1
|
Lin ZJ, Fang CY, Wang TSA. Natural and artificial siderophores: Iron-based applications and beyond. Curr Opin Chem Biol 2025; 87:102601. [PMID: 40412201 DOI: 10.1016/j.cbpa.2025.102601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/26/2025] [Accepted: 04/23/2025] [Indexed: 05/27/2025]
Abstract
Siderophores are iron chelators secreted by microorganisms to scavenge iron from the environment. Natural siderophores have gained remarkable importance because their conjugates can be applied as antibiotics and diagnostic imaging agents. By utilizing the iron uptake system of microorganisms, functional molecules such as antibiotics or imaging agents can be delivered into cells. Notably, artificial siderophores have also been developed to increase stability and broaden metal chelating diversity. Various strategies, including backbone fine-tuning, artificial chelation moieties, and direct metal swapping, can be employed. Therefore, artificial siderophores can bind biorelated metals or radioactive isotopes, expanding their biological and medical applications. The aim of this review is to introduce recent advances in natural and artificial siderophore applications and highlight future challenges in this area of research.
Collapse
Affiliation(s)
- Zih-Jheng Lin
- Department of Chemistry and Center for Emerging Material and Advanced Devices, National Taiwan University, Taipei, 106319, Taiwan, ROC
| | - Cheng-Yu Fang
- Department of Chemistry and Center for Emerging Material and Advanced Devices, National Taiwan University, Taipei, 106319, Taiwan, ROC
| | - Tsung-Shing Andrew Wang
- Department of Chemistry and Center for Emerging Material and Advanced Devices, National Taiwan University, Taipei, 106319, Taiwan, ROC.
| |
Collapse
|
2
|
Castelan-Ramírez I, Flores-Maldonado C, Hernández-Martínez D, Salazar-Villatoro L, Saucedo-Campos AD, Segura-Cobos D, Méndez-Cruz AR, Omaña-Molina M. Advances in the study of extracellular vesicles of Naegleria fowleri and their role in contact-independent pathogenic mechanisms. Parasit Vectors 2025; 18:164. [PMID: 40312759 PMCID: PMC12046931 DOI: 10.1186/s13071-025-06786-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) are spherical membrane particles released by prokaryotic and eukaryotic cells. EVs produced by pathogenic organisms are known to play a role in host-pathogen interactions; however, despite some reports on Naegleria fowleri EVs, their potential role in inducing cytopathic effects remains poorly understood. In this study, we evaluated the role of N. fowleri EVs in contact-independent pathogenic mechanisms. METHODS Extracellular vesicles were characterized via transmission electron microscopy, nanoparticle tracking analysis, SDS-PAGE, mass spectrometry, Western blotting, and zymography. EVs internalization by trophozoites and MDCK epithelial cells was also determined. Finally, mammalian cells were coincubated with EVs to evaluate haemolytic activity, epithelial paracellular ionic permeability alterations, and necrosis. RESULTS Naegleria fowleri extracellular vesicles, ranging from 82.5 to 576.5 nm in size, were isolated, with a mean of 216.8 nm and a mode of 165.3 nm. Proteomic analysis identified 1006 proteins in the EVs, including leishmanolysin, a protein associated with pathogenic mechanisms such as adhesion and enzymatic processes. The proteolytic activity of EVs was found to be primarily due to serine protease. Furthermore, EVs were internalized by both trophozoites and MDCK cells. Additionally, EVs exhibited haemolytic activity in erythrocytes as well as increased ionic permeability and necrosis in MDCK cells 24 h postinteraction. CONCLUSIONS Naegleria fowleri EVs exhibit proteolytic and haemolytic activity and are internalized by trophozoites and MDCK epithelial cell monolayers, increasing the ionic permeability of the monolayer and inducing necrosis. Furthermore, these vesicles contain molecules associated with pathogenic processes such as leishmanolysin. Our results suggest that EVs facilitate paracellular invasion, migration, and damage caused by trophozoites and play a significant role in pathogenic processes as part of a contact-independent mechanism, which, in conjunction with a contact-dependent mechanism, enhances our understanding of the pathogenicity exhibited by this amphizoic amoeba during its invasion of target tissues.
Collapse
Affiliation(s)
- Ismael Castelan-Ramírez
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Ciudad de Mexico, México
- Laboratorio de Amibas Anfizóicas, Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, Estado de México, México
| | | | - Dolores Hernández-Martínez
- Laboratorio de Amibas Anfizóicas, Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, Estado de México, México
| | | | | | - David Segura-Cobos
- Laboratorio de Amibas Anfizóicas, Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, Estado de México, México
| | - Adolfo René Méndez-Cruz
- Laboratorio de Inmunología, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, Estado de México, México
| | - Maritza Omaña-Molina
- Laboratorio de Amibas Anfizóicas, Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, Estado de México, México.
| |
Collapse
|
3
|
Randaisi VR, Bunch ML, Beavers WN, Rogers T, Mesler R, Ashurst TD, Donohoe DR, Monteith AJ, Johnson JG. Efficient gastrointestinal colonization by Campylobacter jejuni requires components of the ChuABCD heme transport system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643992. [PMID: 40166214 PMCID: PMC11957022 DOI: 10.1101/2025.03.18.643992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Previous research demonstrated that Campylobacter jejuni encodes a heme utilization system that facilitates heme-dependent growth under iron-limiting conditions and that transcription of this system is induced during human infection. Despite these observations, it remained unknown whether the heme transport system is required for colonization and disease in a susceptible host. To address this, we created individual non-polar deletion mutants of each component of the heme transport system, as well as a total deletion of the inner membrane transporter, ChuBCD, and examined their ability to promote heme-dependent growth and iron uptake. From this work, we found that only the heme receptor, ChuA, was required for heme-dependent growth and iron acquisition, which supports earlier work of another group. Further, we examined whether intestinal colonization, immune activation, and pathology were altered during infection with these mutants. After establishing that elevated heme and chuABCD expression occurs during C. jejuni infection of IL-10-/- mice, we found that heme transport mutants exhibited significantly reduced fecal shedding and colonization of the cecum and colon. In addition, we found that neutrophil and macrophage recruitment and intestinal pathology often remained intermediately elevated despite decreased bacterial loads. These results suggest that heme utilization promotes efficient colonization and full pathogenicity in C. jejuni, but that neither is completely abrogated in its absence.
Collapse
|
4
|
Lira RLDS, Nogueira FAB, Campos RDFPDC, Ferreira DRM, Roxo PLBT, de Azevedo CCS, Gimenes ECM, Bastos RLC, Nascimento CEC, Nunes FDO, Marques MCP, Campos CDL, Martinez CG, Zagmignan A, Silva LCN, Ribeiro RM, de Azevedo dos Santos APS, Carvalho RC, de Sousa EM. Mycobacterium abscessus subsp. massiliense: Biofilm Formation, Host Immune Response, and Therapeutic Strategies. Microorganisms 2025; 13:447. [PMID: 40005812 PMCID: PMC11858063 DOI: 10.3390/microorganisms13020447] [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: 01/15/2025] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Infection by Mycobacterium abscessus subsp. massiliense poses a growing public health threat, especially to immunocompromised individuals. The pathogenicity of this mycobacterium is directly linked to its ability to form biofilms, complex structures that confer resistance to antibiotics and the host immune response. The extracellular matrix of the biofilm acts as a physical barrier, hindering the penetration of drugs and the action of the immune system, while also inducing a slow-growth state that reduces susceptibility to antibiotics. Current therapies, which involve prolonged use of multiple antibiotics, are often ineffective and cause significant side effects. Therefore, it is essential to explore new strategies targeting bacterial resistance and biofilm destruction. This narrative review explores the biofilm-forming capacity of Mycobacterium abscessus subsp. massiliense and the potential of novel therapeutic strategies. Promising approaches include inhibiting biofilm formation, developing drugs with improved penetration of the extracellular matrix, combination therapies with agents that destabilize the biofilm structure, and modulating the host immune response. Investing in research and development of new therapeutic strategies is essential to combat this resistant bacterium and improve patient outcomes.
Collapse
Affiliation(s)
- 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.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Flávio Augusto Barros Nogueira
- Graduate Program in Biodiversity and Biotechnology, Amazônia—BIONORTE, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (F.A.B.N.); (A.Z.)
| | | | - Dayenne Regina Mota Ferreira
- Graduate Program in Biosciences Applied to Health, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (R.d.F.P.d.C.C.); (D.R.M.F.); (C.G.M.); (L.C.N.S.)
| | - Pedro Lucas Brito Tromps Roxo
- Undergraduate in Medicine, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.L.B.T.R.); (C.C.S.d.A.); (E.C.M.G.); (R.L.C.B.)
| | - Caio César Santana de Azevedo
- Undergraduate in Medicine, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.L.B.T.R.); (C.C.S.d.A.); (E.C.M.G.); (R.L.C.B.)
| | - Eleonôra Costa Monteiro Gimenes
- Undergraduate in Medicine, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.L.B.T.R.); (C.C.S.d.A.); (E.C.M.G.); (R.L.C.B.)
| | - Ruan Lucas Costa Bastos
- Undergraduate in Medicine, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.L.B.T.R.); (C.C.S.d.A.); (E.C.M.G.); (R.L.C.B.)
| | - Camila Evangelista Carnib Nascimento
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Flávia Danyelle Oliveira Nunes
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Mayane Cristina Pereira Marques
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Carmem Duarte Lima Campos
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Camila Guerra Martinez
- Graduate Program in Biosciences Applied to Health, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (R.d.F.P.d.C.C.); (D.R.M.F.); (C.G.M.); (L.C.N.S.)
| | - Adrielle Zagmignan
- Graduate Program in Biodiversity and Biotechnology, Amazônia—BIONORTE, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (F.A.B.N.); (A.Z.)
- Graduate Program in Biosciences Applied to Health, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (R.d.F.P.d.C.C.); (D.R.M.F.); (C.G.M.); (L.C.N.S.)
| | - Luís Cláudio Nascimento Silva
- Graduate Program in Biosciences Applied to Health, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (R.d.F.P.d.C.C.); (D.R.M.F.); (C.G.M.); (L.C.N.S.)
| | - Rachel Melo Ribeiro
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Ana Paula Silva de Azevedo dos Santos
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Rafael Cardoso Carvalho
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Eduardo Martins de Sousa
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
- Graduate Program in Biodiversity and Biotechnology, Amazônia—BIONORTE, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (F.A.B.N.); (A.Z.)
- Graduate Program in Biosciences Applied to Health, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (R.d.F.P.d.C.C.); (D.R.M.F.); (C.G.M.); (L.C.N.S.)
| |
Collapse
|
5
|
Jian J, Wei J. Ferroptosis: A New Pathway in the Interaction between Gut Microbiota and Multiple Sclerosis. FRONT BIOSCI-LANDMRK 2025; 30:26265. [PMID: 39862079 DOI: 10.31083/fbl26265] [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: 08/24/2024] [Revised: 10/28/2024] [Accepted: 11/01/2024] [Indexed: 01/27/2025]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder marked by neuroinflammation, demyelination, and neuronal damage. Recent advancements highlight a novel interaction between iron-dependent cell death, known as ferroptosis, and gut microbiota, which may significantly influences the pathophysiology of MS. Ferroptosis, driven by lipid peroxidation and tightly linked to iron metabolism, is a pivotal contributor to the oxidative stress observed in MS. Concurrently, the gut microbiota, known to affect systemic immunity and neurological health, emerges as an important regulator of iron homeostasis and inflammatory responses, thereby influencing ferroptotic pathways. This review investigates how gut microbiota dysbiosis and ferroptosis impact MS, emphasizing their potential as therapeutic targets. Through an integrated examination of mechanistic pathways and clinical evidence, we discuss how targeting these interactions could lead to novel interventions that not only modulate disease progression but also offer personalized treatment strategies based on gut microbiota profiling. This synthesis aims at deepening insights into the microbial contributions to ferroptosis and their implications in MS, setting the stage for future research and therapeutic exploration.
Collapse
Affiliation(s)
- Junjie Jian
- The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
- Department of Neurology, Yichang Central People's Hospital, 443003 Yichang, Hubei, China
| | - Jun Wei
- The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
- Department of Neurology, Yichang Central People's Hospital, 443003 Yichang, Hubei, China
| |
Collapse
|
6
|
Wander K, Ogunleye OO, Nwagu EN, Unigwe US, Odo AN, Chukwubike CM, Omilabu SA, Salu OB, Owolabi BS, Osikomaiya BI, Ebede SO, Bowale A, Olaitan AO, Chukwu CU, Ndiokwelu CO, Edu-Alamba C, Azubuike C, Odubiyi OA, Hassan YA, Oloniniyi N, Muyiwa Kelvin A, Rashidat Abiola R, Saliu A, Fadipe OO, Anyanwu RA, Orenolu MR, Abdullah MA, Ishaya OD, Agulefo CJ, Akase IE, Gauck ME, Huang Z, Chen MH, Okoror TA, Fujita M. Iron nutrition and COVID-19 among Nigerian healthcare workers. Evol Med Public Health 2024; 12:287-297. [PMID: 39759428 PMCID: PMC11697216 DOI: 10.1093/emph/eoae034] [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: 05/17/2024] [Revised: 10/07/2024] [Indexed: 01/07/2025] Open
Abstract
Background and objectives The optimal iron hypothesis (OIH) posits that risk for infection is lowest at a mild level of iron deficiency. The extent to which this protection results from arms race dynamics in the evolution of iron acquisition and sequestration mechanisms is unclear. We evaluated the OIH with regard to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), an emerging infectious agent. Methodology We tested 304 healthcare workers at baseline for iron deficiency (zinc protoporphyrin:heme), anemia (hemoglobin), and SARS-CoV-2 (salivary PCR), and followed them for ~3 months with biweekly SARS-CoV-2 tests. We fit logistic regression models based on Akaike Information Criterion. Results Adequate data were available for 199 participants. Iron replete (OR: 2.87, 95% CI: 0.85, 9.75) and anemia (OR: 2.48; 95% CI: 0.82, 7.85) were associated with higher risk for SARS-CoV-2 infection after control for covariates. Logistic regression and Cox proportional hazards models of the SARS-CoV-2 outcome were similar. Anemia (OR: 1.81; 95% CI: 0.88, 3.71) was associated with respiratory symptoms regardless of SARS-CoV-2 infection. Conclusions and implications These findings provide partial support for the OIH: SARS-CoV-2 infection risk was elevated at the high end of the range of iron availability; however, the elevated risk among those with anemia was not, as expected, specific to severe iron deficiency. Narrowly, for COVID-19 epidemiology, these findings accord with evidence that SARS-CoV-2's ability to establish infection is enhanced by access to iron. More broadly, these findings suggest that the OIH does not hinge on a long history of evolutionary arms race dynamics in access to host iron.
Collapse
Affiliation(s)
- Katherine Wander
- Department of Anthropology, Binghamton University, Binghamton, NY, USA
| | - Olayinka O Ogunleye
- Department of Medicine, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
- Department of Pharmacology, Therapeutics and Toxicology, Lagos State University College of Medicine, Ikeja, Lagos, Nigeria
| | - Evelyn N Nwagu
- Department of Human Kinetics and Health Education, University of Nigeria, Nsukka, Enugu, Nigeria
| | - Uche S Unigwe
- Department of Medicine, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Amelia N Odo
- Department of Human Kinetics and Health Education, University of Nigeria, Nsukka, Enugu, Nigeria
| | - Chinedu M Chukwubike
- Molecular Virology Unit, Department of Microbiology University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Sunday A Omilabu
- Centre for Human and Zoonotic Virology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
- Department of Medical Microbiology and Parasitology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Olumuyiwa B Salu
- Centre for Human and Zoonotic Virology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
- Department of Medical Microbiology and Parasitology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Bukola S Owolabi
- Department of Surgery, Lagos University Teaching Hospital, Idi-Araba, Mushin, Lagos, Nigeria
| | - Bodunrin I Osikomaiya
- Lagos State Blood Transfusion Service, Lagos, Nigeria
- Department of Haematology, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Samuel O Ebede
- Department of Medical Microbiology, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | | | - Abimbola O Olaitan
- Department of Medicine, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
- Olabisi Onabanjo University Teaching Hospital, Sagamu, Ogun, Nigeria
| | - Christopher U Chukwu
- Family Medicine Department, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Chibuzo O Ndiokwelu
- Department of Paediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Chioma Edu-Alamba
- Molecular Virology Unit, Department of Microbiology University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Constance Azubuike
- Molecular Virology Unit, Department of Microbiology University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | | | - Yusuf A Hassan
- Medical Emergency Unit, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | | | | | | | - Amina Saliu
- Lagos State Health Service Commission, Lagos, Nigeria
| | - Ololade O Fadipe
- Medical Emergency Unit, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Roosevelt A Anyanwu
- Centre for Human and Zoonotic Virology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Mercy R Orenolu
- Centre for Human and Zoonotic Virology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Maryam A Abdullah
- Centre for Human and Zoonotic Virology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Onyinye D Ishaya
- Usmanu Danfodiyo University Teaching Hospital, Garba Nadama, Sokoto, Nigeria
| | - Chinenye J Agulefo
- Centre for Human and Zoonotic Virology, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| | - Iorhen E Akase
- Lagos University Teaching Hospital, Idi-Araba, Mushin, Lagos, Nigeria
- College of Medicine, University of Lagos (CMUL), Idi-Araba, Lagos, Nigeria
| | - Megan E Gauck
- Department of Anthropology, Binghamton University, Binghamton, NY, USA
| | - Zifan Huang
- Department of Mathematics and Statistics, Binghamton University, Binghamton, NY, USA
| | - Mei-Hsiu Chen
- Department of Mathematics and Statistics, Binghamton University, Binghamton, NY, USA
| | - Titilayo A Okoror
- Department of Africana Studies, Binghamton University, Binghamton, NY, USA
| | - Masako Fujita
- Department of Anthropology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
7
|
Vollenweider V, Rehm K, Chepkirui C, Pérez-Berlanga M, Polymenidou M, Piel J, Bigler L, Kümmerli R. Antimicrobial activity of iron-depriving pyoverdines against human opportunistic pathogens. eLife 2024; 13:RP92493. [PMID: 39693130 DOI: 10.7554/elife.92493] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024] Open
Abstract
The global rise of antibiotic resistance calls for new drugs against bacterial pathogens. A common approach is to search for natural compounds deployed by microbes to inhibit competitors. Here, we show that the iron-chelating pyoverdines, siderophores produced by environmental Pseudomonas spp., have strong antibacterial properties by inducing iron starvation and growth arrest in pathogens. A screen of 320 natural Pseudomonas isolates used against 12 human pathogens uncovered several pyoverdines with particularly high antibacterial properties and distinct chemical characteristics. The most potent pyoverdine effectively reduced growth of the pathogens Acinetobacter baumannii, Klebsiella pneumoniae, and Staphylococcus aureus in a concentration- and iron-dependent manner. Pyoverdine increased survival of infected Galleria mellonella host larvae and showed low toxicity for the host, mammalian cell lines, and erythrocytes. Furthermore, experimental evolution of pathogens combined with whole-genome sequencing revealed limited resistance evolution compared to an antibiotic. Thus, pyoverdines from environmental strains have the potential to become a new class of sustainable antibacterials against specific human pathogens.
Collapse
Affiliation(s)
- Vera Vollenweider
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Karoline Rehm
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Clara Chepkirui
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | | | | | - Jörn Piel
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Laurent Bigler
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Rolf Kümmerli
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
Gräff ÁT, Barry SM. Siderophores as tools and treatments. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:47. [PMID: 39649077 PMCID: PMC11621027 DOI: 10.1038/s44259-024-00053-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/03/2024] [Indexed: 12/10/2024]
Abstract
In the search for iron, an essential element in many biochemical processes, microorganisms biosynthesise dedicated chelators, known as siderophores, to sequester iron from their environment and actively transport the siderophore complex into the cell. This process has been implicated in bacterial pathogenesis and exploited through siderophore-antibiotic conjugates as a method for selective antibiotic delivery. Here we review this Trojan-horse approach including design considerations and potential in diagnostics and infection imaging.
Collapse
Affiliation(s)
- Á. Tamás Gräff
- Department of Chemistry, Faculty of Natural, Mathematical and Engineering Sciences, King’s College London, Britannia House, London, SE1 1DB UK
| | - Sarah M. Barry
- Department of Chemistry, Faculty of Natural, Mathematical and Engineering Sciences, King’s College London, Britannia House, London, SE1 1DB UK
| |
Collapse
|
9
|
Shankar G, Akhter Y. Stealing survival: Iron acquisition strategies of Mycobacteriumtuberculosis. Biochimie 2024; 227:37-60. [PMID: 38901792 DOI: 10.1016/j.biochi.2024.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 06/22/2024]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), faces iron scarcity within the host due to immune defenses. This review explores the importance of iron for Mtb and its strategies to overcome iron restriction. We discuss how the host limits iron as an innate immune response and how Mtb utilizes various iron acquisition systems, particularly the siderophore-mediated pathway. The review illustrates the structure and biosynthesis of mycobactin, a key siderophore in Mtb, and the regulation of its production. We explore the potential of targeting siderophore biosynthesis and uptake as a novel therapeutic approach for TB. Finally, we summarize current knowledge on Mtb's iron acquisition and highlight promising directions for future research to exploit this pathway for developing new TB interventions.
Collapse
Affiliation(s)
- Gauri Shankar
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226 025, India
| | - Yusuf Akhter
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226 025, India.
| |
Collapse
|
10
|
Yeung YWS, Ma Y, Deng Y, Khoo BL, Chua SL. Bacterial Iron Siderophore Drives Tumor Survival and Ferroptosis Resistance in a Biofilm-Tumor Spheroid Coculture Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404467. [PMID: 39135304 PMCID: PMC11496991 DOI: 10.1002/advs.202404467] [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: 04/25/2024] [Indexed: 10/25/2024]
Abstract
Interactions between tumoral cells and tumor-associated bacteria within the tumor microenvironment play a significant role in tumor survival and progression, potentially impacting cancer treatment outcomes. In lung cancer patients, the Gram-negative pathogen Pseudomonas aeruginosa raises questions about its role in tumor survival. Here, a microfluidic-based 3D-human lung tumor spheroid-P. aeruginosa model is developed to study the bacteria's impact on tumor survival. P. aeruginosa forms a tumor-associated biofilm by producing Psl exopolysaccharide and secreting iron-scavenging pyoverdine, which is critical for establishing a bacterial community in tumors. Consequently, pyoverdine promotes cancer progression by reducing susceptibility to iron-induced death (ferroptosis), enhancing cell viability, and facilitating several cancer hallmarks, including epithelial-mesenchymal transition and metastasis. A promising combinatorial therapy approach using antimicrobial tobramycin, ferroptosis-inducing thiostrepton, and anti-cancer doxorubicin could eradicate biofilms and tumors. This work unveils a novel phenomenon of cross-kingdom cooperation, where bacteria protect tumors from death, and it paves the way for future research in developing antibiofilm cancer therapies. Understanding these interactions offers potential new strategies for combatting cancer and enhancing treatment efficacy.
Collapse
Affiliation(s)
- Yoyo Wing Suet Yeung
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityKowloonHong Kong SAR999077China
| | - Yeping Ma
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityKowloonHong Kong SAR999077China
| | - Yanlin Deng
- Department of Biomedical EngineeringCity University of Hong KongHong Kong SAR999077China
| | - Bee Luan Khoo
- Department of Biomedical EngineeringCity University of Hong KongHong Kong SAR999077China
- Hong Kong Center for Cerebro‐Cardiovascular Health Engineering (COCHE)Hong Kong SAR999077China
- City University of Hong Kong‐Shenzhen Futian Research InstituteShenzhen518000China
| | - Song Lin Chua
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityKowloonHong Kong SAR999077China
- State Key Laboratory of Chemical Biology and Drug DiscoveryThe Hong Kong Polytechnic UniversityKowloonHong Kong SAR999077China
- Research Centre of Deep Space Explorations (RCDSE)The Hong Kong Polytechnic UniversityKowloonHong Kong SAR999077China
| |
Collapse
|
11
|
Martinez E, Berg N, Rodriguez C, Daube G, Taminiau B. Influence of microbiota on the growth and gene expression of Clostridioides difficile in an in vitro coculture model. Microbiologyopen 2024; 13:e70001. [PMID: 39404502 PMCID: PMC11633334 DOI: 10.1002/mbo3.70001] [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: 04/02/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 12/13/2024] Open
Abstract
Clostridioides difficile is an anaerobic, spore-forming, Gram-positive pathogenic bacterium. This study aimed to analyze the effect of two samples of healthy fecal microbiota on C. difficile gene expression and growth using an in vitro coculture model. The inner compartment was cocultured with spores of the C. difficile polymerase chain reaction (PCR)-ribotype 078, while the outer compartment contained fecal samples from donors to mimic the microbiota (FD1 and FD2). A fecal-free plate served as a control (CT). RNA-Seq and quantitative PCR confirmation were performed on the inner compartment sample. Similarities in gene expression were observed in the presence of the microbiota. After 12 h, the expression of genes associated with germination, sporulation, toxin production, and growth was downregulated in the presence of the microbiota. At 24 h, in an iron-deficient environment, C. difficile activated several genes to counteract iron deficiency. The expression of genes associated with germination and sporulation was upregulated at 24 h compared with 12 h in the presence of microbiota from donor 1 (FD1). This study confirmed previous findings that C. difficile can use ethanolamine as a primary nutrient source. To further investigate this interaction, future studies will use a simplified coculture model with an artificial bacterial consortium instead of fecal samples.
Collapse
Affiliation(s)
- Elisa Martinez
- Department of Food Sciences, Food MicrobiologyFundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of LiegeLiegeBelgium
| | - Noémie Berg
- Department of Food Sciences, Food MicrobiologyFundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of LiegeLiegeBelgium
| | - Cristina Rodriguez
- Instituto de Investigación Biomédica de Málaga‐IBIMAMálagaSpain
- Unidadde Gestión Clínica de Aparato DigestivoHospital Universitario Virgen de laVictoriaMálagaSpain
| | - Georges Daube
- Department of Food Sciences, Food MicrobiologyFundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of LiegeLiegeBelgium
| | - Bernard Taminiau
- Department of Food Sciences, Food MicrobiologyFundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of LiegeLiegeBelgium
| |
Collapse
|
12
|
Kraihammer M, Petřík M, Rangger C, Gabriel M, Haas H, Nilica B, Virgolini I, Decristoforo C. Automated Production of [ 68Ga]Ga-Desferrioxamine B on Two Different Synthesis Platforms. Pharmaceutics 2024; 16:1231. [PMID: 39339267 PMCID: PMC11435116 DOI: 10.3390/pharmaceutics16091231] [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: 07/23/2024] [Revised: 09/11/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: PET imaging of bacterial infection could potentially provide added benefits for patient care through non-invasive means. [68Ga]Ga-desferrioxamine B-a radiolabelled siderophore-shows specific uptake by human-pathogenic bacteria like Staphylococcus aureus or Pseudomonas aeruginosa and sufficient serum stability for clinical application. In this report, we present data for automated production of [68Ga]Ga-desferrioxamine B on two different cassette-based synthesis modules (Modular-Lab PharmTracer and GRP 3V) utilising commercially obtainable cassettes together with a licensed 68Ge/68Ga radionuclide generator. Methods: Quality control, including the determination of radiochemical purity, as well as a system suitability test, was set up via RP-HPLC on a C18 column. The two described production processes use an acetic acid/acetate buffer system with ascorbic acid as a radical scavenger for radiolabelling, yielding ready-to-use formulations with sufficient activity yield. Results: Batch data analysis demonstrated radiochemical purity of >95% by RP-HPLC combined with ITLC and excellent stability up to 2 h after synthesis. Specifications for routine production were set up and validated with four masterbatches for each synthesis module. Conclusions: Based on this study, an academic clinical trial for imaging of bacterial infection was initiated. Both described synthesis methods enable automated production of [68Ga]Ga-desferrioxamine B in-house with high reproducibility for clinical application.
Collapse
Affiliation(s)
- Martin Kraihammer
- Department of Nuclear Medicine, Medical University Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
- Institute of Nuclear Medicine and Endocrinology, Kepler University Hospital, Krankenhausstrasse 9, A-4021 Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, Altenberger Strasse 69, A-4040 Linz, Austria
| | - Miloš Petřík
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, CZ-77900 Olomouc, Czech Republic
| | - Christine Rangger
- Department of Nuclear Medicine, Medical University Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Michael Gabriel
- Institute of Nuclear Medicine and Endocrinology, Kepler University Hospital, Krankenhausstrasse 9, A-4021 Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, Altenberger Strasse 69, A-4040 Linz, Austria
| | - Hubertus Haas
- Institute of Molecular Biology, Biocenter, Medical University of Innsbruck, A-6020 Innsbruck, Austria
| | - Bernhard Nilica
- Department of Nuclear Medicine, Medical University Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Irene Virgolini
- Department of Nuclear Medicine, Medical University Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| |
Collapse
|
13
|
Liu YS, Zhang C, Khoo BL, Hao P, Chua SL. Dual-species proteomics and targeted intervention of animal-pathogen interactions. J Adv Res 2024:S2090-1232(24)00383-7. [PMID: 39233003 DOI: 10.1016/j.jare.2024.08.038] [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: 06/15/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024] Open
Abstract
INTRODUCTION Host-microbe interactions are important to human health and ecosystems globally, so elucidating the complex host-microbe interactions and associated protein expressions drives the need to develop sensitive and accurate biochemical techniques. Current proteomics techniques reveal information from the point of view of either the host or microbe, but do not provide data on the corresponding partner. Moreover, it remains challenging to simultaneously study host-microbe proteomes that reflect the direct competition between host and microbe. This raises the need to develop a dual-species proteomics method for host-microbe interactions. OBJECTIVES We aim to establish a forward + reverse Stable Isotope Labeling with Amino acids in Cell culture (SILAC) proteomics approach to simultaneously label and quantify newly-expressed proteins of host and microbe without physical isolation, for investigating mechanisms in direct host-microbe interactions. METHODS Using Caenorhabditis elegans-Pseudomonas aeruginosa infection model as proof-of-concept, we employed SILAC proteomics and molecular pathway analysis to characterize the differentially-expressed microbial and host proteins. We then used molecular docking and chemical characterization to identify chemical inhibitors that intercept host-microbe interactions and eliminate microbial infection. RESULTS Based on our proteomics results, we studied the iron competition between pathogen iron scavenger and host iron uptake protein, where P. aeruginosa upregulated pyoverdine synthesis protein (PvdA) (fold-change of 5.2313) and secreted pyoverdine, and C. elegans expressed ferritin (FTN-2) (fold-change of 3.4057). Targeted intervention of iron competition was achieved using Galangin, a ginger-derived phytochemical that inhibited pyoverdine production and biofilm formation in P. aeruginosa. The Galangin-ciprofloxacin combinatorial therapy could eliminate P. aeruginosa biofilms in a fish wound infection model, and enabled animal survival. CONCLUSION Our work provides a novel SILAC-based proteomics method that can simultaneously evaluate host and microbe proteomes, with future applications in higher host organisms and other microbial species. It also provides insights into the mechanisms dictating host-microbe interactions, offering novel strategies for anti-infective therapy.
Collapse
Affiliation(s)
- Yang Sylvia Liu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region
| | - Chengqian Zhang
- School of Life Science and Technology, ShanghaiTech University, China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region; Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong Special Administrative Region; City University of Hong Kong-Shenzhen Futian Research Institute, Shenzhen, China
| | - Piliang Hao
- School of Life Science and Technology, ShanghaiTech University, China.
| | - Song Lin Chua
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region; State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region; Research Centre for Deep Space Explorations (RCDSE), The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region.
| |
Collapse
|
14
|
Lee M, Magante K, Gómez-Garzón C, Payne SM, Smith AT. Structural determinants of Vibrio cholerae FeoB nucleotide promiscuity. J Biol Chem 2024; 300:107663. [PMID: 39128725 PMCID: PMC11406355 DOI: 10.1016/j.jbc.2024.107663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/17/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024] Open
Abstract
Ferrous iron (Fe2+) is required for the growth and virulence of many pathogenic bacteria, including Vibrio cholerae (Vc), the causative agent of the disease cholera. For this bacterium, Feo is the primary system that transports Fe2+ into the cytosol. FeoB, the main component of this system, is regulated by a soluble cytosolic domain termed NFeoB. Recent reanalysis has shown that NFeoBs can be classified as either GTP-specific or NTP-promiscuous, but the structural and mechanistic bases for these differences were not known. To explore this intriguing property of FeoB, we solved the X-ray crystal structures of VcNFeoB in both the apo and the GDP-bound forms. Surprisingly, this promiscuous NTPase displayed a canonical NFeoB G-protein fold like GTP-specific NFeoBs. Using structural bioinformatics, we hypothesized that residues surrounding the nucleobase could be important for both nucleotide affinity and specificity. We then solved the X-ray crystal structures of N150T VcNFeoB in the apo and GDP-bound forms to reveal H-bonding differences surrounding the guanine nucleobase. Interestingly, isothermal titration calorimetry revealed similar binding thermodynamics of the WT and N150T proteins to guanine nucleotides, while the behavior in the presence of adenine nucleotides was dramatically different. AlphaFold models of VcNFeoB in the presence of ADP and ATP showed important conformational changes that contribute to nucleotide specificity among FeoBs. Combined, these results provide a structural framework for understanding FeoB nucleotide promiscuity, which could be an adaptive measure utilized by pathogens to ensure adequate levels of intracellular iron across multiple metabolic landscapes.
Collapse
Affiliation(s)
- Mark Lee
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland, USA
| | - Kate Magante
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland, USA
| | - Camilo Gómez-Garzón
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Shelley M Payne
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA; John Ring LaMontagne Center for Infectious Disease, University of Texas at Austin, Austin, Texas, USA
| | - Aaron T Smith
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland, USA.
| |
Collapse
|
15
|
Kuo J, Uzunovic J, Jacobson A, Dourado M, Gierke S, Rajendram M, Keilberg D, Mar J, Stekol E, Curry J, Verstraete S, Lund J, Liang Y, Tamburini FB, Omattage NS, Masureel M, Rutherford ST, Hackos DH, Tan MW, Byrd AL, Keir ME, Skippington E, Storek KM. Toxigenic Clostridium perfringens Isolated from At-Risk Paediatric Inflammatory Bowel Disease Patients. J Crohns Colitis 2024; 18:985-1001. [PMID: 38267224 PMCID: PMC11302968 DOI: 10.1093/ecco-jcc/jjae016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/22/2023] [Accepted: 01/23/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND AND AIMS This study aimed to identify microbial drivers of inflammatory bowel disease [IBD], by investigating mucosal-associated bacteria and their detrimental products in IBD patients. METHODS We directly cultured bacterial communities from mucosal biopsies from paediatric gastrointestinal patients and examined for pathogenicity-associated traits. Upon identifying Clostridium perfringens as toxigenic bacteria present in mucosal biopsies, we isolated strains and further characterized toxicity and prevalence. RESULTS Mucosal biopsy microbial composition differed from corresponding stool samples. C. perfringens was present in eight of nine patients' mucosal biopsies, correlating with haemolytic activity, but was not present in all corresponding stool samples. Large IBD datasets showed higher C. perfringens prevalence in stool samples of IBD adults [18.7-27.1%] versus healthy controls [5.1%]. In vitro, C. perfringens supernatants were toxic to cell types beneath the intestinal epithelial barrier, including endothelial cells, neuroblasts, and neutrophils, while the impact on epithelial cells was less pronounced, suggesting C. perfringens may be particularly damaging when barrier integrity is compromised. Further characterization using purified toxins and genetic insertion mutants confirmed perfringolysin O [PFO] toxin was sufficient for toxicity. Toxin RNA signatures were found in the original patient biopsies by PCR, suggesting intestinal production. C. perfringens supernatants also induced activation of neuroblast and dorsal root ganglion neurons in vitro, suggesting C. perfringens in inflamed mucosal tissue may directly contribute to abdominal pain, a frequent IBD symptom. CONCLUSIONS Gastrointestinal carriage of certain toxigenic C. perfringens may have an important pathogenic impact on IBD patients. These findings support routine monitoring of C. perfringens and PFO toxins and potential treatment in patients.
Collapse
Affiliation(s)
- James Kuo
- Department of Infectious Diseases and Host-Microbe Interactions, Genentech Inc., South San Francisco, CA, USA
| | - Jasmina Uzunovic
- Department of Bioinformatics, Genentech Inc., South San Francisco, CA, USA
| | - Amanda Jacobson
- Department of Immunology Discovery, Genentech Inc., South San Francisco, CA, USA
| | - Michelle Dourado
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, USA
| | - Sarah Gierke
- Department of Pathology, Genentech Inc., South San Francisco, CA, USA
| | - Manohary Rajendram
- Department of Infectious Diseases and Host-Microbe Interactions, Genentech Inc., South San Francisco, CA, USA
| | - Daniela Keilberg
- Department of Infectious Diseases and Host-Microbe Interactions, Genentech Inc., South San Francisco, CA, USA
| | - Jordan Mar
- Department of Human Pathobiology and OMNI Reverse Translation, Genentech Inc., South San Francisco, CA, USA
| | - Emily Stekol
- Department of Pediatrics, University of California San Francisco Benioff Children’s Hospital, San Francisco, CA, 94158, USA
| | - Joanna Curry
- Department of Pediatrics, University of California San Francisco Benioff Children’s Hospital, San Francisco, CA, 94158, USA
| | - Sofia Verstraete
- Department of Pediatrics, University of California San Francisco Benioff Children’s Hospital, San Francisco, CA, 94158, USA
| | - Jessica Lund
- Department of Microchemistry, Proteomics & Lipidomics, Genentech Inc., South San Francisco, CA, USA
| | - Yuxin Liang
- Department of Microchemistry, Proteomics & Lipidomics, Genentech Inc., South San Francisco, CA, USA
| | - Fiona B Tamburini
- Department of Human Pathobiology and OMNI Reverse Translation, Genentech Inc., South San Francisco, CA, USA
| | - Natalie S Omattage
- Department of Infectious Diseases and Host-Microbe Interactions, Genentech Inc., South San Francisco, CA, USA
| | - Matthieu Masureel
- Department of Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | - Steven T Rutherford
- Department of Infectious Diseases and Host-Microbe Interactions, Genentech Inc., South San Francisco, CA, USA
| | - David H Hackos
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, USA
| | - Man-Wah Tan
- Department of Infectious Diseases and Host-Microbe Interactions, Genentech Inc., South San Francisco, CA, USA
| | - Allyson L Byrd
- Department of Cancer Immunology, Genentech Inc., South San Francisco, CA, USA
| | - Mary E Keir
- Department of Human Pathobiology and OMNI Reverse Translation, Genentech Inc., South San Francisco, CA, USA
| | - Elizabeth Skippington
- Department of Infectious Diseases and Host-Microbe Interactions, Genentech Inc., South San Francisco, CA, USA
- Department of Bioinformatics, Genentech Inc., South San Francisco, CA, USA
| | - Kelly M Storek
- Department of Infectious Diseases and Host-Microbe Interactions, Genentech Inc., South San Francisco, CA, USA
| |
Collapse
|
16
|
Ye D, Nguyen PT, Bourgault S, Couture M. The heme binding protein ChuX is a regulator of heme degradation by the ChuS protein in Escherichia coli O157:H7. J Inorg Biochem 2024; 256:112575. [PMID: 38678912 DOI: 10.1016/j.jinorgbio.2024.112575] [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: 03/01/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Escherichia coli O157:H7 possesses an 8-gene cluster (chu genes) that contains genes involved in heme transport and processing from the human host. Among the chu genes, four encode cytoplasmic proteins (ChuS, ChuX, ChuY and ChuW). ChuX was previously shown to be a heme binding protein and to assist ChuW in heme degradation under anaerobic conditions. The purpose of this work was to investigate if ChuX works in concert with ChuS, which is a protein able to degrade heme by a non-canonical mechanism and release the iron from the porphyrin under aerobic conditions using hydrogen peroxide as the oxidant. We showed that when the heme-bound ChuX and apo-ChuS protein are mixed, heme is efficiently transferred from ChuX to ChuS. Heme-bound ChuX displayed a peroxidase activity with ABTS and H2O2 but not heme-bound ChuS, which is an efficient test to determine the protein to which heme is bound in the ChuS-ChuX complex. We found that ChuX protects heme from chemical oxidation and that it has no heme degradation activity by itself. Unexpectedly, we found that ChuX inhibits heme degradation by ChuS and stops the reaction at an early intermediate. We determined using surface plasmon resonance that ChuX interacts with ChuS and that it forms a relatively stable complex. These results indicate that ChuX in addition to its heme transfer activity is a regulator of ChuS activity, a function that was not described before for any of the heme carrier protein that delivers heme to heme degradation enzymes.
Collapse
Affiliation(s)
- Danrong Ye
- Department of Biochemistry, Microbiology and Bioinformatics, Université Laval, Quebec City, QC, Canada; Institut de Biologie Intégrative et des Systèmes (IBIS) and PROTEO, Université Laval, Quebec city, QC, Canada; Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Montreal, QC, Canada
| | - Phuong Trang Nguyen
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Montreal, QC, Canada; Departement of Chemistry, Université du Québec à Montréal, Montreal, QC, Canada
| | - Steve Bourgault
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Montreal, QC, Canada; Departement of Chemistry, Université du Québec à Montréal, Montreal, QC, Canada
| | - Manon Couture
- Department of Biochemistry, Microbiology and Bioinformatics, Université Laval, Quebec City, QC, Canada; Institut de Biologie Intégrative et des Systèmes (IBIS) and PROTEO, Université Laval, Quebec city, QC, Canada; Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Montreal, QC, Canada.
| |
Collapse
|
17
|
Chen S, Pham S, Terrapon N, Blom J, Walker ED. Elizabethkingia anophelis MSU001 Isolated from Anopheles stephensi: Molecular Characterization and Comparative Genome Analysis. Microorganisms 2024; 12:1079. [PMID: 38930461 PMCID: PMC11206156 DOI: 10.3390/microorganisms12061079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Elizabethkingia anophelis MSU001, isolated from Anopheles stephensi in the laboratory, was characterized by matrix-assisted laser desorption/ionization time of flight mass spectrometry (MALDI-ToF/MS), biochemical testing, and genome sequencing. Average nucleotide identity analysis revealed 99% identity with the type species E. anophelis R26. Phylogenetic placement showed that it formed a clade with other mosquito-associated strains and departed from a clade of clinical isolates. Comparative genome analyses further showed that it shared at least 98.6% of genes with mosquito-associated isolates (except E. anophelis As1), while it shared at most 88.8% of common genes with clinical isolates. Metabolites from MSU001 significantly inhibited growth of E. coli but not the mosquito gut symbionts Serratia marcescens and Asaia sp. W12. Insect-associated E. anophelis carried unique glycoside hydrolase (GH) and auxiliary activities (AAs) encoding genes distinct from those of clinical isolates, indicating their potential role in reshaping chitin structure and other components involved in larval development or formation of the peritrophic matrix. Like other Elizabethkingia, MSU001 also carried abundant genes encoding two-component system proteins (51), transcription factor proteins (188), and DNA-binding proteins (13). E. anophelis MSU001 contains a repertoire of antibiotic resistance genes and several virulence factors. Its potential for opportunistic infections in humans should be further evaluated prior to implementation as a paratransgenesis agent (by transgenesis of a symbiont of the vector).
Collapse
Affiliation(s)
- Shicheng Chen
- Medical Laboratory Sciences Program, College of Health and Human Sciences, Northern Illinois University, DeKalb, IL 60115, USA
| | - Steven Pham
- Corewell Health William Beaumont University Hospital, Royal Oak, MI 48073, USA;
| | - Nicolas Terrapon
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB), UMR7257 CNRS AMU, USC 1408 INRAE, 13009 Marseille, France;
| | - Jochen Blom
- Bioinformatics and Systems Biology, Justus-Liebig University Giessen, 35392 Giessen, Germany;
| | - Edward D. Walker
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI 48824, USA;
| |
Collapse
|
18
|
Zhang W, Li B, Yu R, Xu W, Liu X, Su J, Yuan G. Hepcidin contributes to largemouth bass (Micropterus salmoides) against bacterial infections. Int J Biol Macromol 2024; 266:131144. [PMID: 38556234 DOI: 10.1016/j.ijbiomac.2024.131144] [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: 09/13/2023] [Revised: 01/16/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
The increasing emergence and dissemination of bacterial pathogens in largemouth bass culture accelerate the desire for new treatment measures. Antimicrobial peptides as the host's antimicrobial source dominate the preferred molecules for discovering antibacterial agents. Here, the potential of Hepcidin-1 from largemouth bass (Micropterus salmoides) (MsHep-1) against bacterial infection is demonstrated. MsHep-1 not only improved the survival rate in infection experiments involving Nocardia seriolae (12 %) and Aeromonas hydrophila (18 %) but also coped with iron overload conditions in vivo. Moreover, the antibacterial activity of MsHep-1 in vitro was identified against both gram-negative and gram-positive bacteria. Mechanistic studies show MsHep-1 leads to bacterial death by changing the bacterial membrane potential and disrupting the bacterial membrane structure. These findings demonstrate that MsHep-1 may play an important role in the host response to bacterial infection. It provides promising strategies in the application of immunosuppression prevention and control in fish. AMPs may be a promising and available reservoir for treating the current bacterial diseases.
Collapse
Affiliation(s)
- Weixiang Zhang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Bo Li
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ruying Yu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenyan Xu
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, 450046, China
| | - Xiaoling Liu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jianguo Su
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Gailing Yuan
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
19
|
Silva LC, Sanches MS, Guidone GHM, Montini VH, de Oliva BHD, do Nascimento AB, Galhardi LCF, Kobayashi RKT, Vespero EC, Rocha SPD. Clonal relationship, virulence genes, and antimicrobial resistance of Morganella morganii isolated from community-acquired infections and hospitalized patients: a neglected opportunistic pathogen. Int Microbiol 2024; 27:411-422. [PMID: 37479959 DOI: 10.1007/s10123-023-00400-x] [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: 05/10/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/23/2023]
Abstract
Morganella morganii is a bacterium belonging to the normal intestinal microbiota and the environment; however, in immunocompromised individuals, this bacterium can become an opportunistic pathogen, causing a series of diseases, both in hospitals and in the community, being urinary tract infections more prevalent. Therefore, the objective of this study was to evaluate the prevalence, virulence profile, and resistance to antimicrobials and the clonal relationship of isolates of urinary tract infections (UTI) caused by M. morganii, both in the hospital environment and in the community of the municipality of Londrina-PR, in southern Brazil, in order to better understand the mechanisms for the establishment of the disease caused by this bacterium. Our study showed that M. morganii presents a variety of virulence factors in the studied isolates. Hospital strains showed a higher prevalence for the virulence genes zapA, iutA, and fimH, while community strains showed a higher prevalence for the ireA and iutA genes. Hospital isolates showed greater resistance compared to community isolates, as well as a higher prevalence of multidrug-resistant (MDR) and extended-spectrum beta lactamase (ESBL)-producing isolates. Several M. morganii isolates from both sources showed high genetic similarity. The most prevalent plasmid incompatibility groups detected were FIB and I1, regardless of the isolation source. Thus, M. morganii isolates can accumulate virulence factors and antimicrobial resistance, making them a neglected opportunistic pathogen.
Collapse
Affiliation(s)
- Luana Carvalho Silva
- Laboratory of Bacteriology, Department of Microbiology, Center of Biological Sciences, State University of Londrina, Rodovia Celso Garcia Cid PO-BOX 6001, 86051-980, Londrina, Paraná, Brazil
| | - Matheus Silva Sanches
- Laboratory of Bacteriology, Department of Microbiology, Center of Biological Sciences, State University of Londrina, Rodovia Celso Garcia Cid PO-BOX 6001, 86051-980, Londrina, Paraná, Brazil
| | - Gustavo Henrique Migliorini Guidone
- Laboratory of Bacteriology, Department of Microbiology, Center of Biological Sciences, State University of Londrina, Rodovia Celso Garcia Cid PO-BOX 6001, 86051-980, Londrina, Paraná, Brazil
| | - Victor Hugo Montini
- Laboratory of Bacteriology, Department of Microbiology, Center of Biological Sciences, State University of Londrina, Rodovia Celso Garcia Cid PO-BOX 6001, 86051-980, Londrina, Paraná, Brazil
| | - Bruno Henrique Dias de Oliva
- Laboratory of Bacteriology, Department of Microbiology, Center of Biological Sciences, State University of Londrina, Rodovia Celso Garcia Cid PO-BOX 6001, 86051-980, Londrina, Paraná, Brazil
| | - Arthur Bossi do Nascimento
- Laboratory of Bacteriology, Department of Microbiology, Center of Biological Sciences, State University of Londrina, Rodovia Celso Garcia Cid PO-BOX 6001, 86051-980, Londrina, Paraná, Brazil
| | - Lígia Carla Faccin Galhardi
- Laboratory of Virology, 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, Rodovia Celso Garcia Cid PO-BOX 6001, 86051-980, Londrina, Paraná, Brazil.
| |
Collapse
|
20
|
Hu T, Yang X, Zhu Y, Liu F, Yang X, Xiong Z, Liang J, Lin Z, Ran Y, Guddat LW, Rao Z, Zhang B. Molecular basis for substrate transport of Mycobacterium tuberculosis ABC importer DppABCD. SCIENCE ADVANCES 2024; 10:eadk8521. [PMID: 38507491 PMCID: PMC10954201 DOI: 10.1126/sciadv.adk8521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 02/15/2024] [Indexed: 03/22/2024]
Abstract
The type I adenosine 5'-triphosphate (ATP)-binding cassette (ABC) transporter DppABCD is believed to be responsible for the import of exogenous heme as an iron source into the cytoplasm of the human pathogen Mycobacterium tuberculosis (Mtb). Additionally, this system is also known to be involved in the acquisition of tri- or tetra-peptides. Here, we report the cryo-electron microscopy structures of the dual-function Mtb DppABCD transporter in three forms, namely, the apo, substrate-bound, and ATP-bound states. The apo structure reveals an unexpected and previously uncharacterized assembly mode for ABC importers, where the lipoprotein DppA, a cluster C substrate-binding protein (SBP), stands upright on the translocator DppBCD primarily through its hinge region and N-lobe. These structural data, along with biochemical studies, reveal the assembly of DppABCD complex and the detailed mechanism of DppABCD-mediated transport. Together, these findings provide a molecular roadmap for understanding the transport mechanism of a cluster C SBP and its translocator.
Collapse
Affiliation(s)
- Tianyu Hu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaolin Yang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen 518112, China
| | - Yuanchen Zhu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Fengjiang Liu
- Innovative Center for Pathogen Research, Guangzhou Laboratory, Guangzhou 510005, China
| | - Xiuna Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Zhiqi Xiong
- Laboratory of Structural Biology, Tsinghua University, Beijing 100084, China
| | - Jingxi Liang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300353, China
| | - Zhenli Lin
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuting Ran
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Luke W. Guddat
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zihe Rao
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen 518112, China
- Innovative Center for Pathogen Research, Guangzhou Laboratory, Guangzhou 510005, China
- Laboratory of Structural Biology, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300353, China
| | - Bing Zhang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| |
Collapse
|
21
|
Aftab H, Donegan RK. Regulation of heme biosynthesis via the coproporphyrin dependent pathway in bacteria. Front Microbiol 2024; 15:1345389. [PMID: 38577681 PMCID: PMC10991733 DOI: 10.3389/fmicb.2024.1345389] [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: 11/27/2023] [Accepted: 03/08/2024] [Indexed: 04/06/2024] Open
Abstract
Heme biosynthesis in the Gram-positive bacteria occurs mostly via a pathway that is distinct from that of eukaryotes and Gram-negative bacteria in the three terminal heme synthesis steps. In many of these bacteria heme is a necessary cofactor that fulfills roles in respiration, gas sensing, and detoxification of reactive oxygen species. These varying roles for heme, the requirement of iron and glutamate, as glutamyl tRNA, for synthesis, and the sharing of intermediates with the synthesis of other porphyrin derivatives necessitates the need for many points of regulation in response to nutrient availability and metabolic state. In this review we examine the regulation of heme biosynthesis in these bacteria via heme, iron, and oxygen species. We also discuss our perspective on emerging roles of protein-protein interactions and post-translational modifications in regulating heme biosynthesis.
Collapse
|
22
|
Li T, Cao H, Duan C, Chen S, Xu Z. Activation of CzcS/CzcR during zinc excess regulates copper tolerance and pyochelin biosynthesis of Pseudomonas aeruginosa. Appl Environ Microbiol 2024; 90:e0232723. [PMID: 38376236 PMCID: PMC10952498 DOI: 10.1128/aem.02327-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 01/24/2024] [Indexed: 02/21/2024] Open
Abstract
Zinc is an important transition metal that is essential for numerous physiological processes while excessive zinc is cytotoxic. Pseudomonas aeruginosa is a ubiquitous opportunistic human pathogen equipped with an exquisite zinc homeostatic system, and the two-component system CzcS/CzcR plays a key role in zinc detoxification. Although an increasing number of studies have shown the versatility of CzcS/CzcR, its physiological functions are still not fully understood. In this study, transcriptome analysis was performed, which revealed that CzcS/CzcR is silenced in the absence of the zinc signal but modulates global gene expression when the pathogen encounters zinc excess. CzcR was demonstrated to positively regulate the copper tolerance gene ptrA and negatively regulate the pyochelin biosynthesis regulatory gene pchR through direct binding to their promoters. Remarkably, the upregulation of ptrA and downregulation of pchR were shown to rescue the impaired capacity of copper tolerance and prevent pyochelin overproduction, respectively, caused by zinc excess. This study not only advances our understanding of the regulatory spectrum of CzcS/CzcR but also provides new insights into stress adaptation mediated by two-component systems in bacteria to balance the cellular processes that are disturbed by their signals. IMPORTANCE CzcS/CzcR is a two-component system that has been found to modulate zinc homeostasis, quorum sensing, and antibiotic resistance in Pseudomonas aeruginosa. To fully understand the physiological functions of CzcS/CzcR, we performed a comparative transcriptome analysis in this study and discovered that CzcS/CzcR controls global gene expression when it is activated during zinc excess. In particular, we demonstrated that CzcS/CzcR is critical for maintaining copper tolerance and iron homeostasis, which are disrupted during zinc excess, by inducing the expression of the copper tolerance gene ptrA and repressing the pyochelin biosynthesis genes through pchR. This study revealed the global regulatory functions of CzcS/CzcR and described a new and intricate adaptive mechanism in response to zinc excess in P. aeruginosa. The findings of this study have important implications for novel anti-infective interventions by incorporating metal-based drugs.
Collapse
Affiliation(s)
- Ting Li
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, China
| | - Huiluo Cao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cheng Duan
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, China
| | - Shuzhen Chen
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, China
| | - Zeling Xu
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, China
| |
Collapse
|
23
|
Kalalah AA, Koenig SSK, Bono JL, Bosilevac JM, Eppinger M. Pathogenomes and virulence profiles of representative big six non-O157 serogroup Shiga toxin-producing Escherichia coli. Front Microbiol 2024; 15:1364026. [PMID: 38562479 PMCID: PMC10982417 DOI: 10.3389/fmicb.2024.1364026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Shiga toxin (Stx)-producing Escherichia coli (STEC) of non-O157:H7 serotypes are responsible for global and widespread human food-borne disease. Among these serogroups, O26, O45, O103, O111, O121, and O145 account for the majority of clinical infections and are colloquially referred to as the "Big Six." The "Big Six" strain panel we sequenced and analyzed in this study are reference type cultures comprised of six strains representing each of the non-O157 STEC serogroups curated and distributed by the American Type Culture Collection (ATCC) as a resource to the research community under panel number ATCC MP-9. The application of long- and short-read hybrid sequencing yielded closed chromosomes and a total of 14 plasmids of diverse functions. Through high-resolution comparative phylogenomics, we cataloged the shared and strain-specific virulence and resistance gene content and established the close relationship of serogroup O26 and O103 strains featuring flagellar H-type 11. Virulence phenotyping revealed statistically significant differences in the Stx-production capabilities that we found to be correlated to the strain's individual stx-status. Among the carried Stx1a, Stx2a, and Stx2d phages, the Stx2a phage is by far the most responsive upon RecA-mediated phage mobilization, and in consequence, stx2a + isolates produced the highest-level of toxin in this panel. The availability of high-quality closed genomes for this "Big Six" reference set, including carried plasmids, along with the recorded genomic virulence profiles and Stx-production phenotypes will provide a valuable foundation to further explore the plasticity in evolutionary trajectories in these emerging non-O157 STEC lineages, which are major culprits of human food-borne disease.
Collapse
Affiliation(s)
- Anwar A. Kalalah
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, United States
- South Texas Center for Emerging Infectious Diseases (STCEID), San Antonio, TX, United States
| | - Sara S. K. Koenig
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, United States
- South Texas Center for Emerging Infectious Diseases (STCEID), San Antonio, TX, United States
| | - James L. Bono
- U.S. Department of Agriculture (USDA), Agricultural Research Service (ARS), U.S. Meat Animal Research Center, Clay Center, NE, United States
| | - Joseph M. Bosilevac
- U.S. Department of Agriculture (USDA), Agricultural Research Service (ARS), U.S. Meat Animal Research Center, Clay Center, NE, United States
| | - Mark Eppinger
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, United States
- South Texas Center for Emerging Infectious Diseases (STCEID), San Antonio, TX, United States
| |
Collapse
|
24
|
Senoo A, Hoshino M, Shiomi T, Nakakido M, Nagatoishi S, Kuroda D, Nakagawa I, Tame JRH, Caaveiro JMM, Tsumoto K. Structural basis for the recognition of human hemoglobin by the heme-acquisition protein Shr from Streptococcus pyogenes. Sci Rep 2024; 14:5374. [PMID: 38438508 PMCID: PMC10912661 DOI: 10.1038/s41598-024-55734-x] [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: 12/02/2023] [Accepted: 02/27/2024] [Indexed: 03/06/2024] Open
Abstract
In Gram-positive bacteria, sophisticated machineries to acquire the heme group of hemoglobin (Hb) have evolved to extract the precious iron atom contained in it. In the human pathogen Streptococcus pyogenes, the Shr protein is a key component of this machinery. Herein we present the crystal structure of hemoglobin-interacting domain 2 (HID2) of Shr bound to Hb. HID2 interacts with both, the protein and heme portions of Hb, explaining the specificity of HID2 for the heme-bound form of Hb, but not its heme-depleted form. Further mutational analysis shows little tolerance of HID2 to interfacial mutations, suggesting that its interaction surface with Hb could be a suitable candidate to develop efficient inhibitors abrogating the binding of Shr to Hb.
Collapse
Affiliation(s)
- Akinobu Senoo
- Laboratory of Protein Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Masato Hoshino
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Toshiki Shiomi
- Laboratory of Protein Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan
| | - Makoto Nakakido
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Satoru Nagatoishi
- Medical Device Development and Regulation Research Center, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Daisuke Kuroda
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Ichiro Nakagawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Jeremy R H Tame
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro, Yokohama, Kanagawa, 230-0045, Japan
| | - Jose M M Caaveiro
- Laboratory of Protein Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, 812-8582, Japan.
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
| | - Kouhei Tsumoto
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
- The Institute of Medical Sciences, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8629, Japan.
| |
Collapse
|
25
|
Mridha S, Abt MC. Starvation helps transition to abundance - a ferrosome story. Trends Microbiol 2024; 32:219-220. [PMID: 38281864 PMCID: PMC10967234 DOI: 10.1016/j.tim.2024.01.006] [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: 01/05/2024] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/30/2024]
Abstract
Iron is an essential nutrient for bacterial pathogenesis. In their study, Skaar and colleagues (Pi et al.) discovered and determined the detailed structure of ferrosomes within Clostridioides difficile, the iron-storage organelles that form under iron-limited conditions in anticipation of future iron overload.
Collapse
Affiliation(s)
- Subham Mridha
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael C Abt
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Anand N. Antiparasitic activity of the iron-containing milk protein lactoferrin and its potential derivatives against human intestinal and blood parasites. FRONTIERS IN PARASITOLOGY 2024; 2:1330398. [PMID: 39816822 PMCID: PMC11731944 DOI: 10.3389/fpara.2023.1330398] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/30/2023] [Indexed: 01/18/2025]
Abstract
An iron-containing milk protein named lactoferrin (Lf) has demonstrated antiparasitic and immunomodulatory properties against a variety of human parasites. This protein has shown its capability to bind and transport iron molecules in the vicinity of the host-pathogen environment. The ability of parasites to sequester the iron molecule and to increase their pathogenicity and survival depends on the availability of iron sources. Lf protein has suggested a iron chelating effect on parasites iron and, hence, has shown its antiparasitic effect. Since the parasites have a complex life cycle and have developed drug resistance, vaccines and other treatments are a handful. Therefore, therapeutic research focusing on natural treatment regimens that target the parasite and are non-toxic to host cells is urgently needed. The antiparasitic efficacy of Lf protein has been extensively studied over the past 40 years using both in vitro and in vivo studies. This review article highlighted past important studies on Lf protein that revealed its potential antiparasitic activity against various intracellular and extracellular intestinal or blood-borne human parasites. This review article structures the role of Lf protein in its various forms, such as native, peptide, and nanoformulation, laying the groundwork for its function as an antiparasitic agent and its possible known mechanisms of action.
Collapse
Affiliation(s)
- Namrata Anand
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
27
|
Botti V, De Bei O, Marchetti M, Campanini B, Cannistraro S, Bettati S, Bizzarri AR. Nanoscale dynamical investigation of the hemoglobin complex with the bacterial protein IsdB: is their interaction stabilized by catch bonds? NANOSCALE 2024; 16:4308-4316. [PMID: 38353599 DOI: 10.1039/d3nr05241a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Iron-regulated surface determinant B (IsdB) is a surface protein of Staphylococcus aureus that plays essential roles in host cell invasion by mediating both bacterial adhesion and hemic iron acquisition. Single-molecule experiments have recently revealed that the binding of IsdB to vitronectin and integrins is dramatically strengthened under mechanical stress conditions, promoting staphylococcal adhesion. Here we conducted atomic force spectroscopy (AFS) measurements of the interaction between IsdB and hemoglobin (Hb), in both its oxidized (metHb) and reduced forms (HbCO). While the former represents the natural substrate for IsdB, the latter is resistant to heme extraction. For the unbinding between IsdB and HbCO, we obtained a linear trend in the Bell-Evans plot, indicative of a weakening of the interaction upon mechanical stress. For the unbinding between IsdB and metHb, we found similar behavior at low loading rates. Remarkably, a non-linear trend of the complex interaction force was detected at higher force-pulling rates. Such behavior may provide some cues to the ability of IsdB to form stress-dependent bonds also with Hb, possibly enabling a more efficient heme transfer through stabilization of the transient (in vivo) IsdB-Hb complex.
Collapse
Affiliation(s)
- Valentina Botti
- Biophysics and Nanoscience Centre, DEB, Università della Tuscia, Largo dell'Università, 01100, Viterbo, Italy.
| | - Omar De Bei
- Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125, Parma, Italy.
| | - Marialaura Marchetti
- Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125, Parma, Italy.
| | - Barbara Campanini
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 23/A, 43124, Parma, Italy
| | - Salvatore Cannistraro
- Biophysics and Nanoscience Centre, DEB, Università della Tuscia, Largo dell'Università, 01100, Viterbo, Italy.
| | - Stefano Bettati
- Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125, Parma, Italy.
- Institute of Biophysics, National Research Council, via G. Moruzzi, 56124, Pisa, Italy
| | - Anna Rita Bizzarri
- Biophysics and Nanoscience Centre, DEB, Università della Tuscia, Largo dell'Università, 01100, Viterbo, Italy.
| |
Collapse
|
28
|
Xi D, Garg K, Lambert JS, Rajput-Ray M, Madigan A, Avramovic G, Gilbert L. Scrutinizing Clinical Biomarkers in a Large Cohort of Patients with Lyme Disease and Other Tick-Borne Infections. Microorganisms 2024; 12:380. [PMID: 38399784 PMCID: PMC10893018 DOI: 10.3390/microorganisms12020380] [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: 12/14/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Standard clinical markers can improve tick-borne infection (TBI) diagnoses. We investigated immune and other clinical biomarkers in 110 patients clinically diagnosed with TBIs before (T0) and after antibiotic treatment (T2). At T0, both the initial observation group and patients without seroconversion for tick-borne pathogens exhibited notably low percentages and counts of CD3 percentage (CD3%), CD3+ cells, CD8+ suppressors, CD4 percentage (CD4%), and CD4+ helper cells, with the latter group showing reductions in CD3%, CD3+, and CD8+ counts in approximately 15-22% of cases. Following treatment at the T2 follow-up, patients typically experienced enhancements in their previously low CD3%, CD3+ counts, CD4%, and CD4+ counts; however, there was no notable progress in their low CD8+ counts, and a higher number of patients presented with insufficient transferrin levels. Moreover, among those with negative serology for tick-borne infections, there was an improvement in low CD3% and CD3+ counts, which was more pronounced in patients with deficient transferrin amounts. Among those with CD57+ (n = 37) and CD19+ (n = 101) lymphocyte analysis, 59.46% of patients had a low CD57+ count, 14.85% had a low CD19 count, and 36.63% had a low CD19 percentage (CD19%). Similar findings were observed concerning low CD57+, CD19+, and CD19% markers for negative TBI serology patients. Overall, this study demonstrates that routine standard clinical markers could assist in a TBI diagnosis.
Collapse
Affiliation(s)
- David Xi
- School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland; (D.X.); (J.S.L.); (G.A.)
| | | | - John S. Lambert
- School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland; (D.X.); (J.S.L.); (G.A.)
- Infectious Diseases Department, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
- Infectious Diseases Department, The Rotunda Hospital, D01 P5W9 Dublin, Ireland
| | - Minha Rajput-Ray
- Curaidh Clinic: Innovative Solutions for Pain, Chronic Disease and Work Health, Perth PH2 8EH, UK;
| | - Anne Madigan
- School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland; (D.X.); (J.S.L.); (G.A.)
| | - Gordana Avramovic
- School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland; (D.X.); (J.S.L.); (G.A.)
| | | |
Collapse
|
29
|
Liu YC, Gong YT, Sun QY, Wang B, Yan Y, Chen YX, Zhang LJ, Zhang WD, Luan X. Ferritinophagy induced ferroptosis in the management of cancer. Cell Oncol (Dordr) 2024; 47:19-35. [PMID: 37713105 DOI: 10.1007/s13402-023-00858-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Ferroptosis, a newly form of regulated cell death (RCD), is characterized by iron dyshomeostasis and unrestricted lipid peroxidation. Emerging evidence depicts a pivotal role for ferroptosis in driving some pathological processes, especially in cancer. Triggering ferroptosis can suppress tumor growth and induce an anti-tumor immune response, denoting the therapeutic promises for targeting ferroptosis in the management of cancer. As an autophagic phenomenon, ferritinophagy is critical to induce ferroptosis by degradation of ferritin to release intracellular free iron. Recently, a great deal of effort has gone into designing and developing anti-cancer strategies based on targeting ferritinophagy to induce ferroptosis. CONCLUSION This review delineates the regulatory mechanism of ferritinophagy firstly and summarizes the role of ferritinophagy-induced ferroptosis in cancer. Moreover, the strategies targeting ferritinophagy to induce ferroptosis are highlighted to unveil the therapeutic value of ferritinophagy as a target to manage cancer. Finally, the future research directions on how to cope with the challenges in developing ferritinophagy promoters into clinical therapeutics are discussed.
Collapse
Affiliation(s)
- Yi-Chen Liu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yi-Ting Gong
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qing-Yan Sun
- Shanghai Institute of Pharmaceutical Industry, Shanghai, 200040, China
| | - Bei Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yue Yan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yi-Xu Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li-Jun Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Wei-Dong Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
30
|
Hu M, Ma Y, Chua SL. Bacterivorous nematodes decipher microbial iron siderophores as prey cue in predator-prey interactions. Proc Natl Acad Sci U S A 2024; 121:e2314077121. [PMID: 38190542 PMCID: PMC10801909 DOI: 10.1073/pnas.2314077121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024] Open
Abstract
The minimal levels of biological-available iron in the environment impose growth limitation on all living organisms. Microbes often secrete high iron-binding-affinity siderophores at high concentrations for scavenging iron from the iron-limited habitats. However, the high prevalence of siderophores released by bacteria into the environment raises an intriguing question whether this chemical cue can be detected by bacterivorous predators. Here, we show that the bacterivorous Caenorhabditis elegans nematode could employ its chemosensory receptor Odr-10 to detect pyoverdine, an iron siderophore secreted by an environmental bacterium, Pseudomonas aeruginosa. This enabled the nematode predator to migrate toward the prey. Our soil microcosm study showed that the detection of pyoverdine and subsequent feeding of P. aeruginosa prey by C. elegans could lead to the expansion of its population. These results showed that siderophores are a prey chemical cue by predators, with key implications in predator-prey interactions.
Collapse
Affiliation(s)
- Minqi Hu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region, China
| | - Yeping Ma
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region, China
| | - Song Lin Chua
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region, China
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region, China
- Research Centre for Deep Space Explorations, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region, China
- Research Institute for Future Food, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region, China
| |
Collapse
|
31
|
Kago G, Turnbough CL, Salazar JC, Payne SM. (p)ppGpp is required for virulence of Shigella flexneri. Infect Immun 2024; 92:e0033423. [PMID: 38099658 PMCID: PMC10790822 DOI: 10.1128/iai.00334-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/16/2023] [Indexed: 01/17/2024] Open
Abstract
Infection by the enteric pathogen Shigella flexneri requires transit through the gastrointestinal tract and invasion of and replication within the cells of the host colonic epithelium. This process exposes the pathogen to a range of diverse microenvironments. Furthermore, the unique composition and physical environment of the eukaryotic cell cytosol represents a stressful environment for S. flexneri, and extensive physiological adaptations are needed for the bacterium to thrive. In this work, we show that disrupting synthesis of the stringent response alarmone (p)ppGpp in S. flexneri diminished expression of key virulence genes, including ipaA, ipaB, ipaC, and icsA, and it reduced bacterial invasion and intercellular spread. Deletion of the (p)ppGpp synthase gene relA alone had no effect on S. flexneri virulence, but disruption of both relA and the (p)ppGpp synthase/hydrolase gene spoT resulted in loss of (p)ppGpp synthesis and virulence. While the relA spoT deletion mutant was able to invade a cultured human epithelial cell monolayer, albeit at reduced levels, it was unable to maintain the infection and spread to adjacent cells, as indicated by loss of plaque formation. Complementation with spoT on a plasmid vector restored plaque formation. Thus, SpoT alone is sufficient to provide the necessary level of (p)ppGpp for virulence. These results indicate that (p)ppGpp is required for S. flexneri virulence and adaptation to the intracellular environment, adding to the repertoire of signaling pathways that affect Shigella pathogenesis.
Collapse
Affiliation(s)
- Grace Kago
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Charles L. Turnbough
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Juan Carlos Salazar
- Programa de Microbiología y Micología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Shelley M. Payne
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
- John Ring LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
32
|
Reydams H, Toledo-Silva B, Mertens K, Piepers S, Vereecke N, Souza FN, Haesebrouck F, De Vliegher S. Phenotypic and genotypic assessment of iron acquisition in diverse bovine-associated non-aureus staphylococcal strains. Vet Res 2024; 55:6. [PMID: 38217046 PMCID: PMC10785429 DOI: 10.1186/s13567-023-01260-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/11/2023] [Indexed: 01/14/2024] Open
Abstract
Although the role of iron in bacterial infections has been well described for Staphylococcus (S.) aureus, iron acquisition in (bovine-associated) non-aureus staphylococci and mammaliicocci (NASM) remains insufficiently mapped. This study aimed at elucidating differences between four diverse bovine NASM field strains from two species, namely S. chromogenes and S. equorum, in regards to iron uptake (with ferritin and lactoferrin as an iron source) and siderophore production (staphyloferrin A and staphyloferrin B) by investigating the relationship between the genetic basis of iron acquisition through whole genome sequencing (WGS) with their observed phenotypic behavior. The four field strains were isolated in a previous study from composite cow milk (CCM) and bulk tank milk (BTM) in a Flemish dairy herd. Additionally, two well-studied S. chromogenes isolates originating from a persistent intramammary infection and from a teat apex were included for comparative purpose in all assays. Significant differences between species and strains were identified. In our phenotypical iron acquisition assay, while lactoferrin had no effect on growth recovery for all strains in iron deficient media, we found that ferritin served as an effective source for growth recovery in iron-deficient media for S. chromogenes CCM and BTM strains. This finding was further corroborated by analyzing potential ferritin iron acquisition genes using whole-genome sequencing data, which showed that all S. chromogenes strains contained hits for all three proposed ferritin reductive pathway genes. Furthermore, a qualitative assay indicated siderophore production by all strains, except for S. equorum. This lack of siderophore production in S. equorum was supported by a quantitative assay, which revealed significantly lower or negligible siderophore amounts compared to S. aureus and S. chromogenes. The WGS analysis showed that all tested strains, except for S. equorum, possessed complete staphyloferrin A (SA)-synthesis and export operons, which likely explains the phenotypic absence of siderophore production in S. equorum strains. While analyzing the staphyloferrin A and staphyloferrin B operon landscapes for all strains, we noticed some differences in the proteins responsible for iron acquisition between different species. However, within strains of the same species, the siderophore-related proteins remained conserved. Our findings contribute valuable insights into the genetic elements associated with bovine NASM pathogenesis.
Collapse
Affiliation(s)
- Helena Reydams
- M-Team and Mastitis and Milk Quality Research Unit, Department of Internal Medicine, Reproduction, and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium.
| | - Bruno Toledo-Silva
- M-Team and Mastitis and Milk Quality Research Unit, Department of Internal Medicine, Reproduction, and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Kristien Mertens
- M-Team and Mastitis and Milk Quality Research Unit, Department of Internal Medicine, Reproduction, and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Sofie Piepers
- M-Team and Mastitis and Milk Quality Research Unit, Department of Internal Medicine, Reproduction, and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Nick Vereecke
- Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
- PathoSense BV, Lier, Belgium
| | - Fernando Nogueira Souza
- Veterinary Clinical Immunology Research Group, Department of Internal Medicine, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Prof. Orlando Marques de Paiva Av. 87, São Paulo, 05508-270, Brazil
| | - Freddy Haesebrouck
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Sarne De Vliegher
- M-Team and Mastitis and Milk Quality Research Unit, Department of Internal Medicine, Reproduction, and Population Medicine, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| |
Collapse
|
33
|
Rhoades NS, Cinco IR, Hendrickson SM, Prongay K, Haertel AJ, Flores GE, Slifka MK, Messaoudi I. Infant diarrheal disease in rhesus macaques impedes microbiome maturation and is linked to uncultured Campylobacter species. Commun Biol 2024; 7:37. [PMID: 38182754 PMCID: PMC10770169 DOI: 10.1038/s42003-023-05695-0] [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: 07/04/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024] Open
Abstract
Diarrheal diseases remain one of the leading causes of death for children under 5 globally, disproportionately impacting those living in low- and middle-income countries (LMIC). Campylobacter spp., a zoonotic pathogen, is one of the leading causes of food-borne infection in humans. Yet to be cultured Campylobacter spp. contribute to the total burden in diarrheal disease in children living in LMIC thus hampering interventions. We performed microbiome profiling and metagenomic genome assembly on samples collected from over 100 infant rhesus macaques longitudinally and during cases of clinical diarrhea within the first year of life. Acute diarrhea was associated with long-lasting taxonomic and functional shifts of the infant gut microbiome indicative of microbiome immaturity. We constructed 36 Campylobacter metagenomic assembled genomes (MAGs), many of which fell within 4 yet to be cultured species. Finally, we compared the uncultured Campylobacter MAGs assembled from infant macaques with publicly available human metagenomes to show that these uncultured species are also found in human fecal samples from LMIC. These data highlight the importance of unculturable Campylobacter spp. as an important target for reducing disease burden in LMIC children.
Collapse
Affiliation(s)
- Nicholas S Rhoades
- Department of Molecular biology and Biochemistry, University of California Irvine, Irvine, CA, USA
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Isaac R Cinco
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Sara M Hendrickson
- Division of Neuroscience, Oregon National Primate Research Center, Portland, OR, USA
| | - Kamm Prongay
- Division of Animal Resources and Research Support, Oregon National Primate Research Center, Oregon Health and Science University West Campus, Portland, OR, USA
| | - Andrew J Haertel
- Division of Animal Resources and Research Support, Oregon National Primate Research Center, Oregon Health and Science University West Campus, Portland, OR, USA
| | - Gilberto E Flores
- Department of Biology, California State University, Northridge, Northridge, CA, USA
| | - Mark K Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Portland, OR, USA
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
34
|
Ding X, Robbe-Masselot C, Fu X, Léonard R, Marsac B, Dauriat CJG, Lepissier A, Rytter H, Ramond E, Dupuis M, Euphrasie D, Dubail I, Schimmich C, Qin X, Parraga J, Leite-de-Moraes M, Ferroni A, Chassaing B, Sermet-Gaudelus I, Charbit A, Coureuil M, Jamet A. Airway environment drives the selection of quorum sensing mutants and promote Staphylococcus aureus chronic lifestyle. Nat Commun 2023; 14:8135. [PMID: 38065959 PMCID: PMC10709412 DOI: 10.1038/s41467-023-43863-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Staphylococcus aureus is a predominant cause of chronic lung infections. While the airway environment is rich in highly sialylated mucins, the interaction of S. aureus with sialic acid is poorly characterized. Using S. aureus USA300 as well as clinical isolates, we demonstrate that quorum-sensing dysfunction, a hallmark of S. aureus adaptation, correlates with a greater ability to consume free sialic acid, providing a growth advantage in an air-liquid interface model and in vivo. Furthermore, RNA-seq experiment reveals that free sialic acid triggers transcriptional reprogramming promoting S. aureus chronic lifestyle. To support the clinical relevance of our results, we show the co-occurrence of S. aureus, sialidase-producing microbiota and free sialic acid in the airway of patients with cystic fibrosis. Our findings suggest a dual role for sialic acid in S. aureus airway infection, triggering virulence reprogramming and driving S. aureus adaptive strategies through the selection of quorum-sensing dysfunctional strains.
Collapse
Affiliation(s)
- Xiongqi Ding
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Catherine Robbe-Masselot
- Université Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000, Lille, France
| | - Xiali Fu
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Renaud Léonard
- Université Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000, Lille, France
| | - Benjamin Marsac
- Université Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000, Lille, France
| | - Charlene J G Dauriat
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Team «Mucosal Microbiota in Chronic Inflammatory Diseases», F75014, Paris, France
| | - Agathe Lepissier
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Héloïse Rytter
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Elodie Ramond
- Genoscope, UMR8030, Laboratory of Systems & Synthetic Biology (LISSB), Xenome team, F91057, Evry, France
| | - Marion Dupuis
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Daniel Euphrasie
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Iharilalao Dubail
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Cécile Schimmich
- Anses, Laboratory of Animal Health in Normandy, Physiopathology and epidemiology of equine diseases (PhEED), RD 675, F14430, Goustranville, France
| | - Xiaoquan Qin
- Université Paris Cité, Institut de physique du globe de Paris, CNRS, F75005, Paris, France
| | - Jessica Parraga
- Department of Clinical Microbiology, Necker-Enfants Malades Hospital, AP-HP Centre Université de Paris Cité, F75015, Paris, France
| | - Maria Leite-de-Moraes
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Agnes Ferroni
- Department of Clinical Microbiology, Necker-Enfants Malades Hospital, AP-HP Centre Université de Paris Cité, F75015, Paris, France
| | - Benoit Chassaing
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Team «Mucosal Microbiota in Chronic Inflammatory Diseases», F75014, Paris, France
| | - Isabelle Sermet-Gaudelus
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Alain Charbit
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Mathieu Coureuil
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France.
| | - Anne Jamet
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France.
- Department of Clinical Microbiology, Necker-Enfants Malades Hospital, AP-HP Centre Université de Paris Cité, F75015, Paris, France.
| |
Collapse
|
35
|
Gnanagobal H, Cao T, Hossain A, Vasquez I, Chakraborty S, Chukwu-Osazuwa J, Boyce D, Espinoza MJ, García-Angulo VA, Santander J. Role of riboflavin biosynthesis gene duplication and transporter in Aeromonas salmonicida virulence in marine teleost fish. Virulence 2023; 14:2187025. [PMID: 36895132 PMCID: PMC10012899 DOI: 10.1080/21505594.2023.2187025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
Abstract
Active flavins derived from riboflavin (vitamin B2) are essential for life. Bacteria biosynthesize riboflavin or scavenge it through uptake systems, and both mechanisms may be present. Because of riboflavin's critical importance, the redundancy of riboflavin biosynthetic pathway (RBP) genes might be present. Aeromonas salmonicida, the aetiological agent of furunculosis, is a pathogen of freshwater and marine fish, and its riboflavin pathways have not been studied. This study characterized the A. salmonicida riboflavin provision pathways. Homology search and transcriptional orchestration analysis showed that A. salmonicida has a main riboflavin biosynthetic operon that includes ribD, ribE1, ribBA, and ribH genes. Outside the main operon, putative duplicated genes ribA, ribB and ribE, and a ribN riboflavin importer encoding gene, were found. Monocistronic mRNA ribA, ribB and ribE2 encode for their corresponding functional riboflavin biosynthetic enzyme. While the product of ribBA conserved the RibB function, it lacked the RibA function. Likewise, ribN encodes a functional riboflavin importer. Transcriptomics analysis indicated that external riboflavin affected the expression of a relatively small number of genes, including a few involved in iron metabolism. ribB was downregulated in response to external riboflavin, suggesting negative feedback. Deletion of ribA, ribB and ribE1 showed that these genes are required for A. salmonicida riboflavin biosynthesis and virulence in Atlantic lumpfish (Cyclopterus lumpus). A. salmonicida riboflavin auxotrophic attenuated mutants conferred low protection to lumpfish against virulent A. salmonicida. Overall, A. salmonicida has multiple riboflavin endowment forms, and duplicated riboflavin provision genes are critical for A. salmonicida infection.
Collapse
Affiliation(s)
- Hajarooba Gnanagobal
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St John's, Canada
| | - Trung Cao
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St John's, Canada
| | - Ahmed Hossain
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St John's, Canada
| | - Ignacio Vasquez
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St John's, Canada
| | - Setu Chakraborty
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St John's, Canada
| | - Joy Chukwu-Osazuwa
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St John's, Canada
| | - Danny Boyce
- The Dr. Joe Brown Aquatic Research Building (JBARB), Ocean Sciences Centre, Memorial University of Newfoundland, St John's, Canada
| | - María Jesus Espinoza
- Microbiology and Mycology Program, Institute of Biomedical Sciences, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Víctor Antonio García-Angulo
- Microbiology and Mycology Program, Institute of Biomedical Sciences, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Javier Santander
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St John's, Canada
| |
Collapse
|
36
|
Tan CG, Oberlag NM, McGowan AE, Dawrs SN, Chan YL, Strong M, Hasan NA, Honda JR. Genomic and microbiological analyses of iron acquisition pathways among respiratory and environmental nontuberculous mycobacteria from Hawai'i. Front Microbiol 2023; 14:1268963. [PMID: 38029173 PMCID: PMC10667711 DOI: 10.3389/fmicb.2023.1268963] [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: 08/01/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
As environmental opportunistic pathogens, nontuberculous mycobacteria (NTM) can cause severe and difficult to treat pulmonary disease. In the United States, Hawai'i has the highest prevalence of infection. Rapid growing mycobacteria (RGM) such as Mycobacterium abscessus and M. porcinum and the slow growing mycobacteria (SGM) including M. intracellulare subspecies chimaera are common environmental NTM species and subspecies in Hawai'i. Although iron acquisition is an essential process of many microorganisms, iron acquisition via siderophores among the NTM is not well-characterized. In this study, we apply genomic and microbiological methodologies to better understand iron acquisition via siderophores for environmental and respiratory isolates of M. abscessus, M. porcinum, and M. intracellulare subspecies chimaera from Hawai'i. Siderophore synthesis and transport genes, including mycobactin (mbt), mmpL/S, and esx-3 were compared among 47 reference isolates, 29 respiratory isolates, and 23 environmental Hawai'i isolates. Among all reference isolates examined, respiratory isolates showed significantly more siderophore pertinent genes compared to environmental isolates. Among the Hawai'i isolates, RGM M. abscessus and M. porcinum had significantly less esx-3 and mbt genes compared to SGM M. chimaera when stratified by growth classification. However, no significant differences were observed between the species when grown on low iron culture agar or siderophore production by the chrome azurol S (CAS) assay in vitro. These results indicate the complex mechanisms involved in iron sequestration and siderophore activity among diverse NTM species.
Collapse
Affiliation(s)
| | - Nicole M. Oberlag
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, United States
| | | | - Stephanie N. Dawrs
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, United States
| | | | - Michael Strong
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, United States
| | - Nabeeh A. Hasan
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, United States
| | - Jennifer R. Honda
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, United States
- Department of Cellular and Molecular Biology, School of Medicine, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| |
Collapse
|
37
|
Yamada K, Tazaki A, Ushio-Watanabe N, Usui Y, Takeda A, Matsunaga M, Suzumura A, Shimizu H, Zheng H, Ariefta NR, Yamamoto M, Hara H, Goto H, Sonoda KH, Nishiguchi KM, Kato M, Nishikawa Y, Toyokuni S, Kaneko H. Retinal ferroptosis as a critical mechanism for the induction of retinochoroiditis during ocular toxoplasmosis. Redox Biol 2023; 67:102890. [PMID: 37738924 PMCID: PMC10519826 DOI: 10.1016/j.redox.2023.102890] [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: 09/03/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023] Open
Abstract
Toxoplasmosis is a major infectious disease, affecting approximately one-third of the world's population; its main clinical manifestation, ocular toxoplasmosis (OT), is a severe sight-threatening disease. Nevertheless, the diagnosis of OT is based on clinical findings, which needs improvement, even with biochemical tests, such as polymerase chain reaction and antibody detections. Furthermore, the efficacy of OT-targeted treatment is limited; thus, additional measures for diagnosis and treatments are needed. Here, we for the first time report a significantly reduced iron concentration in the vitreous humor (VH) of human patients infected with OT. To obtain further insights into molecular mechanisms, we established a mouse model of T. gondii infection, in which intravitreally injected tracer 57Fe, was accumulated in the neurosensory retina. T. gondii-infected eyes showed increased lipid peroxidation, reduction of glutathione peroxidase-4 expression and mitochondrial deformity in the photoreceptor as cristae loss. These findings strongly suggest the involvement of ferroptotic process in the photoreceptor of OT. In addition, deferiprone, an FDA-approved iron chelator, reduced the iron uptake but also ameliorated toxoplasma-induced retinochoroiditis by reducing retinal inflammation. In conclusion, the iron levels in the VH could serve as diagnostic markers and iron chelators as potential treatments for OT.
Collapse
Affiliation(s)
- Kazuhisa Yamada
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| | - Akira Tazaki
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| | - Nanako Ushio-Watanabe
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, 080-8555, Japan.
| | - Yoshihiko Usui
- Department of Ophthalmology, Tokyo Medical University, Tokyo, 160-8402, Japan.
| | - Atsunobu Takeda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Masaaki Matsunaga
- Department of Public Health, Fujita Health University School of Medicine, Toyoake, 470-1192, Japan.
| | - Ayana Suzumura
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| | - Hideyuki Shimizu
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| | - Hao Zheng
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| | - Nanang R Ariefta
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, 080-8555, Japan.
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.
| | - Hideaki Hara
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, 501-1196, Japan.
| | - Hiroshi Goto
- Department of Ophthalmology, Tokyo Medical University, Tokyo, 160-8402, Japan.
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Koji M Nishiguchi
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| | - Masashi Kato
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| | - Yoshifumi Nishikawa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, 080-8555, Japan.
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan; Center for Low-Temperature Plasma Sciences, Nagoya University, Furo-Cho, Chikusa-ku, Nagoya, 464-8603, Japan.
| | - Hiroki Kaneko
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| |
Collapse
|
38
|
Durrani B, Mohammad A, Ljubetic BM, Dobberfuhl AD. The Potential Role of Persister Cells in Urinary Tract Infections. Curr Urol Rep 2023; 24:541-551. [PMID: 37907771 DOI: 10.1007/s11934-023-01182-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2023] [Indexed: 11/02/2023]
Abstract
PURPOSE OF REVIEW This review explores the role of persister cells in urinary tract infections (UTIs). UTIs are one of the most common bacterial infections, affecting millions of people worldwide. Persister cells are a subpopulation of bacteria with dormant metabolic activity which allows survival in the presence of antibiotics. RECENT FINDINGS This review summarizes recent research on the pathogenesis of persister cell formation in UTIs, the impact of persister cells on the effectiveness of antibiotics, the challenges they pose for treatment, and the need for new strategies to target these cells. Furthermore, this review examines the current state of research on the identification and characterization of persister cells in UTIs, as well as the future directions for investigations in this field. This review highlights the importance of understanding the role of persister cells in UTIs and the potential impact of targeting these cells in the development of new treatments.
Collapse
Affiliation(s)
- Butool Durrani
- Department of Internal Medicine, Aga Khan University Hospital, National Stadium Rd, Karachi, Karachi City, Pakistan
| | - Ashu Mohammad
- Department of Urology, Center for Academic Medicine, Stanford University School of Medicine, 453 Quarry Road, Urology-5656, Palo Alto, CA, 94304, USA
| | - Bernardita M Ljubetic
- Department of Urology, Center for Academic Medicine, Stanford University School of Medicine, 453 Quarry Road, Urology-5656, Palo Alto, CA, 94304, USA
| | - Amy D Dobberfuhl
- Department of Urology, Center for Academic Medicine, Stanford University School of Medicine, 453 Quarry Road, Urology-5656, Palo Alto, CA, 94304, USA.
| |
Collapse
|
39
|
Zhao Q, Yan J, Wang J, Liu R, Bartlam M. Structural analysis of the ferric-binding protein KfuA from Klebsiella pneumoniae. Biochem Biophys Res Commun 2023; 679:52-57. [PMID: 37669596 DOI: 10.1016/j.bbrc.2023.08.068] [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: 08/09/2023] [Accepted: 08/31/2023] [Indexed: 09/07/2023]
Abstract
Iron acquisition is an essential process of cell physiology for biological systems. In Klebsiella pneumoniae, the siderophore and ferric-acquisition ABC (ATP-Binding-Cassette) transporter KfuABC is utilized for iron uptake. Initial recognition of the various ferric sources in periplasm and transportation across the cytoplasmic membrane is performed by the substrate-binding protein (SBP) KfuA. Here we report the 2.0 Å resolution crystal structure of KfuA from K. pneumoniae, which crystallizes in the space group P1211 with a single monomer in the asymmetric unit. A bound metal ion reveals the residues required for binding ferric ions. Binding analysis shows that ferric iron and the iron-mimicking gallium bind with high affinity to KfuA. Growth curves show that gallium inhibits growth of K. pneumoniae whereas ferric iron enhances it. This work suggests a mechanism whereby gallium effectively competes with ferric iron, disrupting iron-dependent biological functions via binding to KfuA and leading to heightened antimicrobial efficacy. Significantly, humans lack equivalent ABC transporters like SBP KfuA, underscoring the potential of KfuA as an attractive target for therapeutic intervention.
Collapse
Affiliation(s)
- Qi Zhao
- College of Life Sciences, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Jiaqi Yan
- College of Life Sciences, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Jingjing Wang
- College of Life Sciences, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Ruihua Liu
- College of Life Sciences, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.
| | - Mark Bartlam
- College of Life Sciences, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China; Nankai International Advanced Research Institute (Shenzhen Futian), Nankai University, Tianjin, 300071, China.
| |
Collapse
|
40
|
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
|
41
|
Kahl LJ, Stremmel N, Esparza-Mora MA, Wheatley RM, MacLean RC, Ralser M. Interkingdom interactions between Pseudomonas aeruginosa and Candida albicans affect clinical outcomes and antimicrobial responses. Curr Opin Microbiol 2023; 75:102368. [PMID: 37677865 DOI: 10.1016/j.mib.2023.102368] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 09/09/2023]
Abstract
Infections that involve interkingdom microbial communities, such as those between bacteria and yeast pathogens, are difficult to treat, associated with worse patient outcomes, and may be a source of antimicrobial resistance. In this review, we address co-occurrence and co-infections of Candida albicans and Pseudomonas aeruginosa, two pathogens that occupy multiple infection niches in the human body, especially in immunocompromised patients. The interaction between the pathogen species influences microbe-host interactions, the effectiveness of antimicrobials and even infection outcomes, and may thus require adapted treatment strategies. However, the molecular details of bacteria-fungal interactions both inside and outside the infection sites, are insufficiently characterised. We argue that comprehensively understanding the P. aeruginosa-C. albicans interaction network through integrated systems biology approaches will capture the highly dynamic and complex nature of these polymicrobial infections and lead to a more comprehensive understanding of clinical observations such as reshaped immune defences and low antimicrobial treatment efficacy.
Collapse
Affiliation(s)
- Lisa J Kahl
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | - Nina Stremmel
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | | | - Rachel M Wheatley
- University of Oxford, Department of Biology, Oxford OX1 3SZ, United Kingdom
| | - R Craig MacLean
- University of Oxford, Department of Biology, Oxford OX1 3SZ, United Kingdom
| | - Markus Ralser
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany; University of Oxford, The Wellcome Centre for Human Genetics, Nuffield Department of Medicine, Oxford OX3 7BN, United Kingdom; Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany.
| |
Collapse
|
42
|
Ferguson D, Ryder R, Lunsford R, Dash A, Kamali A, Kimura A, Crandall J, Mukhopadhyay R, Dowless H, Ortiz N, Jue NK. Serratia marcescens Outbreak at a Correctional Facility: Environmental Sampling, Laboratory Analyses and Genomic Characterization to Assess Sources and Persistence. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6709. [PMID: 37681849 PMCID: PMC10487681 DOI: 10.3390/ijerph20176709] [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: 07/01/2023] [Revised: 08/12/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023]
Abstract
Serratia marcescens is an environmental bacterium and clinical pathogen that can cause an array of infections. We describe an environmental sampling and comparative genomics approach used to investigate a multi-year outbreak of S. marcescens at a correctional facility. Whole genome sequencing analysis revealed a predominant cluster of clonally related S. marcescens from nine patient cases and items associated with illicit drug use. Closely related strains found among items associated with case-patient cells and diluted Cell Block 64 (CB64), a quaternary ammonium disinfectant, and Break Out (BO), a multipurpose cleaner, highlighted their role as environmental reservoirs for S. marcescens in this outbreak. Comparative genomic analysis suggested outbreak strains were both persistent (identical strains found over long periods and in multiple locations of the correctional facility) and diverse (strains clustered with multiple global samples from NCBI database). No correlation was found between antimicrobial resistance (AMR) genes of outbreak strains; NCBI strains have more AMR genes. Principal component analysis (PCA) of virulence factors associated with persistence and infectivity indicated variation based on phylogroups, including the predominant cluster; identifiable variations among environmental versus clinical strains were not observed. Identification of multiple distinct genetic groups highlights the importance of putting epidemiological genomic studies in a proper genetic context.
Collapse
Affiliation(s)
- Donna Ferguson
- Public Health Laboratory, County of Monterey Health Department, Salinas, CA 93906, USA
| | - Rahil Ryder
- Public Health Laboratory, County of Monterey Health Department, Salinas, CA 93906, USA
| | - Rawni Lunsford
- Public Health Laboratory, County of Monterey Health Department, Salinas, CA 93906, USA
| | - Arie Dash
- Public Health Laboratory, County of Monterey Health Department, Salinas, CA 93906, USA
| | - Amanda Kamali
- Public Health, Medical Services Division, California Correctional Health Care Services, Elk Grove, CA 95758, USA
| | - Akiko Kimura
- Infectious Diseases Branch, Center for Infectious Diseases, California Department of Public Health, Richmond, CA 94804, USA
| | - John Crandall
- Microbial Diseases Laboratory Branch, Center for Laboratory Sciences, California Department of Public Health, Richmond, CA 94804, USA
| | - Rituparna Mukhopadhyay
- Microbial Diseases Laboratory Branch, Center for Laboratory Sciences, California Department of Public Health, Richmond, CA 94804, USA
| | - Heather Dowless
- Public Health, Medical Services Division, California Correctional Health Care Services, Elk Grove, CA 95758, USA
| | - Nancy Ortiz
- Infectious Diseases Branch, Center for Infectious Diseases, California Department of Public Health, Richmond, CA 94804, USA
| | - Nathaniel K. Jue
- Department of Biology and Chemistry, California State University, Monterey Bay, Seaside, CA 93955, USA
| |
Collapse
|
43
|
de Miranda R, Cuthbert BJ, Klevorn T, Chao A, Mendoza J, Arbing M, Sieminski PJ, Papavinasasundaram K, Abdul-Hafiz S, Chan S, Sassetti CM, Ehrt S, Goulding CW. Differentiating the roles of Mycobacterium tuberculosis substrate binding proteins, FecB and FecB2, in iron uptake. PLoS Pathog 2023; 19:e1011650. [PMID: 37747938 PMCID: PMC10553834 DOI: 10.1371/journal.ppat.1011650] [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: 05/07/2023] [Revised: 10/05/2023] [Accepted: 08/31/2023] [Indexed: 09/27/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, poses a great threat to human health. With the emergence of drug resistant Mtb strains, new therapeutics are desperately needed. As iron is critical to the growth and survival of Mtb, mechanisms through which Mtb acquires host iron represent attractive therapeutic targets. Mtb scavenges host iron via Mtb siderophore-dependent and heme iron uptake pathways. While multiple studies describe the import of heme and ferric-siderophores and the export of apo-siderophores across the inner membrane, little is known about their transport across the periplasm and cell-wall environments. Mtb FecB and FecB2 are predicted periplasmic binding proteins implicated in host iron acquisition; however, their precise roles are not well understood. This study sought to differentiate the roles FecB and FecB2 play in Mtb iron acquisition. The crystallographic structures of Mtb FecB and FecB2 were determined to 2.0 Å and 2.2 Å resolution, respectively, and show distinct ligand binding pockets. In vitro ligand binding experiments for FecB and FecB2 were performed with heme and bacterial siderophores from Mtb and other species, revealing that both FecB and FecB2 bind heme, while only FecB binds the Mtb sideophore ferric-carboxymycobactin (Fe-cMB). Subsequent structure-guided mutagenesis of FecB identified a single glutamate residue-Glu339-that significantly contributes to Fe-cMB binding. A role for FecB in the Mtb siderophore-mediated iron acquisition pathway was corroborated by Mycobacterium smegmatis and Mtb pull-down assays, which revealed interactions between FecB and members of the mycobacterial siderophore export and import machinery. Similarly, pull-down assays with FecB2 confirms its role in heme uptake revealing interactions with a potential inner membrane heme importer. Due to ligand preference and protein partners, our data suggest that Mtb FecB plays a role in siderophore-dependent iron and heme acquisition pathways; in addition, we confirm that Mtb FecB2 is involved in heme uptake.
Collapse
Affiliation(s)
- Rodger de Miranda
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Bonnie J. Cuthbert
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Thaís Klevorn
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | - Alex Chao
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Jessica Mendoza
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Mark Arbing
- UCLA-DOE Institute, UCLA, Los Angeles, Calofornia, United States of America
| | - Paul J. Sieminski
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Kadamba Papavinasasundaram
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, Massachusetts, United States of America
| | - Sumer Abdul-Hafiz
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Sum Chan
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | - Christopher M. Sassetti
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, Massachusetts, United States of America
| | - Sabine Ehrt
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | - Celia W. Goulding
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, Califiornia, United States of America
| |
Collapse
|
44
|
Truong VK, Hayles A, Bright R, Luu TQ, Dickey MD, Kalantar-Zadeh K, Vasilev K. Gallium Liquid Metal: Nanotoolbox for Antimicrobial Applications. ACS NANO 2023; 17:14406-14423. [PMID: 37506260 DOI: 10.1021/acsnano.3c06486] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
The proliferation of drug resistance in microbial pathogens poses a significant threat to human health. Hence, treatment measures are essential to surmount this growing problem. In this context, liquid metal nanoparticles are promising. Gallium, a post-transition metal notable for being a liquid at physiological temperature, has drawn attention for its distinctive properties, high antimicrobial efficacy, and low toxicity. Moreover, gallium nanoparticles demonstrate anti-inflammatory properties in immune cells. Gallium can alloy with other metals and be prepared in various composites to modify and tailor its characteristics and functionality. More importantly, the bactericidal mechanism of gallium liquid metal could sidestep the threat of emerging drug resistance mechanisms. Building on this rationale, gallium-based liquid metal nanoparticles can enable impactful and innovative strategic pathways in the battle against antimicrobial resistance. This review outlines the characteristics of gallium-based liquid metals at the nanoscale and their corresponding antimicrobial mechanisms to provide a comprehensive yet succinct overview of their current antimicrobial applications. In addition, challenges and opportunities that require further research efforts have been identified and discussed.
Collapse
Affiliation(s)
- Vi Khanh Truong
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Andrew Hayles
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Richard Bright
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Trong Quan Luu
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Michael D Dickey
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Kourosh Kalantar-Zadeh
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Krasimir Vasilev
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| |
Collapse
|
45
|
Gómez-Garzón C, Payne SM. Divide and conquer: genetics, mechanism, and evolution of the ferrous iron transporter Feo in Helicobacter pylori. Front Microbiol 2023; 14:1219359. [PMID: 37469426 PMCID: PMC10353542 DOI: 10.3389/fmicb.2023.1219359] [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: 05/09/2023] [Accepted: 06/14/2023] [Indexed: 07/21/2023] Open
Abstract
Introduction Feo is the most widespread and conserved system for ferrous iron uptake in bacteria, and it is important for virulence in several gastrointestinal pathogens. However, its mechanism remains poorly understood. Hitherto, most studies regarding the Feo system were focused on Gammaproteobacterial models, which possess three feo genes (feoA, B, and C) clustered in an operon. We found that the human pathogen Helicobacter pylori possesses a unique arrangement of the feo genes, in which only feoA and feoB are present and encoded in distant loci. In this study, we examined the functional significance of this arrangement. Methods Requirement and regulation of the individual H. pylori feo genes were assessed through in vivo assays and gene expression profiling. The evolutionary history of feo was inferred via phylogenetic reconstruction, and AlphaFold was used for predicting the FeoA-FeoB interaction. Results and Discussion Both feoA and feoB are required for Feo function, and feoB is likely subjected to tight regulation in response to iron and nickel by Fur and NikR, respectively. Also, we established that feoA is encoded in an operon that emerged in the common ancestor of most, but not all, helicobacters, and this resulted in feoA transcription being controlled by two independent promoters. The H. pylori Feo system offers a new model to understand ferrous iron transport in bacterial pathogens.
Collapse
Affiliation(s)
- Camilo Gómez-Garzón
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
| | - Shelley M. Payne
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
- John Ring LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
46
|
Lear L, Hesse E, Newsome L, Gaze W, Buckling A, Vos M. The effect of metal remediation on the virulence and antimicrobial resistance of the opportunistic pathogen Pseudomonas aeruginosa. Evol Appl 2023; 16:1377-1389. [PMID: 37492145 PMCID: PMC10363854 DOI: 10.1111/eva.13576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 05/18/2023] [Accepted: 06/19/2023] [Indexed: 07/27/2023] Open
Abstract
Anthropogenic metal pollution can result in co-selection for antibiotic resistance and potentially select for increased virulence in bacterial pathogens. Metal-polluted environments can select for the increased production of siderophore molecules to detoxify non-ferrous metals. However, these same molecules also aid the uptake of ferric iron, a limiting factor for within-host pathogen growth, and are consequently a virulence factor. Anthropogenic methods to remediate environmental metal contamination commonly involve amendment with lime-containing materials. However, whether this reduces in situ co-selection for antibiotic resistance and siderophore-mediated virulence remains unknown. Here, using microcosms containing non-sterile metal-contaminated river water and sediment, we test whether liming reduces co-selection for these pathogenicity traits in the opportunistic pathogen Pseudomonas aeruginosa. To account for the effect of environmental structure, which is known to impact siderophore production, microcosms were incubated under either static or shaking conditions. Evolved P. aeruginosa populations had greater fitness in the presence of toxic concentrations of copper than the ancestral strain and showed increased resistance to the clinically relevant antibiotics apramycin, cefotaxime and trimethoprim, regardless of lime addition or environmental structure. Although we found virulence to be significantly associated with siderophore production, neither virulence nor siderophore production significantly differed between the four treatments. Furthermore, liming did not mitigate metal-imposed selection for antibiotic resistance or virulence in P. aeruginosa. Consequently, metal-contaminated environments may select for antibiotic resistance and virulence traits even when treated with lime.
Collapse
Affiliation(s)
- Luke Lear
- College of Life and Environmental ScienceUniversity of ExeterPenrynUK
| | - Elze Hesse
- College of Life and Environmental ScienceUniversity of ExeterPenrynUK
| | - Laura Newsome
- College of Engineering, Mathematics and Physical SciencesUniversity of ExeterPenrynUK
| | - William Gaze
- European Centre for Environment and Human HealthUniversity of Exeter Medical SchoolPenrynUK
| | - Angus Buckling
- College of Life and Environmental ScienceUniversity of ExeterPenrynUK
| | - Michiel Vos
- European Centre for Environment and Human HealthUniversity of Exeter Medical SchoolPenrynUK
| |
Collapse
|
47
|
Zhou Y, Zhou Z, Zheng L, Gong Z, Li Y, Jin Y, Huang Y, Chi M. Urinary Tract Infections Caused by Uropathogenic Escherichia coli: Mechanisms of Infection and Treatment Options. Int J Mol Sci 2023; 24:10537. [PMID: 37445714 DOI: 10.3390/ijms241310537] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Urinary tract infections (UTIs) are common bacterial infections that represent a severe public health problem. They are often caused by Escherichia coli (E. coli), Klebsiella pneumoniae (K. pneumonia), Proteus mirabilis (P. mirabilis), Enterococcus faecalis (E. faecalis), and Staphylococcus saprophyticus (S. saprophyticus). Among these, uropathogenic E. coli (UPEC) are the most common causative agent in both uncomplicated and complicated UTIs. The adaptive evolution of UPEC has been observed in several ways, including changes in colonization, attachment, invasion, and intracellular replication to invade the urothelium and survive intracellularly. While antibiotic therapy has historically been very successful in controlling UTIs, high recurrence rates and increasing antimicrobial resistance among uropathogens threaten to greatly reduce the efficacy of these treatments. Furthermore, the gradual global emergence of multidrug-resistant UPEC has highlighted the need to further explore its pathogenesis and seek alternative therapeutic and preventative strategies. Therefore, a thorough understanding of the clinical status and pathogenesis of UTIs and the advantages and disadvantages of antibiotics as a conventional treatment option could spark a surge in the search for alternative treatment options, especially vaccines and medicinal plants. Such options targeting multiple pathogenic mechanisms of UPEC are expected to be a focus of UTI management in the future to help combat antibiotic resistance.
Collapse
Affiliation(s)
- Yang Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, 4 Beijing Road, Guiyang 550004, China
- School of Pharmaceutical Sciences, Guizhou Medical University, 4 Beijing Road, Guiyang 550004, China
| | - Zuying Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, 4 Beijing Road, Guiyang 550004, China
- School of Pharmaceutical Sciences, Guizhou Medical University, 4 Beijing Road, Guiyang 550004, China
| | - Lin Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, 4 Beijing Road, Guiyang 550004, China
- School of Pharmaceutical Sciences, Guizhou Medical University, 4 Beijing Road, Guiyang 550004, China
| | - Zipeng Gong
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, 4 Beijing Road, Guiyang 550004, China
| | - Yueting Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, 4 Beijing Road, Guiyang 550004, China
| | - Yang Jin
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, 4 Beijing Road, Guiyang 550004, China
| | - Yong Huang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, 4 Beijing Road, Guiyang 550004, China
- School of Pharmaceutical Sciences, Guizhou Medical University, 4 Beijing Road, Guiyang 550004, China
| | - Mingyan Chi
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, 4 Beijing Road, Guiyang 550004, China
- School of Pharmaceutical Sciences, Guizhou Medical University, 4 Beijing Road, Guiyang 550004, China
| |
Collapse
|
48
|
Yu S, Li L, Zhao H, Liu M, Jiang L, Zhao Y. Citrus flavonoid extracts alter the profiling of rumen antibiotic resistance genes and virulence factors of dairy cows. Front Microbiol 2023; 14:1201262. [PMID: 37362928 PMCID: PMC10289158 DOI: 10.3389/fmicb.2023.1201262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Citrus flavonoid extracts (CFE) have the potential to reduce rumen inflammation, improve ruminal function, and enhance production performance in ruminants. Our previous studies have investigated the effects of CFE on the structure and function of rumen microbiota in dairy cows. However, it remains unclear whether CFE affects the prevalence of antibiotic resistance genes (ARG) and virulence factors genes (VFG) in the rumen. Therefore, metagenomics was used to identify the rumen ARG and VFG in lactating dairy cows fed with CFE diets. The results showed that CFE significantly reduced the levels of Multidrug and Antiphagocytosis in the rumen (p < 0.05) and increased the levels of Tetracycline, Iron uptake system, and Magnesium uptake system (p < 0.05). Furthermore, the changes were found to have associations with the phylum Lentisphaerae. It was concluded that CFE could be utilized as a natural plant product to regulate virulence factors and antibiotic resistance of rumen microbiota, thereby improving rumen homeostasis and the health of dairy cows.
Collapse
Affiliation(s)
- Shiqiang Yu
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Liuxue Li
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Huiying Zhao
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Ming Liu
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Linshu Jiang
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Yuchao Zhao
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
- Beijing Beinong Enterprise Management Co., Ltd., Beijing, China
| |
Collapse
|
49
|
Rabbee MF, Baek KH. Detection of Antagonistic Compounds Synthesized by Bacillus velezensis against Xanthomonas citri subsp. citri by Metabolome and RNA Sequencing. Microorganisms 2023; 11:1523. [PMID: 37375024 DOI: 10.3390/microorganisms11061523] [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: 03/29/2023] [Revised: 05/17/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Biological control of plant diseases has gained attraction for controlling various bacterial diseases at a field trial stage. An isolated endophytic bacterium, Bacillus velezensis 25 (Bv-25), from Citrus species had strong antagonistic activity against Xanthomonas citri subsp. citri (Xcc), which causes citrus canker disease. When Bv-25 was incubated in Landy broth or yeast nutrient broth (YNB), the ethyl acetate extract of Landy broth exhibited higher levels of antagonistic activity against Xcc compared to that of YNB. Therefore, the antimicrobial compounds in the two ethyl acetate extracts were detected by high performance liquid chromatography-mass spectrometry. This comparison revealed an increase in production of several antimicrobial compounds, including difficidin, surfactin, fengycin, and Iturin-A or bacillomycin-D by incubation in Landy broth. RNA sequencing for the Bv-25 grown in Landy broth were performed, and the differential expressions were detected for the genes encoding the enzymes for the synthesis of antimicrobial compounds, such as bacilysin, plipastatin or fengycin, surfactin, and mycosubtilin. Combination of metabolomics analysis and RNA sequencing strongly suggests that several antagonistic compounds, especially bacilysin produced by B. velezensis, exhibit an antagonistic effect against Xcc.
Collapse
Affiliation(s)
- Muhammad Fazle Rabbee
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
50
|
Sun S, Gao Y, Yang X, Yang X, Hu T, Liang J, Xiong Z, Ran Y, Ren P, Bai F, Guddat LW, Yang H, Rao Z, Zhang B. Cryo-EM structures for the Mycobacterium tuberculosis iron-loaded siderophore transporter IrtAB. Protein Cell 2023; 14:448-458. [PMID: 36882106 PMCID: PMC10246721 DOI: 10.1093/procel/pwac060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/23/2022] [Indexed: 11/18/2022] Open
Abstract
The adenosine 5'-triphosphate (ATP)-binding cassette (ABC) transporter, IrtAB, plays a vital role in the replication and viability of Mycobacterium tuberculosis (Mtb), where its function is to import iron-loaded siderophores. Unusually, it adopts the canonical type IV exporter fold. Herein, we report the structure of unliganded Mtb IrtAB and its structure in complex with ATP, ADP, or ATP analogue (AMP-PNP) at resolutions ranging from 2.8 to 3.5 Å. The structure of IrtAB bound ATP-Mg2+ shows a "head-to-tail" dimer of nucleotide-binding domains (NBDs), a closed amphipathic cavity within the transmembrane domains (TMDs), and a metal ion liganded to three histidine residues of IrtA in the cavity. Cryo-electron microscopy (Cryo-EM) structures and ATP hydrolysis assays show that the NBD of IrtA has a higher affinity for nucleotides and increased ATPase activity compared with IrtB. Moreover, the metal ion located in the TM region of IrtA is critical for the stabilization of the conformation of IrtAB during the transport cycle. This study provides a structural basis to explain the ATP-driven conformational changes that occur in IrtAB.
Collapse
Affiliation(s)
- Shan Sun
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Gao
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaolin Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiuna Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Tianyu Hu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jingxi Liang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300353, China
| | - Zhiqi Xiong
- Laboratory of Structural Biology, Tsinghua University, Beijing 100084, China
| | - Yuting Ran
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Pengxuan Ren
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Fang Bai
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Luke W Guddat
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Haitao Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Zihe Rao
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300353, China
- Laboratory of Structural Biology, Tsinghua University, Beijing 100084, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Bing Zhang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| |
Collapse
|