyun-Ah Lee
- Division of Smart Horticulture, Yonam College, Cheonan, 31005, Republic of Korea
| | - Jae-Young Nam
- Department of Horticulture, Division of Applied Life Science (BK21 Four Program), Institute of Agriculture & Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Hakgi Jang
- Department of Horticulture, Division of Applied Life Science (BK21 Four Program), Institute of Agriculture & Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Won-Hee Kang
- Department of Horticulture, Division of Applied Life Science (BK21 Four Program), Institute of Agriculture & Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| |
Collapse
|
12
|
Fortney KR, Brothwell JA, Batteiger TA, Duplantier R, Katz BP, Spinola SM. A Haemophilus ducreyi strain lacking the yfeABCD iron transport system is virulent in human volunteers. Infect Immun 2024; 92:e0005824. [PMID: 38780215 PMCID: PMC11237573 DOI: 10.1128/iai.00058-24] [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: 02/02/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Haemophilus ducreyi causes the genital ulcer disease chancroid and painful cutaneous ulcers in children who live in the tropics. To acquire heme from the host, H. ducreyi expresses a TonB-dependent hemoglobin receptor, HgbA, which is necessary and sufficient for H. ducreyi to progress to the pustular stage of disease in a controlled human infection model. HgbA transports hemoglobin across the outer membrane; how heme is transported across the cytoplasmic membrane is unclear. In previous studies, transcripts encoding the YfeABCD heme transporter were upregulated in experimental lesions caused by H. ducreyi in human volunteers, suggesting the latter may have a role in virulence. Here we constructed a double deletion mutant, 35000HPΔyfeABΔyfeCD, which exhibited growth defects relative to its parent 35000HP in media containing human hemoglobin as an iron source. Five human volunteers were inoculated at three sites on the skin overlying the deltoid with each strain. The results of the trial showed that papules formed at 100% (95% CI, 71.5, 100) at both 35000HP and 35000HPΔyfeABΔyfeCD-inoculated sites (P = 1.0). Pustules formed at 60% (95% CI, 25.9, 94.1) at parent-inoculated sites and 53% (95% CI, 18.3, 88.4) at mutant-inoculated sites (P = 0.79). Thus, the ABC transporter encoded by yfeAB and yfeCD was dispensable for H. ducreyi virulence in humans. In the absence of YfeABCD, H. ducreyi likely utilizes other periplasmic binding proteins and ABC-transporters such as HbpA, SapABCDF, and DppBCDF to shuttle heme from the periplasm into the cytoplasm, underscoring the importance of redundancy of such systems in gram-negative pathogens.
Collapse
Affiliation(s)
- Kate R. Fortney
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Julie A. Brothwell
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Teresa A. Batteiger
- Department of Medicine, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Rory Duplantier
- Department of Medicine, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Barry P. Katz
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Stanley M. Spinola
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
13
|
Wu P, Ong A, O’Brian MR. Bradyrhizobium japonicum HmuP is an RNA-binding protein that positively controls hmuR operon expression by suppression of a negative regulatory RNA element in the 5' untranslated region. Mol Microbiol 2024; 121:1217-1227. [PMID: 38725184 PMCID: PMC11176003 DOI: 10.1111/mmi.15274] [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: 03/07/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 06/14/2024]
Abstract
The hmuR operon encodes proteins for the uptake and utilization of heme as a nutritional iron source in Bradyrhizobium japonicum. The hmuR operon is transcriptionally activated by the Irr protein and is also positively controlled by HmuP by an unknown mechanism. An hmuP mutant does not express the hmuR operon genes nor does it grow on heme. Here, we show that hmuR expression from a heterologous promoter still requires hmuP, suggesting that HmuP does not regulate at the transcriptional level. Replacement of the 5' untranslated region (5'UTR) of an HmuP-independent gene with the hmuR 5'UTR conferred HmuP-dependent expression on that gene. Recombinant HmuP bound an HmuP-responsive RNA element (HPRE) within the hmuR 5'UTR. A 2 nt substitution predicted to destabilize the secondary structure of the HPRE abolished both HmuP binding activity in vitro and hmuR expression in cells. However, deletion of the HPRE partially restored hmuR expression in an hmuP mutant, and it rescued growth of the hmuP mutant on heme. These findings suggest that the HPRE is a negative regulatory RNA element that is suppressed when bound by HmuP to express the hmuR operon.
Collapse
Affiliation(s)
- Peipei Wu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, The University at Buffalo, 955 Main Street, Suite 4102, Buffalo, New York 14203 USA
| | - Alasteir Ong
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, The University at Buffalo, 955 Main Street, Suite 4102, Buffalo, New York 14203 USA
| | - Mark R. O’Brian
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, The University at Buffalo, 955 Main Street, Suite 4102, Buffalo, New York 14203 USA
| |
Collapse
|
14
|
Deng H, Zhang Y, Cai X, Yin Z, Yang Y, Dong Q, Qiu Y, Chen Z. Dual-Targeted Graphitic Cascade Nanozymes for Recognition and Treatment of Helicobacter pylori. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306155. [PMID: 37991257 DOI: 10.1002/smll.202306155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/08/2023] [Indexed: 11/23/2023]
Abstract
Helicobacter pylori (H. pylori) is the major etiological factor of a variety of gastric diseases. However, the treatment of H. pylori is challenged by the destruction of targeted drugs by gastric acid and pepsin. Herein, a dual-targeted cascade catalytic nanozyme PtCo@Graphene@Hemin-2(L-arginine) (PtCo@G@H2A) is designed for the treatment of H. pylori. The dual-targeting ability of PtCo@G@H2A is derived from directly targeting the receptor protein of H. pylori through hemin and responding to the acidic environment to cause charge reversal (protonation of L-arginine) to capture H. pylori, achieving efficient targeting effect. Compared with the single-targeting strategy relying on hemin, the dual-targeting strategy can greatly improve the targeting rate, achieving an increase of 850% targeting rate. At the concentration of NaHCO3 in intestinal fluid, the surface potential of PtCo@G@H2A can be quickly restored to avoid side effects. Meanwhile, PtCo@G@H2A has pH-responsive oxidase-like activity, which can generate nitric oxide (NO) through a cascade catalytic process that first generates reactive oxygen species (ROS) with oxygen, and further oxidizes L-arginine through ROS, realizing a superior acid-selective bactericidal effect. Overall, it proposes a promising strategy for the treatment of H. pylori that maintains high targeting and therapeutic effects in the environment of gastric acid and pepsin.
Collapse
Affiliation(s)
- Hui Deng
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Yi Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Xinqi Cai
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Zhiwei Yin
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Yanxia Yang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Qian Dong
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Ye Qiu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Zhuo Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| |
Collapse
|
15
|
Saichuer P, Khrisanapant P, Senapin S, Rattanarojpong T, Somsoros W, Khunrae P, Sangsuriya P. Evaluate the potential use of TonB-dependent receptor protein as a subunit vaccine against Aeromonas veronii infection in Nile tilapia (Oreochromis niloticus). Protein Expr Purif 2024; 215:106412. [PMID: 38104792 DOI: 10.1016/j.pep.2023.106412] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023]
Abstract
Aeromonas veronii is an emerging bacterial pathogen that causes serious systemic infections in cultured Nile tilapia (Oreochromis niloticus), leading to massive deaths. Therefore, there is an urgent need to identify effective vaccine candidates to control the spread of this emerging disease. TonB-dependent receptor (Tdr) of A. veronii, which plays a role in the virulence factor of the organism, could be useful in terms of protective antigens for vaccine development. This study aims to evaluate the potential use of Tdr protein as a novel subunit vaccine against A. veronii infection in Nile tilapia. The Tdr gene from A. veronii was cloned into the pET28b expression vector, and the recombinant protein was subsequently produced in Escherichia coli strain BL21 (DE3). Tdr was expressed as an insoluble protein and purified by affinity chromatography. Antigenicity test indicated that this protein was recognized by serum from A. veronii infected fish. When Nile tilapia were immunized with the Tdr protein, specific antibody levels increased significantly (p-value <0.05) at 7 days post-immunization (dpi), and peaked at 21 dpi compared to antibody levels at 0 dpi. Furthermore, bacterial agglutination activity was observed in the fish serum immunized with the Tdr protein, indicating that specific antibodies in the serum can detect Tdr on the bacterial cell surface. These results suggest that Tdr protein has potential as a vaccine candidate. However, challenging tests with A.veronii in Nile tilapia needs to be investigated to thoroughly evaluate its protective efficacy for future applications.
Collapse
Affiliation(s)
- Pornpavee Saichuer
- Department of Microbiology, Faculty of Science, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand
| | - Prit Khrisanapant
- Department of Microbiology, Faculty of Science, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand
| | - Saengchan Senapin
- Fish Health Platform, Center of Excellence for Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand; National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, 12120, Thailand
| | - Triwit Rattanarojpong
- Department of Microbiology, Faculty of Science, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand
| | - Wasusit Somsoros
- Department of Microbiology, Faculty of Science, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand
| | - Pongsak Khunrae
- Department of Microbiology, Faculty of Science, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand.
| | - Pakkakul Sangsuriya
- Aquatic Molecular Genetics and Biotechnology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, 12120, Thailand.
| |
Collapse
|
16
|
Mukherjee P, Agarwal S, Mallick SB, Dasgupta J. PAS domain of flagellar histidine kinase FlrB has a unique architecture and binds heme as a sensory ligand in an unconventional fashion. Structure 2024; 32:200-216.e5. [PMID: 38157857 DOI: 10.1016/j.str.2023.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/28/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024]
Abstract
Phosphorylation of the σ54-dependent transcription activator FlrC by the sensor histidine kinase FlrB is essential for flagellar synthesis of Vibrio cholerae. Despite that, the structure, sensory signal, and mechanistic basis of function of FlrB were elusive. Here, we report the crystal structure of the sensory PAS domain of FlrB in its functional dimeric state that exhibits a unique architecture. Series of biochemical/biophysical experiments on different constructs and mutants established that heme binds hydrophobically as sensory ligand in the shallow ligand-binding cleft of FlrB-PAS without axial coordination. Intriguingly, ATP binding to the C-terminal ATP-binding (CA) domain assists PAS domain to bind heme, vis-à-vis, heme binding to the PAS facilitates ATP binding to the CA domain. We hypothesize that synergistic binding of heme and ATP triggers conformational signaling in FlrB, leading to downstream flagellar gene transcription. Enhanced swimming motility of V. cholerae with increased heme uptake supports this proposition.
Collapse
Affiliation(s)
- Peeali Mukherjee
- Department of Biotechnology, St. Xavier's College (Autonomous), 30 Mother Teresa Sarani, Kolkata 700016, India
| | - Shubhangi Agarwal
- Department of Biotechnology, St. Xavier's College (Autonomous), 30 Mother Teresa Sarani, Kolkata 700016, India
| | - Sritapa Basu Mallick
- Department of Biotechnology, St. Xavier's College (Autonomous), 30 Mother Teresa Sarani, Kolkata 700016, India
| | - Jhimli Dasgupta
- Department of Biotechnology, St. Xavier's College (Autonomous), 30 Mother Teresa Sarani, Kolkata 700016, India.
| |
Collapse
|
17
|
Ullah I, Lang M. Key players in the regulation of iron homeostasis at the host-pathogen interface. Front Immunol 2023; 14:1279826. [PMID: 37942316 PMCID: PMC10627961 DOI: 10.3389/fimmu.2023.1279826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/03/2023] [Indexed: 11/10/2023] Open
Abstract
Iron plays a crucial role in the biochemistry and development of nearly all living organisms. Iron starvation of pathogens during infection is a striking feature utilized by a host to quell infection. In mammals and some other animals, iron is essentially obtained from diet and recycled from erythrocytes. Free iron is cytotoxic and is readily available to invading pathogens. During infection, most pathogens utilize host iron for their survival. Therefore, to ensure limited free iron, the host's natural system denies this metal in a process termed nutritional immunity. In this fierce battle for iron, hosts win over some pathogens, but others have evolved mechanisms to overdrive the host barriers. Production of siderophores, heme iron thievery, and direct binding of transferrin and lactoferrin to bacterial receptors are some of the pathogens' successful strategies which are highlighted in this review. The intricate interplay between hosts and pathogens in iron alteration systems is crucial for understanding host defense mechanisms and pathogen virulence. This review aims to elucidate the current understanding of host and pathogen iron alteration systems and propose future research directions to enhance our knowledge in this field.
Collapse
Affiliation(s)
- Inam Ullah
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Minglin Lang
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Beijing, China
- College of Life Science, Agricultural University of Hebei, Baoding, China
| |
Collapse
|
18
|
Guidone GHM, Cardozo JG, Silva LC, Sanches MS, Galhardi LCF, Kobayashi RKT, Vespero EC, Rocha SPD. Epidemiology and characterization of Providencia stuartii isolated from hospitalized patients in southern Brazil: a possible emerging pathogen. Access Microbiol 2023; 5:000652.v4. [PMID: 37970084 PMCID: PMC10634494 DOI: 10.1099/acmi.0.000652.v4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/08/2023] [Indexed: 11/17/2023] Open
Abstract
This study aimed to characterize the virulence factors and antimicrobial resistance of Providencia stuartii , an opportunistic pathogen that causes human infections. We examined 45 isolates of P. stuartii both genotypically and phenotypically by studying their adherence to HeLa cells, biofilm formation, cytotoxicity and antimicrobial resistance, and analysed their genomes for putative virulence and resistance genes. This study found that most isolates possessed multiple virulence genes, including fimA, mrkA, fptA, iutA, ireA and hlyA, and were cytotoxic to Vero cells. All the isolates were resistant to amoxicillin plus clavulanic acid, levofloxacin and sulfamethoxazole plus trimethoprim, and most were resistant to ceftriaxone and cefepime. All isolates harboured extended-spectrum beta-lactamase coding genes such as bla CTX-M-2 and 23/45(51.11 %) of them also harboured bla CTX-M-9. The gene KPC-2 (carbapenemase) was detected in 8/45(17.77 %) isolates. This study also found clonality among the isolates, indicating the possible spread of the pathogen among patients at the hospital. These results have significant clinical and epidemiological implications and emphasize the importance of a continued understanding of the virulence and antimicrobial resistance of this pathogen for the prevention and treatment of future infections.
Collapse
Affiliation(s)
| | - Jennifer Germiniani Cardozo
- Laboratory of Basic and Applied Bacteriology, Department of Microbiology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Luana Carvalho Silva
- Laboratory of Bacteriology, Department of Microbiology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Matheus Silva Sanches
- Laboratory of Bacteriology, Department of Microbiology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Ligia Carla Faccin Galhardi
- Virology Laboratory, Department of Microbiology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Renata Katsuko Takayama Kobayashi
- Laboratory of Basic and Applied Bacteriology, Department of Microbiology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Eliana Carolina Vespero
- Department of Pathology, Clinical and Toxicological Analysis, Health Sciences Center, University Hospital of Londrina, State University of Londrina, Paraná, Brazil
| | - Sergio Paulo Dejato Rocha
- Laboratory of Bacteriology, Department of Microbiology, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| |
Collapse
|
19
|
de Oliveira PV, de Santana Lira RL, de Abreu Lima R, Mendes YC, Martins AB, de Melo BDO, Goiano MF, Filho RL, de Farias Nunes FBB, Aliança ASDS, Firmo WDCA, Carvalho RC, Zagminan A, de Sousa EM. Bibliometric Review on New Possibilities of Antimycobacterial Agents: Exploring Siderophore Desferrioxamine's Applications as an Antimicrobial Agent. Pharmaceuticals (Basel) 2023; 16:1335. [PMID: 37765143 PMCID: PMC10536058 DOI: 10.3390/ph16091335] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/07/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
Mycobacteria cause tuberculosis and other serious diseases. Understanding their mechanisms of resistance to our immune system and exploring novel drugs are critical strategies to combat infections. A bibliometric analysis was performed to identify publication trends and critical research areas in the field of the antimicrobial activity of desferrioxamine. A total of twenty-four publications on the topic, from 2012 to 2023, were retrieved from databases including Web of Science, Scopus, PubMed, and Embase, using specific keywords. The quality of the publications was assessed using impact and productivity metrics, with an average annual publication rate of 2.1 articles. The United States emerged as the most productive country, with medicine (23.4%, 11 publications) and biochemistry, genetics, and molecular biology (21.3%, 10 publications) as the top research fields. The five most cited publications accounted for 672 citations, with a relatively low h-index (11:11). In conclusion, there has been a lack of publications on this topic in the last decade. The United States dominates production and publication in this area, and there appears to be limited exchange of knowledge, ideas, and technology within the field. Therefore, fostering international cooperation through funding is essential to facilitate further research and development of desferrioxamine-related studies.
Collapse
Affiliation(s)
- Patrícia Vieira de Oliveira
- Graduate Program in Microbial Biology, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.V.d.O.); (Y.C.M.); (A.S.d.S.A.); (W.d.C.A.F.); (A.Z.)
| | - Roseane Lustosa de Santana Lira
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (R.d.A.L.); (R.C.C.)
| | - Rafael de Abreu Lima
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (R.d.A.L.); (R.C.C.)
| | - Yasmim Costa Mendes
- Graduate Program in Microbial Biology, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.V.d.O.); (Y.C.M.); (A.S.d.S.A.); (W.d.C.A.F.); (A.Z.)
| | - Antenor Bezerra Martins
- Graduate Program in Health and Services Management, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil;
| | - Bruna de Oliveira de Melo
- Graduate Program in Biodiversity and Biotechnology—BIONORTE Amazonian Network, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil;
| | | | - Rivaldo Lira Filho
- Graduate Program in Nursing, St. Therese College—CEST, São Luís 65045-180, Brazil;
| | | | - Amanda Silva dos Santos Aliança
- Graduate Program in Microbial Biology, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.V.d.O.); (Y.C.M.); (A.S.d.S.A.); (W.d.C.A.F.); (A.Z.)
- Graduate Program in Health and Services Management, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil;
| | - Wellyson da Cunha Araújo Firmo
- Graduate Program in Microbial Biology, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.V.d.O.); (Y.C.M.); (A.S.d.S.A.); (W.d.C.A.F.); (A.Z.)
- Graduate Program in Health and Services Management, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil;
| | - Rafael Cardoso Carvalho
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (R.d.A.L.); (R.C.C.)
| | - Adrielle Zagminan
- Graduate Program in Microbial Biology, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.V.d.O.); (Y.C.M.); (A.S.d.S.A.); (W.d.C.A.F.); (A.Z.)
- Graduate Program in Health and Services Management, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil;
| | - Eduardo Martins de Sousa
- Graduate Program in Microbial Biology, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.V.d.O.); (Y.C.M.); (A.S.d.S.A.); (W.d.C.A.F.); (A.Z.)
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (R.d.A.L.); (R.C.C.)
- Graduate Program in Health and Services Management, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil;
- Graduate Program in Biodiversity and Biotechnology—BIONORTE Amazonian Network, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil;
| |
Collapse
|
20
|
Zhurilov PA, Andriyanov PA, Tutrina AI, Razheva IV, Liskova EA, Gladkova NA, Kashina DD, Yashin IV, Blokhin AA. Characterization and comparative analysis of the Escherichia marmotae M-12 isolate from bank vole (Myodes glareolus). Sci Rep 2023; 13:13949. [PMID: 37626115 PMCID: PMC10457355 DOI: 10.1038/s41598-023-41223-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/23/2023] [Indexed: 08/27/2023] Open
Abstract
The Escherichia marmotae is a bacterium of the Enterobacterales order, which was first isolated from the Himalayan marmot (Marmota himalayana). Recently E. marmotae has been shown to cause severe infections in humans. Wild animals were suggested to be a natural reservoir of this bacterium. The present study describes the first case of E. marmotae isolation from an apparently healthy wild bank vole (Myodes glareolus). Phenotype, as well as genotype-based techniques, were applied to characterize E. marmotae M-12 isolate. E. marmotae M-12 had the capsule-positive phenotype, high adhesion to human erythrocytes and HEp-2 cells as well as a low invasion into HEp-2 cells. E. marmotae M-12 was avirulent in mice. The phylogenomic analyses of E. marmotae showed dispersed phylogenetic structure among isolates of different origins. Virulome analysis of M-12 isolate revealed the presence of the following factors: siderophores, heme uptake systems, capsule synthesis, curli and type I fimbriae, flagella proteins, OmpA porin, etc. Comparative virulome analysis among available E. marmotae genomes revealed the presence of capsule K1 genes mostly in pathogenic isolates and OmpA porin presence among all strains. We assume that the K1 capsule and OmpA porin play a key role in the virulence of E. marmotae. Pathogenesis of the latter might be similar to extraintestinal pathogenic E. coli.
Collapse
Affiliation(s)
- Pavel A Zhurilov
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia.
| | - Pavel A Andriyanov
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Anastasia I Tutrina
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Irina V Razheva
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Elena A Liskova
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Nadezda A Gladkova
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Daria D Kashina
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Ivan V Yashin
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| | - Andrey A Blokhin
- Federal Research Center for Virology and Microbiology, Branch in Nizhny Novgorod, 603950, Nizhny Novgorod, Russia
| |
Collapse
|
21
|
Villalobos-Escobedo JM, Mercado-Esquivias MB, Adams C, Kauffman WB, Malmstrom RR, Deutschbauer AM, Glass NL. Genome-wide fitness profiling reveals molecular mechanisms that bacteria use to interact with Trichoderma atroviride exometabolites. PLoS Genet 2023; 19:e1010909. [PMID: 37651474 PMCID: PMC10516422 DOI: 10.1371/journal.pgen.1010909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 09/22/2023] [Accepted: 08/07/2023] [Indexed: 09/02/2023] Open
Abstract
Trichoderma spp. are ubiquitous rhizosphere fungi capable of producing several classes of secondary metabolites that can modify the dynamics of the plant-associated microbiome. However, the bacterial-fungal mechanisms that mediate these interactions have not been fully characterized. Here, a random barcode transposon-site sequencing (RB-TnSeq) approach was employed to identify bacterial genes important for fitness in the presence of Trichoderma atroviride exudates. We selected three rhizosphere bacteria with RB-TnSeq mutant libraries that can promote plant growth: the nitrogen fixers Klebsiella michiganensis M5aI and Herbaspirillum seropedicae SmR1, and Pseudomonas simiae WCS417. As a non-rhizosphere species, Pseudomonas putida KT2440 was also included. From the RB-TnSeq data, nitrogen-fixing bacteria competed mainly for iron and required the siderophore transport system TonB/ExbB for optimal fitness in the presence of T. atroviride exudates. In contrast, P. simiae and P. putida were highly dependent on mechanisms associated with membrane lipid modification that are required for resistance to cationic antimicrobial peptides (CAMPs). A mutant in the Hog1-MAP kinase (Δtmk3) gene of T. atroviride showed altered expression patterns of many nonribosomal peptide synthetase (NRPS) biosynthetic gene clusters with potential antibiotic activity. In contrast to exudates from wild-type T. atroviride, bacterial mutants containing lesions in genes associated with resistance to antibiotics did not show fitness defects when RB-TnSeq libraries were exposed to exudates from the Δtmk3 mutant. Unexpectedly, exudates from wild-type T. atroviride and the Δtmk3 mutant rescued purine auxotrophic mutants of H. seropedicae, K. michiganensis and P. simiae. Metabolomic analysis on exudates from wild-type T. atroviride and the Δtmk3 mutant showed that both strains excrete purines and complex metabolites; functional Tmk3 is required to produce some of these metabolites. This study highlights the complex interplay between Trichoderma-metabolites and soil bacteria, revealing both beneficial and antagonistic effects, and underscoring the intricate and multifaceted nature of this relationship.
Collapse
Affiliation(s)
- José Manuel Villalobos-Escobedo
- Plant and Microbial Biology Department, The University of California, Berkeley, California, United States of America
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Maria Belen Mercado-Esquivias
- Plant and Microbial Biology Department, The University of California, Berkeley, California, United States of America
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Catharine Adams
- Plant and Microbial Biology Department, The University of California, Berkeley, California, United States of America
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - W. Berkeley Kauffman
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Rex R. Malmstrom
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Adam M. Deutschbauer
- Plant and Microbial Biology Department, The University of California, Berkeley, California, United States of America
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - N. Louise Glass
- Plant and Microbial Biology Department, The University of California, Berkeley, California, United States of America
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| |
Collapse
|
22
|
Zamarreño Beas J, Videira MAM, Karavaeva V, Lourenço FM, Almeida MR, Sousa F, Saraiva LM. In Campylobacter jejuni, a new type of chaperone receives heme from ferrochelatase. Front Genet 2023; 14:1199357. [PMID: 37415606 PMCID: PMC10320005 DOI: 10.3389/fgene.2023.1199357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/02/2023] [Indexed: 07/08/2023] Open
Abstract
Intracellular heme formation and trafficking are fundamental processes in living organisms. Bacteria and archaea utilize three biogenesis pathways to produce iron protoporphyrin IX (heme b) that diverge after the formation of the common intermediate uroporphyrinogen III (uro'gen III). In this study, we identify and provide a detailed characterization of the enzymes involved in the transformation of uro'gen III into heme in Campylobacter jejuni, demonstrating that this bacterium utilizes the protoporphyrin-dependent (PPD) pathway. In general, limited knowledge exists regarding the mechanisms by which heme b reaches its target proteins after this final step. Specifically, the chaperones necessary for trafficking heme to prevent the cytotoxic effects associated with free heme remain largely unidentified. In C. jejuni, we identified a protein named CgdH2 that binds heme with a dissociation constant of 4.9 ± 1.0 µM, and this binding is impaired upon mutation of residues histidine 45 and 133. We demonstrate that C. jejuni CgdH2 establishes protein-protein interactions with ferrochelatase, suggesting its role in facilitating heme transfer from ferrochelatase to CgdH2. Furthermore, phylogenetic analysis reveals that C. jejuni CgdH2 is evolutionarily distinct from the currently known chaperones. Therefore, CgdH2 is the first protein identified as an acceptor of intracellularly formed heme, expanding our knowledge of the mechanisms underlying heme trafficking within bacterial cells.
Collapse
Affiliation(s)
- Jordi Zamarreño Beas
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Marco A. M. Videira
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Val Karavaeva
- Department of Functional and Evolutionary Ecology, University of Vienna, Wien, Austria
| | - Frederico M. Lourenço
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Mafalda R. Almeida
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Filipa Sousa
- Department of Functional and Evolutionary Ecology, University of Vienna, Wien, Austria
| | - Lígia M. Saraiva
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
23
|
Tsiftsoglou SA. Heme Interactions as Regulators of the Alternative Pathway Complement Responses and Implications for Heme-Associated Pathologies. Curr Issues Mol Biol 2023; 45:5198-5214. [PMID: 37367079 DOI: 10.3390/cimb45060330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023] Open
Abstract
Heme (Fe2+-protoporphyrin IX) is a pigment of life, and as a prosthetic group in several hemoproteins, it contributes to diverse critical cellular processes. While its intracellular levels are tightly regulated by networks of heme-binding proteins (HeBPs), labile heme can be hazardous through oxidative processes. In blood plasma, heme is scavenged by hemopexin (HPX), albumin and several other proteins, while it also interacts directly with complement components C1q, C3 and factor I. These direct interactions block the classical pathway (CP) and distort the alternative pathway (AP). Errors or flaws in heme metabolism, causing uncontrolled intracellular oxidative stress, can lead to several severe hematological disorders. Direct interactions of extracellular heme with alternative pathway complement components (APCCs) may be implicated molecularly in diverse conditions at sites of abnormal cell damage and vascular injury. In such disorders, a deregulated AP could be associated with the heme-mediated disruption of the physiological heparan sulphate-CFH coat of stressed cells and the induction of local hemostatic responses. Within this conceptual frame, a computational evaluation of HBMs (heme-binding motifs) aimed to determine how heme interacts with APCCs and whether these interactions are affected by genetic variation within putative HBMs. Combined computational analysis and database mining identified putative HBMs in all of the 16 APCCs examined, with 10 exhibiting disease-associated genetic (SNPs) and/or epigenetic variation (PTMs). Overall, this article indicates that among the pleiotropic roles of heme reviewed, the interactions of heme with APCCs could induce differential AP-mediated hemostasis-driven pathologies in certain individuals.
Collapse
Affiliation(s)
- Stefanos A Tsiftsoglou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
24
|
Simpson AC, Eedara VVR, Singh NK, Damle N, Parker CW, Karouia F, Mason CE, Venkateswaran K. Comparative genomic analysis of Cohnella hashimotonis sp. nov. isolated from the International Space Station. Front Microbiol 2023; 14:1166013. [PMID: 37396358 PMCID: PMC10308117 DOI: 10.3389/fmicb.2023.1166013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/24/2023] [Indexed: 07/04/2023] Open
Abstract
A single strain from the family Paenibacillaceae was isolated from the wall behind the Waste Hygiene Compartment aboard the International Space Station (ISS) in April 2018, as part of the Microbial Tracking mission series. This strain was identified as a gram-positive, rod-shaped, oxidase-positive, catalase-negative motile bacterium in the genus Cohnella, designated as F6_2S_P_1T. The 16S sequence of the F6_2S_P_1T strain places it in a clade with C. rhizosphaerae and C. ginsengisoli, which were originally isolated from plant tissue or rhizosphere environments. The closest 16S and gyrB matches to strain F6_2S_P_1T are to C. rhizosphaerae with 98.84 and 93.99% sequence similarity, while a core single-copy gene phylogeny from all publicly available Cohnella genomes places it as more closely related to C. ginsengisoli. Average nucleotide identity (ANI) and digital DNA-DNA hybridization (dDDH) values to any described Cohnella species are <89 and <22%, respectively. The major fatty acids for strain F6_2S_P_1T are anteiso-C15:0 (51.7%), iso-C16:0 (23.1%), and iso-C15:0 (10.5%), and it is able to metabolize a wide range of carbon compounds. Given the results of the ANI and dDDH analyses, this ISS strain is a novel species within the genus Cohnella for which we propose the name Cohnella hashimotonis, with the type strain F6_2S_P_1T (=NRRL B-65657T and DSMZ 115098T). Because no closely related Cohnella genomes were available, this study generated the whole-genome sequences (WGSs) of the type strains for C. rhizosphaerae and C. ginsengisoli. Phylogenetic and pangenomic analysis reveals that F6_2S_P_1T, C. rhizosphaerae, and C. ginsengisoli, along with two uncharacterized Cohnella strains, possess a shared set of 332 gene clusters which are not shared with any other WGS of Cohnella species, and form a distinct clade branching off from C. nanjingensis. Functional traits were predicted for the genomes of strain F6_2S_P_1T and other members of this clade.
Collapse
Affiliation(s)
- Anna C. Simpson
- California Institute of Technology, Jet Propulsion Laboratory, Pasadena, CA, United States
| | - V. V. Ramprasad Eedara
- Department of Plant Science, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Nitin K. Singh
- California Institute of Technology, Jet Propulsion Laboratory, Pasadena, CA, United States
| | - Namita Damle
- Department of Physiology and Biophysics, and the WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, United States
| | - Ceth W. Parker
- California Institute of Technology, Jet Propulsion Laboratory, Pasadena, CA, United States
| | | | - Christopher E. Mason
- Department of Physiology and Biophysics, and the WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, United States
| | - Kasthuri Venkateswaran
- California Institute of Technology, Jet Propulsion Laboratory, Pasadena, CA, United States
| |
Collapse
|
25
|
Sankey N, Merrick H, Singh P, Rogers J, Reddi A, Hartson SD, Mitra A. Role of the Mycobacterium tuberculosis ESX-4 Secretion System in Heme Iron Utilization and Pore Formation by PPE Proteins. mSphere 2023; 8:e0057322. [PMID: 36749044 PMCID: PMC10117145 DOI: 10.1128/msphere.00573-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/16/2023] [Indexed: 02/08/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is transmitted through aerosols and primarily colonizes within the lung. The World Health Organization estimates that Mtb kills ~1.4 million people every year. A key aspect that makes Mtb such a successful pathogen is its ability to overcome iron limitation mounted by the host immune response. In our previous studies, we have shown that Mtb can utilize iron from heme, the largest source of iron in the human host, and that it uses two redundant heme utilization pathways. In this study, we show that the ESX-4 type VII secretion system (T7SS) is necessary for extracellular heme uptake into the Mtb cell through both heme utilization pathways. ESX-4 influences the secretion of the culture filtrate proteins Rv0125 and Rv1085c, which are also necessary for efficient heme utilization. We also discovered that deletion of the alternative sigma factor SigM significantly reduced Mtb heme utilization through both pathways and predict that SigM is a global positive regulator of core heme utilization genes of both pathways. Finally, we present the first direct evidence that some mycobacterial PPE (proline-proline-glutamate motif) proteins of the PPE protein family are pore-forming membrane proteins. Altogether, we identified core components of both Mtb Heme utilization pathways that were previously unknown and identified a novel channel-forming membrane protein of Mtb. IMPORTANCE M. tuberculosis (Mtb) is completely dependent on iron acquisition in the host to cause disease. The largest source of iron for Mtb in the human host is heme. Here, we show that the ancestral ESX-4 type VII secretion system is required for the efficient utilization of heme as a source of iron, which is an essential nutrient. This is another biological function identified for ESX-4 in Mtb, whose contribution to Mtb physiology is poorly understood. A most exciting finding is that some mycobacterial PPE (proline-proline-glutamate motif) proteins that have been implicated in the nutrient acquisition are membrane proteins that can form channels in a lipid bilayer. These observations have far-reaching implications because they support an emerging theme that PPE proteins can function as channel proteins in the outer mycomembrane for nutrient acquisition. Mtb has evolved a heme uptake system that is drastically different from all other known bacterial heme acquisition systems.
Collapse
Affiliation(s)
- November Sankey
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Haley Merrick
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Padam Singh
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Janet Rogers
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Amit Reddi
- School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Steven D. Hartson
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Avishek Mitra
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
26
|
Zhao J, Zhao B, Kong N, Li F, Liu J, Wang L, Song L. Increased abundances of potential pathogenic bacteria and expressions of inflammatory cytokines in the intestine of oyster Crassostrea gigas after high temperature stress. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 141:104630. [PMID: 36603795 DOI: 10.1016/j.dci.2022.104630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 06/17/2023]
Abstract
High temperature stress is a significant threat to the health of oysters, but the effects on their intestinal performances are not well understood. In this study, the effects of high temperature stress on the intestinal histology, immune response and associated microbiota were investigated in Crassostrea gigas after rearing at 20, 25 and 28 °C for 21 days. With the increase of temperature, shortened and shed microvilli as well as increased goblet cells were observed in the intestines of oysters. The transcripts of cytokines CgIL17-5, CgTNF-2 and CgTGF-β and apoptosis-related gene CgCaspase-3 in intestine increased with the increasing temperature. Further, the diversity and composition of the oyster intestinal microbiota changed after high temperature stress. The 16S rRNA gene copy number per ng of DNA in the T25 (5.16 × 105) and T28 (1.63 × 105) groups were higher than that in the control group (8.62 × 104). The Chao 1 index in the T25 (238.00) and T28 (240.17) groups was lower than that in the control group (279.00). The Shannon index decreased progressively with the increasing temperature, with the value in the T28 group (4.44) significantly lower than that in the control group (5.40) (p < 0.05). The abundances of potential pathogenic bacteria such as Acinetobacter, Pseudomonas, Vibrio and Endozoicomonas increased while that of probiotic bacteria Bacillus decreased after high temperature exposure. Functional prediction indicated that the pathways associated with bacterial proliferation were enriched at 25 °C, while those involved in protein synthesis were blocked at 28 °C. Collectively, these results suggested that high temperature stress led to an increased abundances of potential pathogenic bacteria and expressions of inflammatory cytokines in the intestine, which may consequently affect the functional integrity of the intestinal barrier in oysters.
Collapse
Affiliation(s)
- Junyan Zhao
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Bao Zhao
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Ning Kong
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
| | - Fuzhe Li
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Jinyu Liu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| |
Collapse
|
27
|
Meslé MM, Gray CR, Dlakić M, DuBois JL. Bacteroides thetaiotaomicron, a Model Gastrointestinal Tract Species, Prefers Heme as an Iron Source, Yields Protoporphyrin IX as a Product, and Acts as a Heme Reservoir. Microbiol Spectr 2023; 11:e0481522. [PMID: 36862015 PMCID: PMC10100974 DOI: 10.1128/spectrum.04815-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/03/2023] [Indexed: 03/03/2023] Open
Abstract
Members of the phylum Bacteroidetes are abundant in healthy gastrointestinal (GI) tract flora. Bacteroides thetaiotaomicron is a commensal heme auxotroph and representative of this group. Bacteroidetes are sensitive to host dietary iron restriction but proliferate in heme-rich environments that are also associated with colon cancer. We hypothesized that B. thetaiotaomicron may act as a host reservoir for iron and/or heme. In this study, we defined growth-promoting quantities of iron for B. thetaiotaomicron. B. thetaiotaomicron preferentially consumed and hyperaccumulated iron in the form of heme when presented both heme and nonheme iron sources in excess of its growth needs, leading to an estimated 3.6 to 8.4 mg iron in a model GI tract microbiome consisting solely of B. thetaiotaomicron. Protoporphyrin IX was identified as an organic coproduct of heme metabolism, consistent with anaerobic removal of iron from the heme leaving the intact tetrapyrrole as the observed product. Notably, no predicted or discernible pathway for protoporphyrin IX generation exists in B. thetaiotaomicron. Heme metabolism in congeners of B. thetaiotaomicron has previously been associated with the 6-gene hmu operon, based on genetic studies. A bioinformatics survey demonstrated that the intact operon is widespread in but confined to members of the Bacteroidetes phylum and ubiquitous in healthy human GI tract flora. Anaerobic heme metabolism by commensal Bacteroidetes via hmu is likely a major contributor to human host metabolism of the heme from dietary red meat and a driver for the selective growth of these species in the GI tract consortium. IMPORTANCE Research on bacterial iron metabolism has historically focused on the host-pathogen relationship, where the host suppresses pathogen growth by cutting off access to iron. Less is known about how host iron is shared with bacterial species that live commensally in the anaerobic human GI tract, typified by members of phylum Bacteroidetes. While many facultative pathogens avidly produce and consume heme iron, most GI tract anaerobes are heme auxotrophs whose metabolic preferences we aimed to describe. Understanding iron metabolism by model microbiome species like Bacteroides thetaiotaomicron is essential for modeling the ecology of the GI tract, which serves the long-term biomedical goals of manipulating the microbiome to facilitate host metabolism of iron and remediate dysbiosis and associated pathologies (e.g., inflammation and cancer).
Collapse
Affiliation(s)
- Margaux M. Meslé
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, USA
| | - Chase R. Gray
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, USA
| | - Mensur Dlakić
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Jennifer L. DuBois
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
28
|
Rocha MFG, Diógenes EM, Carvalho VL, Marmontel M, da Costa MO, da Silva VMF, de Souza Amaral R, Gravena W, do Carmo NAS, Marigo J, Ocadaque CJ, Freitas AS, Pinheiro RM, de Lima-Neto RG, de Aguiar Cordeiro R, de Aquino Pereira-Neto W, de Melo Guedes GM, Sidrim JJC, de Souza Collares Maia Castelo-Branco D. Virulence factors of Gram-negative bacteria from free-ranging Amazon river dolphins (Inia geoffrensis). Antonie Van Leeuwenhoek 2023; 116:447-462. [PMID: 36841923 DOI: 10.1007/s10482-023-01812-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 01/19/2023] [Indexed: 02/27/2023]
Abstract
Freshwater cetaceans play a significant role as sentinel animals, providing important data on animal species and aquatic ecosystem health. They also may serve as potential reservoirs of emerging pathogens and host virulence genes in their microbiota. In this study, we evaluated virulence factors produced by Gram-negative bacteria recovered from individuals belonging to two populations of free-ranging Amazon river dolphins (Inia geoffrensis). A total of 132 isolates recovered from the oral cavity, blowhole, genital opening and rectum of 21 river dolphins, 13 from Negro River and 8 from Tapajós River, Brazil, were evaluated for the production of virulence factors, such as biofilms and exoproducts (proteases, hemolysins and siderophores), in planktonic and biofilm forms. In planktonic form, 81.1% (107/132) of the tested bacteria of free-ranging Amazon river dolphins were able to produce virulence factors, with 44/132 (33.4%), 65/132 (49,2%) and 54/132 (40,9%) positive for protease, hemolysin and siderophore production, respectively. Overall, 57/132 (43.2%) of the isolates produced biofilms and, under this form of growth, 66/132 (50%), 88/132 (66.7%) and 80/132 (60.6%) of the isolates were positive for protease, hemolysin and siderophore production. In general, the isolates showed a higher release of exoproducts in biofilm than in planktonic form (P < 0.001). The present findings show that Amazon river dolphins harbor potentially pathogenic bacteria in their microbiota, highlighting the importance of monitoring the micro-organisms from wild animals, as they may emerge as pathogens for humans and other animals.
Collapse
Affiliation(s)
- Marcos Fábio Gadelha Rocha
- Postgraduate Program in Veterinary Sciences, School of Veterinary, State University of Ceará, Fortaleza, Ceará, Brazil.,Laboratory of Emerging and Reemerging Pathogens, Postgraduate Program in Medical Microbiology, Federal University of Ceará, Rua Coronel Nunes de Melo, 1315. Fortaleza, CEP: 60.430-275, FortalezaCeará, Brazil
| | - Expedito Maia Diógenes
- Group of Applied Medical Microbiology, Postgraduate Program in Medical Microbiology, Federal University of Ceará, Rua Coronel Nunes de Melo, 1315. Fortaleza, CEP: 60.430-275, FortalezaCeará, Brazil
| | - Vitor Luz Carvalho
- Associação de Pesquisa E Preservação de Ecossistemas Aquáticos (AQUASIS), Av. José Alencar, 150. Praia de Iparana, CEP. 61.627-210, Caucaia, Ceará, Brasil.
| | - Miriam Marmontel
- Mamirauá Sustainable Development Institute, Tefé, Amazonas, Brazil
| | | | - Vera M F da Silva
- National Institute of Amazon Research-Inpa/Aquatic Mammals Laboratory, Manaus, Amazon, Brazil
| | - Rodrigo de Souza Amaral
- Federal Institute of Education, Science and Technology of the Amazonas - IFAM, Amazonas, Brazil
| | - Waleska Gravena
- Federal University of Amazonas-UFAM, Campus Coari, Amazonas, Brazil
| | - Nívia A S do Carmo
- Federal University of Pará-UFPA, Belém, Pará, Brazil.,Brazilian Agricultural Research Corporation Eastern Amazon-EMBRAPA, Belém, Pará, Brazil
| | - Juliana Marigo
- Laboratory of Comparative Pathology of Wild Animals, School of Veterinary Medicine and Animal Science, University of São Paulo (LAPCOM, FMVZ-USP), São Paulo, Brazil
| | - Crister José Ocadaque
- Group of Applied Medical Microbiology, Postgraduate Program in Medical Microbiology, Federal University of Ceará, Rua Coronel Nunes de Melo, 1315. Fortaleza, CEP: 60.430-275, FortalezaCeará, Brazil
| | - Alyne Soares Freitas
- Group of Applied Medical Microbiology, Postgraduate Program in Medical Microbiology, Federal University of Ceará, Rua Coronel Nunes de Melo, 1315. Fortaleza, CEP: 60.430-275, FortalezaCeará, Brazil
| | - Rodrigo Machado Pinheiro
- Group of Applied Medical Microbiology, Postgraduate Program in Medical Microbiology, Federal University of Ceará, Rua Coronel Nunes de Melo, 1315. Fortaleza, CEP: 60.430-275, FortalezaCeará, Brazil
| | | | - Rossana de Aguiar Cordeiro
- Laboratory of Emerging and Reemerging Pathogens, Postgraduate Program in Medical Microbiology, Federal University of Ceará, Rua Coronel Nunes de Melo, 1315. Fortaleza, CEP: 60.430-275, FortalezaCeará, Brazil
| | - Waldemiro de Aquino Pereira-Neto
- Laboratory of Emerging and Reemerging Pathogens, Postgraduate Program in Medical Microbiology, Federal University of Ceará, Rua Coronel Nunes de Melo, 1315. Fortaleza, CEP: 60.430-275, FortalezaCeará, Brazil
| | - Glaucia Morgana de Melo Guedes
- Laboratory of Emerging and Reemerging Pathogens, Postgraduate Program in Medical Microbiology, Federal University of Ceará, Rua Coronel Nunes de Melo, 1315. Fortaleza, CEP: 60.430-275, FortalezaCeará, Brazil. .,Group of Applied Medical Microbiology, Postgraduate Program in Medical Microbiology, Federal University of Ceará, Rua Coronel Nunes de Melo, 1315. Fortaleza, CEP: 60.430-275, FortalezaCeará, Brazil.
| | - José Júlio Costa Sidrim
- Laboratory of Emerging and Reemerging Pathogens, Postgraduate Program in Medical Microbiology, Federal University of Ceará, Rua Coronel Nunes de Melo, 1315. Fortaleza, CEP: 60.430-275, FortalezaCeará, Brazil
| | - Débora de Souza Collares Maia Castelo-Branco
- Laboratory of Emerging and Reemerging Pathogens, Postgraduate Program in Medical Microbiology, Federal University of Ceará, Rua Coronel Nunes de Melo, 1315. Fortaleza, CEP: 60.430-275, FortalezaCeará, Brazil.,Group of Applied Medical Microbiology, Postgraduate Program in Medical Microbiology, Federal University of Ceará, Rua Coronel Nunes de Melo, 1315. Fortaleza, CEP: 60.430-275, FortalezaCeará, Brazil
| |
Collapse
|
29
|
Wilkinson IVL, Bottlinger M, El Harraoui Y, Sieber SA. Profiling the Heme-Binding Proteomes of Bacteria Using Chemical Proteomics. Angew Chem Int Ed Engl 2023; 62:e202212111. [PMID: 36495310 DOI: 10.1002/anie.202212111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Heme is a cofactor with myriad roles and essential to almost all living organisms. Beyond classical gas transport and catalytic functions, heme is increasingly appreciated as a tightly controlled signalling molecule regulating protein expression. However, heme acquisition, biosynthesis and regulation is poorly understood beyond a few model organisms, and the heme-binding proteome has not been fully characterised in bacteria. Yet as heme homeostasis is critical for bacterial survival, heme-binding proteins are promising drug targets. Herein we report a chemical proteomics method for global profiling of heme-binding proteins in live cells for the first time. Employing a panel of heme-based clickable and photoaffinity probes enabled the profiling of 32-54 % of the known heme-binding proteomes in Gram-positive and Gram-negative bacteria. This simple-to-implement profiling strategy could be interchangeably applied to different cell types and systems and fuel future research into heme biology.
Collapse
Affiliation(s)
- Isabel V L Wilkinson
- Centre for Functional Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Straße 8, 85748, Garching, Germany
| | - Max Bottlinger
- Centre for Functional Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Straße 8, 85748, Garching, Germany
| | - Yassmine El Harraoui
- Centre for Functional Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Straße 8, 85748, Garching, Germany
| | - Stephan A Sieber
- Centre for Functional Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Straße 8, 85748, Garching, Germany
| |
Collapse
|
30
|
Abuga KM, Nairz M, MacLennan CA, Atkinson SH. Severe anaemia, iron deficiency, and susceptibility to invasive bacterial infections. Wellcome Open Res 2023; 8:48. [PMID: 37600584 PMCID: PMC10439361 DOI: 10.12688/wellcomeopenres.18829.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2023] [Indexed: 08/22/2023] Open
Abstract
Severe anaemia and invasive bacterial infections remain important causes of hospitalization and death among young African children. The emergence and spread of antimicrobial resistance demand better understanding of bacteraemia risk factors to inform prevention strategies. Epidemiological studies have reported an association between severe anaemia and bacteraemia. In this review, we explore evidence that severe anaemia is associated with increased risk of invasive bacterial infections in young children. We describe mechanisms of iron dysregulation in severe anaemia that might contribute to increased risk and pathogenesis of invasive bacteria, recent advances in knowledge of how iron deficiency and severe anaemia impair immune responses to bacterial infections and vaccines, and the gaps in our understanding of mechanisms underlying severe anaemia, iron deficiency, and the risk of invasive bacterial infections.
Collapse
Affiliation(s)
- Kelvin M. Abuga
- Kenya Medical Research Institute (KEMRI) Centre for Geographical Medicine Research-Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, 80108, Kenya
- Open University, KEMRI-Wellcome Trust Research Programme – Accredited Research Centre, Kilifi, 80108, Kenya
| | - Manfred Nairz
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, 6020, Austria
| | - Calman A. MacLennan
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ, UK
| | - Sarah H. Atkinson
- Kenya Medical Research Institute (KEMRI) Centre for Geographical Medicine Research-Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, 80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7LG, UK
- Department of Paediatrics, University of Oxford, Oxford, OX3 9DU, UK
| |
Collapse
|
31
|
Dong J, Sun C, Tian Y, Zhang H, Liu Z, Gao F, Ye X. Genomic organization and gene evolution of two warm temperature acclimation proteins (Wap65s) of Micropterus salmoides and their responses to temperature and bacterial/viral infections. Int J Biol Macromol 2023; 227:340-353. [PMID: 36529221 DOI: 10.1016/j.ijbiomac.2022.12.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/23/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
Warm temperature acclimation-related 65-kDa proteins (Wap65s) are fish plasma acute-phase glycoproteins homologous to hemopexin with high affinity and clearance for heme. The study characterized Mswap65-1 and Mswap65-2 genes in Micropterus salmoides. Structural analysis showed MsWap65s contained conserved heme-binding sites. MsWap65-1 had a chloride-binding site similar to hemopexin, while MsWap65-2 had an additional calcium-binding site. Phylogenetic and Ka/Ks analysis showed that fish Wap65s were evolutionarily conserved and underwent strong purifying selection. Functional divergence analysis indicated that fish Wap65-2 retained the putative function of ancestral Wap65, while Wap65-1 underwent neofunctional differentiation. QPCR showed Mswap65s were predominantly expressed in liver, but prolonged hyperthermy inhibited Mswap65-2 expression. Mswap65-2 expression was up-regulated in liver and spleen after Nocardia seriolae infection, while Mswap65-1 was down-regulated. MsWap65-2 may be associated with pathogenesis and play potential role in pathogen resistance. LMBV infection resulted in both significant downregulation of Mswap65s were both significantly down-regulated, with differences observed between sexes. We speculated the immune system might suppress expression after viral infection. Exogenous rMsWap65s were prepared, and injection of rMsWap65s alleviated phenylhydrazine-induced hemolysis and inhibited increases in heme, complement C3 and inflammatory symptoms. Our results contribute to an advanced understanding of the functions and mechanisms of MsWap65s in stress resistance.
Collapse
Affiliation(s)
- Junjian Dong
- Key Laboratory of Tropical and Subtropical Fisheries Resource Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Chengfei Sun
- Key Laboratory of Tropical and Subtropical Fisheries Resource Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Yuanyuan Tian
- Key Laboratory of Tropical and Subtropical Fisheries Resource Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Hetong Zhang
- Key Laboratory of Tropical and Subtropical Fisheries Resource Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Zhigang Liu
- Key Laboratory of Tropical and Subtropical Fisheries Resource Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Fengying Gao
- Key Laboratory of Tropical and Subtropical Fisheries Resource Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China.
| | - Xing Ye
- Key Laboratory of Tropical and Subtropical Fisheries Resource Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
32
|
Gouda AM, Soltan MA, Abd-Elghany K, Sileem AE, Elnahas HM, Ateya MAM, Elbatreek MH, Darwish KM, Bogari HA, Lashkar MO, Aldurdunji MM, Elhady SS, Ahmad TA, Said AM. Integration of immunoinformatics and cheminformatics to design and evaluate a multitope vaccine against Klebsiella pneumoniae and Pseudomonas aeruginosa coinfection. Front Mol Biosci 2023; 10:1123411. [PMID: 36911530 PMCID: PMC9999731 DOI: 10.3389/fmolb.2023.1123411] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/26/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction: Klebsiella pneumoniae (K. pneumoniae) and Pseudomonas aeruginosa (P. aeruginosa) are the most common Gram-negative bacteria associated with pneumonia and coinfecting the same patient. Despite their high virulence, there is no effective vaccine against them. Methods: In the current study, the screening of several proteins from both pathogens highlighted FepA and OmpK35 for K. pneumonia in addition to HasR and OprF from P. aeruginosa as promising candidates for epitope mapping. Those four proteins were linked to form a multitope vaccine, that was formulated with a suitable adjuvant, and PADRE peptides to finalize the multitope vaccine construct. The final vaccine's physicochemical features, antigenicity, toxicity, allergenicity, and solubility were evaluated for use in humans. Results: The output of the computational analysis revealed that the designed multitope construct has passed these assessments with satisfactory scores where, as the last stage, we performed a molecular docking study between the potential vaccine construct and K. pneumonia associated immune receptors, TLR4 and TLR2, showing affinitive to both targets with preferentiality for the TLR4 receptor protein. Validation of the docking studies has proceeded through molecular dynamics simulation, which estimated a strong binding and supported the nomination of the designed vaccine as a putative solution for K. pneumoniae and P. aeruginosa coinfection. Here, we describe the approach for the design and assessment of our potential vaccine.
Collapse
Affiliation(s)
- Ahmed M Gouda
- Department of Pharmacy Practice, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed A Soltan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University-Kantara Branch, Ismailia, Egypt
| | - Khalid Abd-Elghany
- Department of Microbiology-Microbial Biotechnology, Egyptian Drug Authority, Giza, Egypt
| | - Ashraf E Sileem
- Department of Chest Diseases, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Hanan M Elnahas
- Department of Pharmaceutical and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | | | - Mahmoud H Elbatreek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Khaled M Darwish
- Department of Medicinal Chemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Hanin A Bogari
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Manar O Lashkar
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed M Aldurdunji
- Department of Clinical Pharmacy, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sameh S Elhady
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia.,Center for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tarek A Ahmad
- Library Sector, Bibliotheca Alexandrina, Alexandria, Egypt
| | - Ahmed Mohamed Said
- Department of Chest Diseases, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
33
|
Iron acquisition strategies in pseudomonads: mechanisms, ecology, and evolution. Biometals 2022:10.1007/s10534-022-00480-8. [PMID: 36508064 PMCID: PMC10393863 DOI: 10.1007/s10534-022-00480-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
AbstractIron is important for bacterial growth and survival, as it is a common co-factor in essential enzymes. Although iron is very abundant in the earth crust, its bioavailability is low in most habitats because ferric iron is largely insoluble under aerobic conditions and at neutral pH. Consequently, bacteria have evolved a plethora of mechanisms to solubilize and acquire iron from environmental and host stocks. In this review, I focus on Pseudomonas spp. and first present the main iron uptake mechanisms of this taxa, which involve the direct uptake of ferrous iron via importers, the production of iron-chelating siderophores, the exploitation of siderophores produced by other microbial species, and the use of iron-chelating compounds produced by plants and animals. In the second part of this review, I elaborate on how these mechanisms affect interactions between bacteria in microbial communities, and between bacteria and their hosts. This is important because Pseudomonas spp. live in diverse communities and certain iron-uptake strategies might have evolved not only to acquire this essential nutrient, but also to gain relative advantages over competitors in the race for iron. Thus, an integrative understanding of the mechanisms of iron acquisition and the eco-evolutionary dynamics they drive at the community level might prove most useful to understand why Pseudomonas spp., in particular, and many other bacterial species, in general, have evolved such diverse iron uptake repertoires.
Collapse
|
34
|
Mayneris-Perxachs J, Moreno-Navarrete JM, Fernández-Real JM. The role of iron in host-microbiota crosstalk and its effects on systemic glucose metabolism. Nat Rev Endocrinol 2022; 18:683-698. [PMID: 35986176 DOI: 10.1038/s41574-022-00721-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/01/2022] [Indexed: 11/09/2022]
Abstract
Iron is critical for the appearance and maintenance of life on Earth. Almost all organisms compete or cooperate for iron acquisition, demonstrating the importance of this essential element for the biological and physiological processes that are key for the preservation of metabolic homeostasis. In humans and other mammals, the bidirectional interactions between the bacterial component of the gut microbiota and the host for iron acquisition shape both host and microbiota metabolism. Bacterial functions influence host iron absorption, whereas the intake of iron, iron deficiency and iron excess in the host affect bacterial biodiversity, taxonomy and function, resulting in changes in bacterial virulence. These consequences of the host-microbial crosstalk affect systemic levels of iron, its storage in different tissues and host glucose metabolism. At the interface between the host and the microbiota, alterations in the host innate immune system and in circulating soluble factors that regulate iron (that is, hepcidin, lipocalin 2 and lactoferrin) are associated with metabolic disease. In fact, patients with obesity-associated metabolic dysfunction and insulin resistance exhibit dysregulation in iron homeostasis and alterations in their gut microbiota profile. From an evolutionary point of view, the pursuit of two important nutrients - glucose and iron - has probably driven human evolution towards the most efficient pathways and genes for human survival and health.
Collapse
Affiliation(s)
- Jordi Mayneris-Perxachs
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain.
- Department of Medicine, Universitat de Girona, Girona, Spain.
| |
Collapse
|
35
|
Murdoch CC, Skaar EP. Nutritional immunity: the battle for nutrient metals at the host-pathogen interface. Nat Rev Microbiol 2022; 20:657-670. [PMID: 35641670 PMCID: PMC9153222 DOI: 10.1038/s41579-022-00745-6] [Citation(s) in RCA: 255] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2022] [Indexed: 12/21/2022]
Abstract
Trace metals are essential micronutrients required for survival across all kingdoms of life. From bacteria to animals, metals have critical roles as both structural and catalytic cofactors for an estimated third of the proteome, representing a major contributor to the maintenance of cellular homeostasis. The reactivity of metal ions engenders them with the ability to promote enzyme catalysis and stabilize reaction intermediates. However, these properties render metals toxic at high concentrations and, therefore, metal levels must be tightly regulated. Having evolved in close association with bacteria, vertebrate hosts have developed numerous strategies of metal limitation and intoxication that prevent bacterial proliferation, a process termed nutritional immunity. In turn, bacterial pathogens have evolved adaptive mechanisms to survive in conditions of metal depletion or excess. In this Review, we discuss mechanisms by which nutrient metals shape the interactions between bacterial pathogens and animal hosts. We explore the cell-specific and tissue-specific roles of distinct trace metals in shaping bacterial infections, as well as implications for future research and new therapeutic development.
Collapse
Affiliation(s)
- Caitlin C Murdoch
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Eric P Skaar
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
36
|
Al-Madboly LA. A Novel Triple Combination To Combat Serious Infections with Carbapenem-Resistant Acinetobacter baumannii in a Mouse Pneumonia Model. Microbiol Spectr 2022; 10:e0271021. [PMID: 35975993 PMCID: PMC9603289 DOI: 10.1128/spectrum.02710-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/17/2022] [Indexed: 01/04/2023] Open
Abstract
The ongoing crisis of antimicrobial resistance demands novel combinations between antimicrobials and nonantimicrobials to manage infections caused by highly resistant pathogens. This study aimed to evaluate the effect of combining sodium ascorbate and/or apo-transferrin with imipenem, forming double and triple combinations, against 20 multiple-carbapenemase-producing Acinetobacter baumannii strains using the checkerboard test, time-kill assay, and disc diffusion test. The results of the checkerboard assay revealed that all double combinations showed indifference, while only triple combination recorded a synergistic effect (fractional inhibitory concentration index [FICI] < 0.8) in 95% the test isolates. Moreover, the MIC of imipenem (MICimp) was strongly reduced (up to 128-fold reduction) after treatment with the triple combination against highly resistant isolates and reached the susceptible range. The time-kill assay revealed that the triple combination led to a 4-log10 reduction in the CFU at 8 h compared with the initial bacterial count, and no viable count was recorded at 10 h. The mouse pneumonia model showed restoration of lung function and structure, with mild to moderate residual inflammation and moderately congested vessels observed 8 h following administration of the triple rescue therapy. Additionally, normal lungs with normal patent alveoli were detected 72 h following treatment. Accordingly, sodium ascorbate and apo-transferrin are promising adjunct biological agents with the potential to restore the effectiveness of critically essential antibiotics like imipenem, commonly used for the treatment of A. baumannii infections. IMPORTANCE Combination therapy provides a perspective to threat multidrug-resistant (MDR) strains. The present study sheds light on a novel and effective triple combination against carbapenem-resistant A. baumannii. Our in vitro results showed that combining imipenem with apo-transferrin and sodium ascorbate yielded synergism in 95% of test isolates, and this was associated with a marked reduction in imipenem MIC, shifting it below the breakpoint. Furthermore, a bactericidal effect was recorded, with no viable count detected at 10 h. An in vivo murine model of pneumonia was induced to mimic human disease. The triple combination therapy restored lung function and structure, with mild to moderate residual inflammation and moderately congested vessels observed 8 h following the initiation of therapy. Therefore, our findings suggest novel insights about a promising new combination therapy against highly resistant carbapenemase-producing A. baumannii to restore the effectiveness of imipenem.
Collapse
Affiliation(s)
- Lamiaa A. Al-Madboly
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
37
|
Kannon M, Nebane NM, Ruiz P, McKellip S, Vinson PN, Mitra A. A Novel Approach To Identify Inhibitors of Iron Acquisition Systems of Pseudomonas aeruginosa. Microbiol Spectr 2022; 10:e0243722. [PMID: 36098531 PMCID: PMC9604216 DOI: 10.1128/spectrum.02437-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/26/2022] [Indexed: 01/04/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that has been declared by the World Health Organization as a "priority 1 critical pathogen" needing immediate new strategies for chemotherapy. During infection, P. aeruginosa uses redundant mechanisms to acquire ferric, heme (Hm), or ferrous iron from the host to survive and colonize. Significant efforts have been undertaken to develop siderophore blockers to inhibit ferric iron acquisition by P. aeruginosa, but there is a lack of inhibitors that can block Hm or ferrous iron acquisition by P. aeruginosa. We developed and employed a targeted high-throughput screen (HTS) and identified a molecule(s) that can specifically inhibit the Hm and ferrous iron acquisition systems of P. aeruginosa. Our targeted approach relies on screening a small-molecule library against P. aeruginosa under three growth conditions, where the only variable was the iron source (ferric, Hm, or ferrous iron). Each condition served as a counterscreen for the other, and we identified molecules that inhibit the growth of P. aeruginosa in the presence of only Hm or ferrous iron. Our data indicate that econazole, bithionate, and raloxifene inhibit the growth of P. aeruginosa in the presence of Hm and that oxyquinoline inhibits the growth of P. aeruginosa in the presence of ferrous iron. These iron-specific inhibitors do not interfere with the activity of meropenem, a commercial antipseudomonal, and can also increase meropenem activity. In conclusion, we present a proof of concept of a successful targeted conditional screening method by which we can identify specific iron acquisition inhibitors. This approach is highly adaptable and can easily be extended to any other pathogen. IMPORTANCE Since acquiring iron is paramount to P. aeruginosa's survival and colonization in the human host, developing novel strategies to block the access of P. aeruginosa to host iron will allow us to starve it of an essential nutrient. P. aeruginosa uses siderophore, heme, or ferrous iron uptake systems to acquire iron in the human host. We have developed a novel approach through which we can directly identify molecules that can prevent P. aeruginosa from utilizing heme or ferrous iron. This approach overcomes the need for the in silico design of molecules and identifies structurally diverse biologically active inhibitor molecules. This screening approach is adaptable and can be extended to any pathogen. Since Gram-negative pathogens share many similarities in iron acquisition at both the mechanistic and molecular levels, our screening approach presents a significant opportunity to develop novel broad-spectrum iron acquisition inhibitors of Gram-negative pathogens.
Collapse
Affiliation(s)
- Mamie Kannon
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - N. Miranda Nebane
- High Throughput Screening Center, Southern Research, Birmingham, Alabama, USA
| | - Pedro Ruiz
- High Throughput Screening Center, Southern Research, Birmingham, Alabama, USA
| | - Sara McKellip
- High Throughput Screening Center, Southern Research, Birmingham, Alabama, USA
| | - Paige N. Vinson
- High Throughput Screening Center, Southern Research, Birmingham, Alabama, USA
| | - Avishek Mitra
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
38
|
Mukti IJ, Sardari RRR, Kristjansdottir T, Hreggvidsson GO, Karlsson EN. Medium development and production of carotenoids and exopolysaccharides by the extremophile Rhodothermus marinus DSM16675 in glucose-based defined media. Microb Cell Fact 2022; 21:220. [PMID: 36274123 PMCID: PMC9590192 DOI: 10.1186/s12934-022-01946-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The marine thermophilic bacterium Rhodothermus marinus can degrade many polysaccharides which makes it interesting as a future cell factory. Progress using this bacterium has, however, been hampered by limited knowledge on media and conditions for biomass production, often resulting in low cell yields and low productivity, highlighting the need to develop conditions that allow studies of the microbe on molecular level. This study presents development of defined conditions that support growth, combined with evaluation of production of carotenoids and exopolysaccharides (EPSs) by R. marinus strain DSM 16675. RESULTS Two defined media were initially prepared: one including a low addition of yeast extract (modified Wolfe's medium) and one based on specific components (defined medium base, DMB) to which two amino acids (N and Q), were added. Cultivation trials of R. marinus DSM 16675 in shake flasks, resulted in maximum cell densities (OD620 nm) of 2.36 ± 0.057, cell dry weight (CDW) 1.2 ± 0.14 mg/L, total carotenoids 0.59 × 10-3 mg/L, and EPSs 1.72 ± 0.03 mg/L using 2 g/L glucose in DMB. In Wolfe's medium (supplemented by 0.05 g/L yeast extract and 2.5 g/L glucose), maximum OD620 nm was 2.07 ± 0.05, CDW 1.05 ± 0.07 mg/L, total carotenoids 0.39 × 10-3 mg/L, and EPSs 1.74 ± 0.2 mg/L. Growth trials at 5 g/L glucose in these media either failed or resulted in incomplete substrate utilization. To improve reproducibility and increase substrate utilization, a screening of macroelements (e.g. phosphate) in DMB, was combined with use of trace elements and vitamins of the modified Wolfe's medium. The resulting defined minimal R. marinus medium, (DRM), allowed reproducible cultivations to a final OD620nm of 6.6 ± 0.05, CDW 2.85 ± 0.07 mg/L, a maximum specific growth rate (µmax) of 0.26 h-1, total carotenoids 0.77 × 10-3 mg/L and EPSs 3.4 ± 0.17 mg/L in cultivations supplemented with up to 5 g/L glucose. CONCLUSION A minimal defined medium (DRM) was designed that resulted in reproducible growth and an almost doubled formation of both total carotenoids and EPSs. Such defined conditions, are necessary for systematic studies of metabolic pathways, to determine the specific requirements for growth and fully characterize metabolite production.
Collapse
Affiliation(s)
- Israt Jahan Mukti
- Division of Biotechnology, Department of Chemistry, Lund University, Naturvetarvägen 14, 22100, Lund, Sweden
| | - Roya R R Sardari
- Division of Biotechnology, Department of Chemistry, Lund University, Naturvetarvägen 14, 22100, Lund, Sweden.
| | - Thordis Kristjansdottir
- Matis Ohf, Vinlandsleid 12, 113, Reykjavik, Iceland.,Department of Biology, School of Engineering and Natural Sciences, University of Iceland, Sturlugata 7, 102, Reykjavik, Iceland
| | - Gudmundur O Hreggvidsson
- Matis Ohf, Vinlandsleid 12, 113, Reykjavik, Iceland.,Department of Biology, School of Engineering and Natural Sciences, University of Iceland, Sturlugata 7, 102, Reykjavik, Iceland
| | - Eva Nordberg Karlsson
- Division of Biotechnology, Department of Chemistry, Lund University, Naturvetarvägen 14, 22100, Lund, Sweden
| |
Collapse
|
39
|
Goodman AZ, Papudeshi B, Doane MP, Mora M, Kerr E, Torres M, Nero Moffatt J, Lima L, Nosal AP, Dinsdale E. Epidermal Microbiomes of Leopard Sharks ( Triakis semifasciata) Are Consistent across Captive and Wild Environments. Microorganisms 2022; 10:microorganisms10102081. [PMID: 36296361 PMCID: PMC9610875 DOI: 10.3390/microorganisms10102081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 01/24/2023] Open
Abstract
Characterizations of shark-microbe systems in wild environments have outlined patterns of species-specific microbiomes; however, whether captivity affects these trends has yet to be determined. We used high-throughput shotgun sequencing to assess the epidermal microbiome belonging to leopard sharks (Triakis semifasciata) in captive (Birch Aquarium, La Jolla California born and held permanently in captivity), semi-captive (held in captivity for <1 year in duration and scheduled for release; Scripps Institute of Oceanography, San Diego, CA, USA) and wild environments (Moss Landing and La Jolla, CA, USA). Here, we report captive environments do not drive epidermal microbiome compositions of T. semifasciata to significantly diverge from wild counterparts as life-long captive sharks maintain a species-specific epidermal microbiome resembling those associated with semi-captive and wild populations. Major taxonomic composition shifts observed were inverse changes of top taxonomic contributors across captive duration, specifically an increase of Pseudoalteromonadaceae and consequent decrease of Pseudomonadaceae relative abundance as T. semifasciata increased duration in captive conditions. Moreover, we show captivity did not lead to significant losses in microbial α-diversity of shark epidermal communities. Finally, we present a novel association between T. semifasciata and the Muricauda genus as Metagenomes associated genomes revealed a consistent relationship across captive, semi-captive, and wild populations. Since changes in microbial communities is often associated with poor health outcomes, our report illustrates that epidermally associated microbes belonging to T. semifasciata are not suffering detrimental impacts from long or short-term captivity. Therefore, conservation programs which house sharks in aquariums are providing a healthy environment for the organisms on display. Our findings also expand on current understanding of shark epidermal microbiomes, explore the effects of ecologically different scenarios on benthic shark microbe associations, and highlight novel associations that are consistent across captive gradients.
Collapse
Affiliation(s)
- Asha Z. Goodman
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
- Correspondence: (A.Z.G.); (E.D.)
| | - Bhavya Papudeshi
- College of Science and Engineering, Flinders University, Bedford Park, SA 3929, Australia
| | - Michael P. Doane
- College of Science and Engineering, Flinders University, Bedford Park, SA 3929, Australia
| | - Maria Mora
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Emma Kerr
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Melissa Torres
- Scripps Institution of Oceanography, Universtity of California, San Diego, CA 92093, USA
| | - Jennifer Nero Moffatt
- Scripps Institution of Oceanography, Universtity of California, San Diego, CA 92093, USA
| | - Lais Lima
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Andrew P. Nosal
- Department of Biology, Point Loma Nazarene University, San Diego, CA 92106, USA
| | - Elizabeth Dinsdale
- College of Science and Engineering, Flinders University, Bedford Park, SA 3929, Australia
- Correspondence: (A.Z.G.); (E.D.)
| |
Collapse
|
40
|
Sestok AE, O'Sullivan SM, Smith AT. A general protocol for the expression and purification of the intact transmembrane transporter FeoB. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183973. [PMID: 35636558 PMCID: PMC9203943 DOI: 10.1016/j.bbamem.2022.183973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 04/27/2023]
Abstract
Ferrous iron (Fe2+) transport is an essential process that supports the growth, intracellular survival, and virulence of several drug-resistant pathogens, and the ferrous iron transport (Feo) system is the most important and widespread protein complex that mediates Fe2+ transport in these organisms. The Feo system canonically comprises three proteins (FeoA/B/C). FeoA and FeoC are both small, accessory proteins localized to the cytoplasm, and their roles in the Fe2+ transport process have been of great debate. FeoB is the only wholly-conserved component of the Feo system and serves as the inner membrane-embedded Fe2+ transporter with a soluble G-protein-like N-terminal domain. In vivo studies have underscored the importance of Feo during infection, emphasizing the need to better understand Feo-mediated Fe2+ uptake, although a paucity of research exists on intact FeoB. To surmount this problem, we designed an overproduction and purification system that can be applied generally to a suite of intact FeoBs from several organisms. Importantly, we noted that FeoB is extremely sensitive to excess salt while in the membrane of a recombinant host, and we designed a workflow to circumvent this issue. We also demonstrated effective protein extraction from the lipid bilayer through small-scale solubilization studies. We then applied this approach to the large-scale purifications of Escherichia coli and Pseudomonas aeruginosa FeoBs to high purity and homogeneity. Lastly, we show that our protocol can be generally applied to various FeoB proteins. Thus, this workflow allows for isolation of suitable quantities of FeoB for future biochemical and biophysical characterization.
Collapse
Affiliation(s)
- Alex E Sestok
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD, 21250, USA
| | - Sean M O'Sullivan
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD, 21250, USA
| | - Aaron T Smith
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD, 21250, USA.
| |
Collapse
|
41
|
Sankari S, Babu VM, Bian K, Alhhazmi A, Andorfer MC, Avalos DM, Smith TA, Yoon K, Drennan CL, Yaffe MB, Lourido S, Walker GC. A haem-sequestering plant peptide promotes iron uptake in symbiotic bacteria. Nat Microbiol 2022; 7:1453-1465. [PMID: 35953657 PMCID: PMC9420810 DOI: 10.1038/s41564-022-01192-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/29/2022] [Indexed: 11/09/2022]
Abstract
Symbiotic partnerships with rhizobial bacteria enable legumes to grow without nitrogen fertilizer because rhizobia convert atmospheric nitrogen gas into ammonia via nitrogenase. After Sinorhizobium meliloti penetrate the root nodules that they have elicited in Medicago truncatula, the plant produces a family of about 700 nodule cysteine-rich (NCR) peptides that guide the differentiation of endocytosed bacteria into nitrogen-fixing bacteroids. The sequences of the NCR peptides are related to the defensin class of antimicrobial peptides, but have been adapted to play symbiotic roles. Using a variety of spectroscopic, biophysical and biochemical techniques, we show here that the most extensively characterized NCR peptide, 24 amino acid NCR247, binds haem with nanomolar affinity. Bound haem molecules and their iron are initially made biologically inaccessible through the formation of hexamers (6 haem/6 NCR247) and then higher-order complexes. We present evidence that NCR247 is crucial for effective nitrogen-fixing symbiosis. We propose that by sequestering haem and its bound iron, NCR247 creates a physiological state of haem deprivation. This in turn induces an iron-starvation response in rhizobia that results in iron import, which itself is required for nitrogenase activity. Using the same methods as for L-NCR247, we show that the D-enantiomer of NCR247 can bind and sequester haem in an equivalent manner. The special abilities of NCR247 and its D-enantiomer to sequester haem suggest a broad range of potential applications related to human health.
Collapse
Affiliation(s)
- Siva Sankari
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Vignesh M.P. Babu
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Ke Bian
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Areej Alhhazmi
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Mary C. Andorfer
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Dante M. Avalos
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Harvard Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA
| | - Tyler A. Smith
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Kwan Yoon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Catherine L. Drennan
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Michael B. Yaffe
- Departments of Biology and Biological Engineering, and Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute for Technology, Cambridge, MA 02139, USA.,Divisions of Acute Care Surgery, Trauma, and Surgical Critical Care, and Surgical Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215 USA
| | - Sebastian Lourido
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Graham C. Walker
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| |
Collapse
|
42
|
Malesza IJ, Bartkowiak-Wieczorek J, Winkler-Galicki J, Nowicka A, Dzięciołowska D, Błaszczyk M, Gajniak P, Słowińska K, Niepolski L, Walkowiak J, Mądry E. The Dark Side of Iron: The Relationship between Iron, Inflammation and Gut Microbiota in Selected Diseases Associated with Iron Deficiency Anaemia—A Narrative Review. Nutrients 2022; 14:nu14173478. [PMID: 36079734 PMCID: PMC9458173 DOI: 10.3390/nu14173478] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 12/21/2022] Open
Abstract
Iron is an indispensable nutrient for life. A lack of it leads to iron deficiency anaemia (IDA), which currently affects about 1.2 billion people worldwide. The primary means of IDA treatment is oral or parenteral iron supplementation. This can be burdened with numerous side effects such as oxidative stress, systemic and local-intestinal inflammation, dysbiosis, carcinogenic processes and gastrointestinal adverse events. Therefore, this review aimed to provide insight into the physiological mechanisms of iron management and investigate the state of knowledge of the relationship between iron supplementation, inflammatory status and changes in gut microbiota milieu in diseases typically complicated with IDA and considered as having an inflammatory background such as in inflammatory bowel disease, colorectal cancer or obesity. Understanding the precise mechanisms critical to iron metabolism and the awareness of serious adverse effects associated with iron supplementation may lead to the provision of better IDA treatment. Well-planned research, specific to each patient category and disease, is needed to find measures and methods to optimise iron treatment and reduce adverse effects.
Collapse
Affiliation(s)
- Ida J. Malesza
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | | | - Jakub Winkler-Galicki
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Aleksandra Nowicka
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | | | - Marta Błaszczyk
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Paulina Gajniak
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Karolina Słowińska
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Leszek Niepolski
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Edyta Mądry
- Department of Physiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
- Correspondence:
| |
Collapse
|
43
|
Wu Z, Shao J, Zheng J, Liu B, Li Z, Shen N. A zero-sum game or an interactive frame? Iron competition between bacteria and humans in infection war. Chin Med J (Engl) 2022; 135:1917-1926. [PMID: 35830263 PMCID: PMC9746790 DOI: 10.1097/cm9.0000000000002233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT Iron is an essential trace element for both humans and bacteria. It plays a vital role in life, such as in redox reactions and electron transport. Strict regulatory mechanisms are necessary to maintain iron homeostasis because both excess and insufficient iron are harmful to life. Competition for iron is a war between humans and bacteria. To grow, reproduce, colonize, and successfully cause infection, pathogens have evolved various mechanisms for iron uptake from humans, principally Fe 3+ -siderophore and Fe 2+ -heme transport systems. Humans have many innate immune mechanisms that regulate the distribution of iron and inhibit bacterial iron uptake to help resist bacterial invasion and colonization. Meanwhile, researchers have invented detection test strips and coupled antibiotics with siderophores to create tools that take advantage of this battle for iron, to help eliminate pathogens. In this review, we summarize bacterial and human iron metabolism, competition for iron between humans and bacteria, siderophore sensors, antibiotics coupled with siderophores, and related phenomena. We also discuss how competition for iron can be used for diagnosis and treatment of infection in the future.
Collapse
Affiliation(s)
- Zhenchao Wu
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
- Center for Infectious Diseases, Peking University Third Hospital, Beijing 100191, China
| | - Jiqi Shao
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jiajia Zheng
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Beibei Liu
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Zhiyuan Li
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Ning Shen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
- Center for Infectious Diseases, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
44
|
Catlett JL, Carr S, Cashman M, Smith MD, Walter M, Sakkaff Z, Kelley C, Pierobon M, Cohen MB, Buan NR. Metabolic Synergy between Human Symbionts Bacteroides and Methanobrevibacter. Microbiol Spectr 2022; 10:e0106722. [PMID: 35536023 PMCID: PMC9241691 DOI: 10.1128/spectrum.01067-22] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022] Open
Abstract
Trophic interactions between microbes are postulated to determine whether a host microbiome is healthy or causes predisposition to disease. Two abundant taxa, the Gram-negative heterotrophic bacterium Bacteroides thetaiotaomicron and the methanogenic archaeon Methanobrevibacter smithii, are proposed to have a synergistic metabolic relationship. Both organisms play vital roles in human gut health; B. thetaiotaomicron assists the host by fermenting dietary polysaccharides, whereas M. smithii consumes end-stage fermentation products and is hypothesized to relieve feedback inhibition of upstream microbes such as B. thetaiotaomicron. To study their metabolic interactions, we defined and optimized a coculture system and used software testing techniques to analyze growth under a range of conditions representing the nutrient environment of the host. We verify that B. thetaiotaomicron fermentation products are sufficient for M. smithii growth and that accumulation of fermentation products alters secretion of metabolites by B. thetaiotaomicron to benefit M. smithii. Studies suggest that B. thetaiotaomicron metabolic efficiency is greater in the absence of fermentation products or in the presence of M. smithii. Under certain conditions, B. thetaiotaomicron and M. smithii form interspecies granules consistent with behavior observed for syntrophic partnerships between microbes in soil or sediment enrichments and anaerobic digesters. Furthermore, when vitamin B12, hematin, and hydrogen gas are abundant, coculture growth is greater than the sum of growth observed for monocultures, suggesting that both organisms benefit from a synergistic mutual metabolic relationship. IMPORTANCE The human gut functions through a complex system of interactions between the host human tissue and the microbes which inhabit it. These diverse interactions are difficult to model or examine under controlled laboratory conditions. We studied the interactions between two dominant human gut microbes, B. thetaiotaomicron and M. smithii, using a seven-component culturing approach that allows the systematic examination of the metabolic complexity of this binary microbial system. By combining high-throughput methods with machine learning techniques, we were able to investigate the interactions between two dominant genera of the gut microbiome in a wide variety of environmental conditions. Our approach can be broadly applied to studying microbial interactions and may be extended to evaluate and curate computational metabolic models. The software tools developed for this study are available as user-friendly tutorials in the Department of Energy KBase.
Collapse
Affiliation(s)
- Jennie L. Catlett
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Sean Carr
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Mikaela Cashman
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
| | - Megan D. Smith
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Mary Walter
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Zahmeeth Sakkaff
- Department of Computer Science & Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Christine Kelley
- Department of Mathematics, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Massimiliano Pierobon
- Department of Computer Science & Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Myra B. Cohen
- Department of Computer Science & Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Department of Computer Science, Iowa State University, Ames, Iowa, USA
| | - Nicole R. Buan
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
45
|
Sun Y, Wang X, Gong Q, Li J, Huang H, Xue F, Dai J, Tang F. Extraintestinal Pathogenic Escherichia coli Utilizes Surface-Located Elongation Factor G to Acquire Iron from Holo-Transferrin. Microbiol Spectr 2022; 10:e0166221. [PMID: 35477220 PMCID: PMC9045202 DOI: 10.1128/spectrum.01662-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/14/2022] [Indexed: 11/20/2022] Open
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) can cause systemic infections in both humans and animals. As an essential nutrient, iron is strictly sequestered by the host. Circumventing iron sequestration is a determinant factor for ExPEC infection. However, the ExPEC iron acquisition mechanism, particularly the mechanism of transferrin (TF) acquisition, remains unclear. This study reports that iron-saturated holo-TF can be utilized by ExPEC to promote its growth in culture medium and survival in macrophages. ExPEC specifically bound to holo-TF instead of iron-free apo-TF via the surface located elongation factor G (EFG) in both culture medium and macrophages. As a moonlighting protein, EFG specifically bound holo-TF and also released iron in TF. These two functions were performed by different domains of EFG, in which the N-terminal domains were responsible for holo-TF binding and the C-terminal domains were responsible for iron release. The functions of EFG and its domains have also been further confirmed by surface-display vectors. The surface overexpression of EFG bound significantly more holo-TF in macrophages and significantly improved bacterial intracellular survival ability. Our findings reveal a novel iron acquisition mechanism involving EFG, which suggests novel research avenues into the molecular mechanism of ExPEC resistance to nutritional immunity. IMPORTANCE Extraintestinal pathogenic Escherichia coli (ExPEC) is an important pathogen causing systemic infections in humans and animals. The competition for iron between ExPEC and the host is a determinant for ExPEC to establish a successful infection. Here, we sought to elucidate the role of transferrin (TF) in the interaction between ExPEC and the host. Our results revealed that holo-TF could be utilized by ExPEC to enhance its growth in culture medium and survival in macrophages. Furthermore, the role of elongation factor G (EFG), a novel holo-TF-binding and TF-iron release protein, was confirmed in this study. Our work provides insights into the iron acquisition mechanism of ExPEC, deepens understanding of the interaction between holo-TF and pathogens, and broadens further researches into the molecular mechanism of ExPEC pathogenicity.
Collapse
Affiliation(s)
- Yu Sun
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xuhang Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Qianwen Gong
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jin Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Haosheng Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Feng Xue
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jianjun Dai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fang Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
46
|
Abstract
We hereby present the first descriptions of human-invasive infections caused by Escherichia marmotae, a recently described species that encompasses the former “Escherichia cryptic clade V.” We describe four cases, one acute sepsis of unknown origin, one postoperative sepsis after cholecystectomy, one spondylodiscitis, and one upper urinary tract infection. Cases were identified through unsystematic queries in a single clinical lab over 6 months. Through genome sequencing of the causative strains combined with available genomes from elsewhere, we demonstrate Es. marmotae to be a likely ubiquitous species containing genotypic virulence traits associated with Escherichia pathogenicity. The invasive isolates were scattered among isolates from a range of nonhuman sources in the phylogenetic analyses, thus indicating inherent virulence in multiple lineages. Pan genome analyses indicate that Es. marmotae has a large accessory genome and is likely to obtain ecologically advantageous traits, such as genes encoding antimicrobial resistance. Reliable identification might be possible by matrix-assisted laser desorption ionization–time of flight mass spectrometry (MALDI-TOF MS), but relevant spectra are missing in commercial databases. It can be identified through 16S rRNA gene sequencing. Escherichia marmotae could represent a relatively common human pathogen, and improved diagnostics will provide a better understanding of its clinical importance. IMPORTANCEEscherichia coli is the most common pathogen found in blood cultures and urine and among the most important pathogenic species in the realm of human health. The notion that some of these isolates are not Es. coli but rather another species within the same genus may have implications for what Es. coli constitutes. We only recently have obtained methods to separate the two species, which means that possible differences in important clinical aspects, such as antimicrobial resistance rates, virulence, and phylogenetic structure, may exist. We believe that Es. marmotae as a common pathogen is new merely because we have not looked or bothered to distinguish between the thousands of invasive Escherichia passing through microbiological laboratories each day.
Collapse
|
47
|
Sestok AE, Brown JB, Obi JO, O'Sullivan SM, Garcin ED, Deredge DJ, Smith AT. A fusion of the Bacteroides fragilis ferrous iron import proteins reveals a role for FeoA in stabilizing GTP-bound FeoB. J Biol Chem 2022; 298:101808. [PMID: 35271852 PMCID: PMC8980893 DOI: 10.1016/j.jbc.2022.101808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 12/25/2022] Open
Abstract
Iron is an essential element for nearly all organisms, and under anoxic and/or reducing conditions, Fe2+ is the dominant form of iron available to bacteria. The ferrous iron transport (Feo) system is the primary prokaryotic Fe2+ import machinery, and two constituent proteins (FeoA and FeoB) are conserved across most bacterial species. However, how FeoA and FeoB function relative to one another remains enigmatic. In this work, we explored the distribution of feoAB operons encoding a fusion of FeoA tethered to the N-terminal, G-protein domain of FeoB via a connecting linker region. We hypothesized that this fusion poises FeoA to interact with FeoB to affect function. To test this hypothesis, we characterized the soluble NFeoAB fusion protein from Bacteroides fragilis, a commensal organism implicated in drug-resistant infections. Using X-ray crystallography, we determined the 1.50-Å resolution structure of BfFeoA, which adopts an SH3-like fold implicated in protein–protein interactions. Using a combination of structural modeling, small-angle X-ray scattering, and hydrogen–deuterium exchange mass spectrometry, we show that FeoA and NFeoB interact in a nucleotide-dependent manner, and we mapped the protein–protein interaction interface. Finally, using guanosine triphosphate (GTP) hydrolysis assays, we demonstrate that BfNFeoAB exhibits one of the slowest known rates of Feo-mediated GTP hydrolysis that is not potassium-stimulated. Importantly, truncation of FeoA from this fusion demonstrates that FeoA–NFeoB interactions function to stabilize the GTP-bound form of FeoB. Taken together, our work reveals a role for FeoA function in the fused FeoAB system and suggests a function for FeoA among prokaryotes.
Collapse
Affiliation(s)
- Alex E Sestok
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland, 21250 USA
| | - Janae B Brown
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland, 21250 USA
| | - Juliet O Obi
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, 21201 USA
| | - Sean M O'Sullivan
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland, 21250 USA
| | - Elsa D Garcin
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland, 21250 USA; Laboratoire d'Information Génomique et Structurale, UMR7256, Aix-Marseille Université, Campus de Luminy, 13288 Marseille, France
| | - Daniel J Deredge
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, 21201 USA
| | - Aaron T Smith
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland, 21250 USA.
| |
Collapse
|
48
|
Cheng X, Liu W, Wang Z, Yang R, Yu L, Du Q, Ge A, Liu C, Chi Z. Improved triple-module fluorescent biosensor for the rapid and ultrasensitive detection of Campylobacter jejuni in livestock and dairy. Food Control 2022. [DOI: 10.1016/j.foodcont.2022.108905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
49
|
Tony-Odigie A, Wilke L, Boutin S, Dalpke AH, Yi B. Commensal Bacteria in the Cystic Fibrosis Airway Microbiome Reduce P. aeruginosa Induced Inflammation. Front Cell Infect Microbiol 2022; 12:824101. [PMID: 35174108 PMCID: PMC8842722 DOI: 10.3389/fcimb.2022.824101] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022] Open
Abstract
Chronic Pseudomonas aeruginosa infections play an important role in the progress of lung disease in patients suffering from cystic fibrosis (CF). Recent studies indicate that polymicrobial microbiome profiles in the airway are associated with less inflammation. Thus, the hypothesis was raised that certain commensal bacteria might protect the host from inflammation. We therefore performed a screening study with commensals isolated from CF airway microbiome samples to identify potential beneficial commensals. We isolated more than 80 aerobic or facultative anaerobic commensal strains, including strains from genera Streptococcus, Neisseria, Actinomyces, Corynebacterium, Dermabacter, Micrococcus and Rothia. Through a screening experiment of co-infection in human epithelial cell lines, we identified multiple commensal strains, especially strains belonging to Streptococcus mitis, that reduced P. aeruginosa triggered inflammatory responses. The results were confirmed by co-infection experiments in ex-vivo precision cut lung slices (PCLS) from mice. The underlying mechanisms of the complex host-pathogen-commensal crosstalk were investigated from both the host and the bacterial sides with a focus on S. mitis. Transcriptome changes in the host in response to co-infection and mono-infection were evaluated, and the results indicated that several signalling pathways mediating inflammatory responses were downregulated by co-infection with S. mitis and P. aeruginosa compared to P. aeruginosa mono-infection, such as neutrophil extracellular trap formation. The genomic differences among S. mitis strains with and without protective effects were investigated by whole genome sequencing, revealing genes only present in the S. mitis strains showing protective effects. In summary, through both in vitro and ex vivo studies, we could identify a variety of commensal strains that may reduce host inflammatory responses induced by P. aeruginosa infection. These findings support the hypothesis that CF airway commensals may protect the host from inflammation.
Collapse
Affiliation(s)
- Andrew Tony-Odigie
- Institute of Medical Microbiology and Virology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Leonie Wilke
- Institute of Medical Microbiology and Virology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sébastien Boutin
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University of Heidelberg, Heidelberg, Germany
| | - Alexander H. Dalpke
- Institute of Medical Microbiology and Virology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Buqing Yi
- Institute of Medical Microbiology and Virology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- *Correspondence: Buqing Yi,
| |
Collapse
|
50
|
Krüger A, Keppel M, Sharma V, Frunzke J. The diversity of heme sensor systems - heme-responsive transcriptional regulation mediated by transient heme protein interactions. FEMS Microbiol Rev 2022; 46:6506450. [PMID: 35026033 DOI: 10.1093/femsre/fuac002] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/21/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
Heme is a versatile molecule that is vital for nearly all cellular life by serving as prosthetic group for various enzymes or as nutritional iron source for diverse microbial species. However, elevated levels of heme molecule are toxic to cells. The complexity of this stimulus has shaped the evolution of diverse heme sensor systems, which are involved in heme-dependent transcriptional regulation in eukaryotes and prokaryotes. The functions of these systems are manifold - ranging from the specific control of heme detoxification or uptake systems to the global integration of heme and iron homeostasis. This review focuses on heme sensor systems, regulating heme homeostasis by transient heme protein interaction. We provide an overview of known heme-binding motifs in prokaryotic and eukaryotic transcription factors. Besides the central ligands, the surrounding amino acid environment was shown to play a pivotal role in heme binding. The diversity of heme-regulatory systems therefore illustrates that prediction based on pure sequence information is hardly possible and requires careful experimental validation. Comprehensive understanding of heme-regulated processes is not only important for our understanding of cellular physiology, but also provides a basis for the development of novel antibacterial drugs and metabolic engineering strategies.
Collapse
Affiliation(s)
- Aileen Krüger
- Forschungszentrum Jülich GmbH, Institute for Bio- and Geosciences 1, IBG1, 52425 Jülich, Germany
| | - Marc Keppel
- Forschungszentrum Jülich GmbH, Institute for Bio- and Geosciences 1, IBG1, 52425 Jülich, Germany
| | - Vikas Sharma
- Forschungszentrum Jülich GmbH, Institute for Bio- and Geosciences 1, IBG1, 52425 Jülich, Germany
| | - Julia Frunzke
- Forschungszentrum Jülich GmbH, Institute for Bio- and Geosciences 1, IBG1, 52425 Jülich, Germany
| |
Collapse
|