1
|
Jiang K, Pang X, Li W, Xu X, Yang Y, Shang C, Gao X. Interbacterial warfare in the human gut: insights from Bacteroidales' perspective. Gut Microbes 2025; 17:2473522. [PMID: 40038576 PMCID: PMC11901371 DOI: 10.1080/19490976.2025.2473522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/19/2025] [Accepted: 02/21/2025] [Indexed: 03/06/2025] Open
Abstract
Competition and cooperation are fundamental to the stability and evolution of ecological communities. The human gut microbiota, a dense and complex microbial ecosystem, plays a critical role in the host's health and disease, with competitive interactions being particularly significant. As a dominant and extensively studied group in the human gut, Bacteroidales serves as a successful model system for understanding these intricate dynamic processes. This review summarizes recent advances in our understanding of the intricate antagonism mechanisms among gut Bacteroidales at the biochemical or molecular-genetic levels, focusing on interference and exploitation competition. We also discuss unresolved questions and suggest strategies for studying the competitive mechanisms of Bacteroidales. The review presented here offers valuable insights into the molecular basis of bacterial antagonism in the human gut and may inform strategies for manipulating the microbiome to benefit human health.
Collapse
Affiliation(s)
- Kun Jiang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xinxin Pang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Weixun Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xiaoning Xu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Yan Yang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Chengbin Shang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xiang Gao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| |
Collapse
|
2
|
Shao J, Wang X, Liu Q, Lv H, Qi Q, Li C, Zhang J, Chen X, Chen X. Eucommia ulmoides leaf extracts combined with Astragalus polysaccharides: Effects on growth, antioxidant capacity, and intestinal inflammation in juvenile large yellow croaker (Larimichthys crocea). FISH & SHELLFISH IMMUNOLOGY 2025; 161:110229. [PMID: 40015491 DOI: 10.1016/j.fsi.2025.110229] [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: 12/04/2024] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/01/2025]
Abstract
Eucommia ulmoides leaf extract (ELE) and Astragalus polysaccharides (APS) have been widely used as immunopotentiators in aquaculture. Our prior research on large yellow croaker (Larimichthys crocea) demonstrated that dietary 1 g/kg APS bolstered fish immunity and antioxidant defense. However, the combined effect of ELE and APS in juvenile large yellow croaker remains unknown. Hence, this study aimed to investigate the synergistic effect of ELE and APS on the growth, antioxidant capacity, and intestinal inflammation in large yellow croaker. A total of 1200 fish were divided into five groups and fed diets with 1 g/kg APS and varying ELE levels: 0 g/kg (ELE0), 0.25 g/kg (ELE0.25), 0.5 g/kg (ELE0.5), 1 g/kg (ELE1), and 2 g/kg (ELE2). After an 8-week feeding period, the ELE0.5 and ELE1 groups showed superior weight gain rate, specific growth rate, and feed efficiency compared to other groups. The ELE1 group also had elevated trypsin and lipase activities in the intestine, whereas α-amylase activity was not influenced by ELE addition. Antioxidant enzyme activities, such as hepatopancreas superoxide dismutase (SOD) and glutathione peroxidase (GPX) in the ELE1 group were significantly enhanced, while malondialdehyde (MDA) levels decreased with increasing ELE. Intestinal morphology revealed the highest villi height in proximal and distal intestines of ELE1 group, with no significant change in mucosal thickness. In terms of cytokines, the ELE1 group showed significant down-regulation of pro-inflammatory (tnf-α, il-1β and il-6) and up-regulation of anti-inflammatory (il-4/13a, il-10 and tgf-β) markers, modulated by MAPK and mTOR signaling. In conclusion, this study indicates that supplementing diets with 1 g/kg ELE alongside 1 g/kg APS in juvenile large yellow croaker offers the best synergistic effect on fish immunity, including enhanced growth, antioxidant capacity, and relieved intestinal inflammation through MAPK and mTOR signaling.
Collapse
Affiliation(s)
- Jianchun Shao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Fuzhou Institute of Oceanography, Fuzhou, 350108, China
| | - Xuexi Wang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Fuzhou Institute of Oceanography, Fuzhou, 350108, China
| | - Qianqian Liu
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Huiyuan Lv
- Beijing Engineering Technology Research Center for Traditional Chinese Veterinary Medicine, Beijing Centre Biology Co., Ltd., Beijing, 100000, China
| | - Qiong Qi
- Beijing Engineering Technology Research Center for Traditional Chinese Veterinary Medicine, Beijing Centre Biology Co., Ltd., Beijing, 100000, China
| | - Changhui Li
- Key Laboratory of Special Aquatic Formula Feed of Fujian Province, Fujian Tianma Science and Technology Group Co., Ltd., Fuzhou, 350002, China
| | - Jiaonan Zhang
- Key Laboratory of Special Aquatic Formula Feed of Fujian Province, Fujian Tianma Science and Technology Group Co., Ltd., Fuzhou, 350002, China
| | - Xiaojuan Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Fuzhou Institute of Oceanography, Fuzhou, 350108, China.
| | - Xinhua Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Fuzhou Institute of Oceanography, Fuzhou, 350108, China.
| |
Collapse
|
3
|
Zhou L, Mao HQ, Wen YH, Chen Z, Zhang L. Cuproptosis Aggravates Pulpitis by Inhibiting the Pentose Phosphate Pathway. J Dent Res 2025; 104:541-550. [PMID: 39953718 DOI: 10.1177/00220345251313797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2025] Open
Abstract
Excessive copper becomes toxic, driving inflammation, and, when copper exceeds a certain threshold, it even leads to a novel programmed cell death termed cuproptosis. However, disordered copper metabolism and its mechanism in pulpitis remain unclear. In this work, we found that lipoteichoic acid (LTA) or lipopolysaccharides (LPS) triggered copper deposition in pulpitis and consequently intensified cuproptosis by impeding the pentose phosphate pathway (PPP). We initially assessed the copper content in pulpitis tissues via inductively coupled plasma mass spectrometry and observed significantly greater concentrations than in healthy pulp tissues. We found that a relatively high copper content was triggered by LTA or LPS, leading cells to cuproptosis. Stimulation of LTA or LPS induced copper deposition and cuproptosis, worsening the progression of pulpitis in vivo. Mechanistically, we found that copper detoxification is dependent on the PPP. We used a 13C-glucose stable isotope-tracing experiment to assess the effect of glucose utilization on cuproptosis. Excessive copper hindered the PPP, resulting in an inadequate generation of nicotinamide adenine dinucleotide phosphate to replenish glutathione and counteract copper toxicity. The PPP regulates the phenotype, function, and survival of preodontoblast-like cells in cuproptosis. Our findings revealed the intricate interplay among bacteria, copper homeostasis, and metabolic reprogramming, providing potential strategies for host-targeted therapy in pulpitis.
Collapse
Affiliation(s)
- L Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Cariology and Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - H-Q Mao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Y-H Wen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Z Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Cariology and Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - L Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Cariology and Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
4
|
Yang MC, Huang WL, Chen HY, Lin SH, Chang YS, Tseng KY, Lo HJ, Wang IC, Lin CJ, Lan CY. Deletion of RAP1 affects iron homeostasis, azole resistance, and virulence in Candida albicans. mSphere 2025:e0015525. [PMID: 40265929 DOI: 10.1128/msphere.00155-25] [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: 03/11/2025] [Accepted: 03/29/2025] [Indexed: 04/24/2025] Open
Abstract
Rap1 is a DNA-binding protein conserved from yeast to mammals for its role in telomeric maintenance. Here, to explore additional functions of Candida albicans Rap1, we performed RNA sequencing analysis. Experimental validations further showed that Rap1 plays a role in iron regulation, especially under low-iron conditions. Moreover, Rap1 was involved in iron acquisition and modulation of iron-related genes. Rap1 was found to be associated with fluconazole resistance in a low-iron condition. Finally, we demonstrated that the deletion of RAP1 leads to reduced C. albicans virulence in a mouse model of infection. Together, this study reveals new functions of C. albicans Rap1, particularly in iron homeostasis, azole resistance, and virulence. IMPORTANCE Candida albicans is an important pathogenic fungus that can cause superficial to life-threatening infections. Iron is essential for almost all organisms, yet it is highly restricted within the human host to defend against pathogens. To grow and survive in the iron-limited host environment, C. albicans has evolved multiple iron acquisition mechanisms. Understanding the regulation of iron homeostasis is, therefore, critical for elucidating C. albicans pathogenesis and virulence. This study explores the novel functions of C. albicans Rap1, with a focus on its contribution to iron acquisition and utilization. Our findings further highlight how iron availability impacts antifungal resistance and virulence through Rap1, providing insight into the complex iron regulatory machinery of C. albicans.
Collapse
Affiliation(s)
- Min-Chi Yang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Wei-Luen Huang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsuan-Yu Chen
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Shin-Huey Lin
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Shan Chang
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Kuo-Yun Tseng
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
- Taiwan Mycology Reference Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Hsiu-Jung Lo
- Taiwan Mycology Reference Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - I-Ching Wang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Chi-Jan Lin
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Chung-Yu Lan
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
5
|
Yokoyama T, Miyazaki R, Suzuki T, Dohmae N, Nagai H, Tsukazaki T, Kubori T, Akiyama Y. Cleavage cascade of the sigma regulator FecR orchestrates TonB-dependent signal transduction. Proc Natl Acad Sci U S A 2025; 122:e2500366122. [PMID: 40244679 DOI: 10.1073/pnas.2500366122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
TonB-dependent signal transduction is a versatile mechanism observed in gram-negative bacteria that integrates energy-dependent substrate transport with signal relay. In Escherichia coli, the TonB-ExbBD motor complex energizes the TonB-dependent outer membrane transporter FecA, facilitating ferric citrate import. FecA also acts as a sensor, transmitting signals to the cytoplasmic membrane protein FecR, which eventually activates the cytoplasmic sigma factor FecI, driving transcription of the fec operon. Building on our previous finding that FecR undergoes functional maturation through a three-step cleavage process [T. Yokoyama et al., J. Biol. Chem. 296, 100673 (2021)], we here describe the complete mechanism of FecR-mediated ferric citrate signaling involving FecA and TonB. The cleavage cascade begins with FecR autoproteolysis prior to membrane integration. The soluble C-terminal domain (CTD) fragment of FecR is cotranslocated with the N-terminal domain (NTD) fragment through a twin-arginine translocation (Tat) system-mediated process. In the periplasm, the interaction between the CTD and NTD fragments prevents further cleavage. Binding of ferric citrate induces a conformational change in FecA, exposing its TonB box to the periplasmic space. This structural alteration is transmitted to the interacting FecR CTD via the motor function of TonB, resulting in the release of the CTD blockage from the NTD. Consequently, the successive cleavage of FecR's NTD is initiated, culminating in the ferric citrate signal-induced activation of fec gene expression. Our findings reveal that the regulation of FecR cleavage, controlled by the TonB-FecA axis, plays a central role in the bacterial response to ferric citrate signals.
Collapse
Affiliation(s)
- Tatsuhiko Yokoyama
- Department of Microbiology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
| | - Ryoji Miyazaki
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Takehiro Suzuki
- Biomolecular Characterization Unit, Technology Platform Division, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Naoshi Dohmae
- Biomolecular Characterization Unit, Technology Platform Division, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Hiroki Nagai
- Department of Microbiology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
- Center for One Medicine Innovative Translational Research, Institute for Advanced Study, Gifu University, Gifu 501-1194, Japan
| | - Tomoya Tsukazaki
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Tomoko Kubori
- Department of Microbiology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
| | - Yoshinori Akiyama
- Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
6
|
Weiner JM, Lee WH, Nolan EM, Oglesby AG. Calprotectin elicits aberrant iron starvation responses in Pseudomonas aeruginosa under anaerobic conditions. J Bacteriol 2025; 207:e0002925. [PMID: 40135923 PMCID: PMC12004955 DOI: 10.1128/jb.00029-25] [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: 01/24/2025] [Accepted: 01/31/2025] [Indexed: 03/27/2025] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that uses several mechanisms to survive in the iron-limiting host environment. The innate immune protein calprotectin (CP) sequesters ferrous iron [Fe(II)], among other divalent transition metal ions, to limit its availability to pathogens. CP levels are increased in individuals with cystic fibrosis (CF), a hereditary disease that leads to chronic pulmonary infection by P. aeruginosa. We previously showed that aerobic CP treatment of P. aeruginosa induces a multi-metal starvation response that alters expression of several virulence properties. However, the CF lung is a hypoxic environment due to the growth of P. aeruginosa in dense biofilms. Here, we report that anaerobic CP treatment of P. aeruginosa induces many processes associated with an aerobic iron starvation response, including decreased phenazine production and increased expression of the PrrF small regulatory RNAs (sRNAs). However, the iron starvation response elicited by CP in anaerobic conditions shows characteristics that are distinct from responses observed in aerobic growth, including a lack of siderophore production and increased induction of genes for the FeoAB Fe(II) and Phu heme uptake systems. Also distinct from aerobic conditions, CP treatment induces expression of genes for predicted manganese transporters MntH1 and MntH2 during anaerobic growth while eliciting a less robust zinc starvation response compared to aerobic conditions. Induction of mntH2 is dependent on the PrrF sRNAs, suggesting a novel example of metal regulatory cross-talk. Thus, anaerobic CP treatment results in a multi-metal starvation response with key distinctions from aerobic conditions, revealing differences in P. aeruginosa metal homeostasis during anaerobic growth.IMPORTANCEIron is critical for most microbial pathogens, and the innate immune system sequesters this metal to limit microbial growth. Pathogens must overcome iron sequestration to survive during infection. For many pathogens, iron homeostasis has primarily been studied in aerobic conditions. Nevertheless, some host environments are hypoxic, including chronic lung infection sites in individuals with cystic fibrosis (CF). Here, we use the innate immune protein calprotectin, which sequesters divalent metal ions including Fe(II), to study the anaerobic iron starvation response of a common CF lung pathogen, Pseudomonas aeruginosa. We report several distinctions of this response during anaerobiosis, highlighting the importance of carefully considering the host environment when investigating the role of nutritional immunity in host-pathogen interactions.
Collapse
Affiliation(s)
- Jacob M. Weiner
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Wei Hao Lee
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Elizabeth M. Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Amanda G. Oglesby
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Freeman MJ, Eral NJ, Sauer JD. Listeria monocytogenes requires phosphotransferase systems to facilitate intracellular growth and virulence. PLoS Pathog 2025; 21:e1012492. [PMID: 40233105 DOI: 10.1371/journal.ppat.1012492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 03/27/2025] [Indexed: 04/17/2025] Open
Abstract
The metabolism of bacterial pathogens is exquisitely evolved to support virulence in the nutrient-limiting host. Many bacterial pathogens utilize bipartite metabolism to support intracellular growth by splitting carbon utilization between two carbon sources and dividing flux to distinct metabolic needs. For example, previous studies suggest that the professional cytosolic pathogen Listeria monocytogenes (L. monocytogenes) utilizes glycerol and hexose phosphates (e.g., Glucose-6-Phosphate) as catabolic and anabolic carbon sources in the host cytosol, respectively. However, the role of this putative bipartite metabolism in L. monocytogenes virulence has not been fully assessed. Here, we demonstrate that when L. monocytogenes is unable to consume either glycerol (ΔglpD/ΔgolD), hexose phosphates (ΔuhpT), or both (ΔglpD/ΔgolD/ΔuhpT), it is still able to grow in the host cytosol and is 10- to 100-fold attenuated in vivo suggesting that L. monocytogenes consumes alternative carbon source(s) in the host. An in vitro metabolic screen using BioLog's phenotypic microarrays unexpectedly demonstrated that WT and PrfA* (G145S) L. monocytogenes, a strain with constitutive virulence gene expression, use phosphotransferase system (PTS) mediated carbon sources. These findings contrast with the existing metabolic model that cytosolic L. monocytogenes expressing PrfA does not use PTS mediated carbon sources. We next demonstrate that two independent and universal phosphocarrier proteins (PtsI [EI] and PtsH [HPr]), essential for the function of all PTS, are critical for intracellular growth and virulence in vivo. Constitutive virulence gene expression using a PrfA* (G145S) allele in ΔglpD/ΔgolD/ΔuhpT and ΔptsI failed to rescue in vivo virulence defects suggesting phenotypes are due to metabolic disruption and not virulence gene regulation. Finally, in vivo attenuation of ΔptsI and ΔptsH was additive to ΔglpD/ΔgolD/ΔuhpT, suggesting that hexose phosphates and glycerol and PTS mediated carbon source are relevant metabolites. Taken together, these studies indicate that PTS are critical virulence factors for the cytosolic growth and virulence of L. monocytogenes.
Collapse
Affiliation(s)
- Matthew J Freeman
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Noah J Eral
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
8
|
Wiebe M, Ingebritson A, Sholeh M, Tichenor C, Visek C, Victoria J, Beck M, Tiwari R, Hardwidge P, Zhu L. Streptococcus suis manganese transporter mutant as a live attenuated vaccine: Safety, efficacy, and virulence reversion mechanisms. Vet Microbiol 2025; 305:110521. [PMID: 40239440 DOI: 10.1016/j.vetmic.2025.110521] [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: 02/24/2025] [Revised: 04/02/2025] [Accepted: 04/11/2025] [Indexed: 04/18/2025]
Abstract
Streptococcus suis is the leading cause of mortality in piglets and is responsible for severe economic losses in the global pork industry. Severe invasive diseases caused by S. suis include sepsis, meningitis, arthritis, and endocarditis. S. suis disease prevention is hampered by the lack of safe and efficacious vaccines. In this study, we constructed an S. suis live attenuated vaccine candidate lacking the major streptococcal manganese transporter, a known virulence determinant of this organism. The safety and efficacy of this live vaccine were evaluated in swine. Our clinical study results showed that when administered at a dose of 1010 CFU, the vaccine strain was safe and efficacious. However, a lower dose of 109 CFU failed to generate significant immune protection. To investigate if an adjuvant could enhance the efficacy of the vaccine at a lower dose, we spiked the vaccine with a polymeric adjuvant and evaluated its performance. Surprisingly, four pigs receiving the adjuvanted vaccine died during the vaccination phase. Pathology, microbiology, and genetic analyses suggested that the vaccine strain reverted to virulence in these animals. Functional genetic analysis found that the vaccine strain acquired compensatory mutations that upregulated the expression of a secondary manganese transporter, which in turn restored the virulence of the vaccine strain. Our results provide a new understanding of S. suis host adaptation mechanisms and useful information for the design of future live-attenuated vaccines.
Collapse
Affiliation(s)
- Michelle Wiebe
- Boehringer Ingelheim Animal Health USA, Inc., Ames, IA, USA
| | | | - Melody Sholeh
- Boehringer Ingelheim Animal Health USA, Inc., Ames, IA, USA
| | | | - Callie Visek
- Boehringer Ingelheim Animal Health USA, Inc., Ames, IA, USA
| | | | - Michael Beck
- Boehringer Ingelheim Animal Health USA, Inc., Ames, IA, USA
| | - Raksha Tiwari
- Boehringer Ingelheim Animal Health USA, Inc., Ames, IA, USA
| | | | - Luchang Zhu
- Boehringer Ingelheim Animal Health USA, Inc., Ames, IA, USA.
| |
Collapse
|
9
|
Yang R, Han S, Yu Y, Li H, Helmann JD, Schaufler K, Johnson MDL, Yang QE, Rensing C. The Klebsiella pneumoniae tellurium resistance gene terC contributes to both tellurite and zinc resistance. Microbiol Spectr 2025:e0263424. [PMID: 40202338 DOI: 10.1128/spectrum.02634-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
Klebsiella pneumoniae is widely recognized as a pathogen responsible for hospital-acquired infections and community-acquired invasive infections. It has rapidly become a significant global public health threat due to the emergence of hypervirulent and multidrug-resistant strains, which have increased the challenges associated with treating life-threatening infections. Tellurium resistance genes are widespread on virulence plasmids in K. pneumoniae isolates. However, the core function of the ter operon (terZABCDEF) in K. pneumoniae remains unclear. In this study, the multidrug-resistant K. pneumoniae P1927 strain was isolated from the sputum of a hospitalized pneumonia patient. The ter operon, along with antimicrobial resistance and virulence genes, was identified on a large hybrid plasmid in K. pneumoniae P1927. We generated a terC deletion mutant and demonstrated that this mutant exhibited reduced virulence in a Galleria mellonella larva infection model. Further physiological functional analysis revealed that terC is not only important for Te(IV) resistance but also for resistance to Zn(II), Mn(II), and phage infection. All genes of the ter operon were highly inducible by Zn(II), which is a stronger inducer than Te(IV), and the terBCDE genes were also induced by Mn(II). Collectively, our study demonstrates novel physiological functions of TerC in Zn(II) resistance and virulence in K. pneumoniae.IMPORTANCEKlebsiella pneumoniae has rapidly become a global threat to public health. Although the ter operon is widely identified in clinical isolates, its physiological function remains unclear. It has been proposed that proteins encoded by the ter operon form a multi-site metal-binding complex, but its exact function is still unknown. TerC, a central component of the tellurium resistance determinant, was previously shown to interact with outer membrane proteins OmpA and KpsD in Escherichia coli, suggesting potential changes in outer membrane structure and properties. Here, we report that TerC confers resistance to Zn(II), Mn(II), and phage infection, and Zn(II) was shown to be a strong inducer of the ter operon. Furthermore, TerC was identified as a novel virulence factor. Taken together, our results expand our understanding of the physiological functions encoded by the ter operon and its role in the virulence of K. pneumoniae, providing deeper insights into the link between heavy metal(loid) resistance determinants and virulence in pathogenic bacteria.
Collapse
Affiliation(s)
- Ruixiang Yang
- Institute of Environmental Microbiology, College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Shuang Han
- Fujian Provincial Key Laboratory of Soil Environmental Health and Regulation, College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Yanshuang Yu
- Institute of Environmental Microbiology, College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Soil Environmental Health and Regulation, College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Hongru Li
- Fujian Provincial Key Laboratory of Medical Big Data Engineering, Fujian Provincial Hospital, Shengli Clinical College of Fujian Medical University, Fuzhou, Fujian, China
- Department of Respiratory and Critical Care Medicine, Fujian Shengli Medical College, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - John D Helmann
- Department of Microbiology, Cornell University, Ithaca, New York, USA
| | - Katharina Schaufler
- Department of Epidemiology and Ecology of Antimicrobial Resistance, Helmholtz Centre for Infection Research HZI, Helmholtz Institute for One Health, Greifswald, Mecklenburg-Vorpommern, Germany
| | - Michael D L Johnson
- Department of Immunobiology, The University of Arizona College of Medicine Tucson, Tucson, Arizona, USA
| | - Qiu E Yang
- Fujian Provincial Key Laboratory of Soil Environmental Health and Regulation, College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Christopher Rensing
- Institute of Environmental Microbiology, College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| |
Collapse
|
10
|
Garg R, Zhu Z, Hernandez FG, Wang Y, David MS, Bruno VM, Culotta VC. A response to iron involving carbon metabolism in the opportunistic fungal pathogen Candida albicans. mSphere 2025:e0004025. [PMID: 40183578 DOI: 10.1128/msphere.00040-25] [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: 01/16/2025] [Accepted: 03/09/2025] [Indexed: 04/05/2025] Open
Abstract
Iron (Fe) is an essential micronutrient, and during infection, the host attempts to starve pathogens of this vital element through a process known as nutritional immunity. Successful pathogens have evolved means to evade this attack, an example being Candida albicans, the most prevalent human fungal pathogen. When Fe-starved, C. albicans induces multiple pathways for Fe uptake using the SEF1 trans-regulator, and we now describe a previously unrecognized effect of Fe on C. albicans metabolism that occurs independent of SEF1. Specifically, Fe limitation leads to inhibition of pyruvate dehydrogenase (PDH) connecting glycolysis to mitochondrial respiration. PDH inactivation involves loss of the LAT1 catalytic subunit harboring a lipoic acid co-factor. Protein lipoylation is a Fe-S dependent process, and lipoylated alpha-ketoglutarate dehydrogenase is also inhibited in Fe-starved C. albicans. SEF1 does not protect against PDH inactivation, and despite SEF1 induction of Fe import genes, cellular Fe levels drop dramatically during chronic Fe starvation. Such loss of LAT1 and lipoylation is also seen in Fe-starved bakers' yeast Saccharomyces cerevisiae. In both yeast species, glucose is diverted toward the pentose phosphate pathway (PPP) and PPP production of NADPH is increased in response to low Fe and PDH loss. Additionally, glucose consumption is lowered in Fe-starved C. albicans, and non-PDH alternatives to producing Ac-CoA are induced, including pyruvate bypass and fatty acid oxidation pathways. C. albicans can adapt well to the effects of micronutrient loss on cell metabolism. IMPORTANCE We describe a new response to Fe-starvation in a fungal pathogen involving carbon metabolism. Pyruvate dehydrogenase (PDH) that is central to glucose metabolism is inactivated at the post-translational level in Fe-starved cells. Nevertheless, the fungal pathogen can thrive by activating backup systems for metabolizing glucose. Methods that inhibit these compensatory pathways for carbon metabolism may prove beneficial in future anti-fungal strategies.
Collapse
Affiliation(s)
- Ritu Garg
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Zhengkai Zhu
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Francisco G Hernandez
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Yiran Wang
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Marika S David
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Vincent M Bruno
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Valeria C Culotta
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
11
|
Derbise A, Guillas C, Echenique-Rivera H, Carniel E, Gerke C, Pizarro-Cerdá J, Demeure CE. Contribution of the type 3 secretion system to adaptive and innate immunity induced by a live Yersinia pseudotuberculosis plague vaccine. Vaccine 2025; 51:126887. [PMID: 39978224 DOI: 10.1016/j.vaccine.2025.126887] [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: 09/05/2024] [Revised: 01/25/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
Yersinia pestis, the causative agent of plague, remains a threat to public health worldwide. From the perspective of developing safe and effective vaccines, we present a derived version of our Y. pseudotuberculosis VTnF1 live attenuated vaccine candidate that lacks the pYV virulence plasmid coding for the Type 3 Secretion system (T3SS) and carries no antibiotic resistance cassettes (VTnF1-S). This strain, named VpYV-, fails to cause disease in immunocompromised mice when given orally, and can be considered as avirulent in such conditions. It retains a tropism for Peyer's patches and mesenteric lymph nodes, whilst rarely reaching the spleen and liver. When compared to VTnF1-S, VpYV- elicited equivalent production of IgG directed to the F1 antigen, but less IgG directed to other Yersinia antigens. A single oral dose of VpYV- induced 100 % protection against bubonic and pneumonic forms of plague. Four months after vaccination, the protection induced by VpYV- had decreased more than that induced by VTnF1-S. Furthermore, VpYV- was 30 % less protective against F1-negative Y. pestis, revealing that the T3SS components encoded by pYV are mandatory to obtain a large spectrum protection. Finally, VTnF1-S and VpYV- were compared for their ability to induce immediate immune activity against co-infecting Y. pestis, which could be a potential therapeutic strategy against early-stage infections. Like the historical Y. pestis vaccine EV76, VTnF1-S was able to induce such a protection. The process involved nutritional immunity in serum, indicating a fast activation of innate immune mechanisms. By contrast, VpYV- failed to protect mice, revealing an importance of the T3SS in this mechanism. Overall, VTnF1 and its derivative strain VpYV-, offer a choice between better vaccine performance or greater vaccine safety. They represent useful tools to prevent and treat Y. pestis infection in healthy or immunocompromised individuals.
Collapse
Affiliation(s)
- Anne Derbise
- Institut Pasteur, Unité de recherche Yersinia,; Université Paris Cité; CNRS UMR6047, F-75015, Paris, France; Institut Pasteur, Université Paris Cité, WHO Collaborating Research and Reference Center for Plague FRA-146, F-75015, Paris, France
| | - Chloé Guillas
- Institut Pasteur, Unité de recherche Yersinia,; Université Paris Cité; CNRS UMR6047, F-75015, Paris, France
| | - Hebert Echenique-Rivera
- Institut Pasteur, Unité de recherche Yersinia,; Université Paris Cité; CNRS UMR6047, F-75015, Paris, France; Institut Pasteur, Université Paris Cité, WHO Collaborating Research and Reference Center for Plague FRA-146, F-75015, Paris, France
| | - Elisabeth Carniel
- Institut Pasteur, Unité de recherche Yersinia,; Université Paris Cité; CNRS UMR6047, F-75015, Paris, France; Institut Pasteur, Université Paris Cité, WHO Collaborating Research and Reference Center for Plague FRA-146, F-75015, Paris, France; Institut Pasteur, Université Paris Cité, Yersinia National Reference Laboratory, F-75015, Paris, France
| | - Chris Gerke
- Institut Pasteur, Innovation Office, Vaccine Programs, F-75015, Paris, France
| | - Javier Pizarro-Cerdá
- Institut Pasteur, Unité de recherche Yersinia,; Université Paris Cité; CNRS UMR6047, F-75015, Paris, France; Institut Pasteur, Université Paris Cité, WHO Collaborating Research and Reference Center for Plague FRA-146, F-75015, Paris, France; Institut Pasteur, Université Paris Cité, Yersinia National Reference Laboratory, F-75015, Paris, France
| | - Christian E Demeure
- Institut Pasteur, Unité de recherche Yersinia,; Université Paris Cité; CNRS UMR6047, F-75015, Paris, France; Institut Pasteur, Université Paris Cité, WHO Collaborating Research and Reference Center for Plague FRA-146, F-75015, Paris, France.
| |
Collapse
|
12
|
Anne S, McDonald MR, Lu Y, Peterson RL. Pseudogymnoascus destructans transcriptional response to chronic copper stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.646060. [PMID: 40236230 PMCID: PMC11996344 DOI: 10.1101/2025.03.28.646060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Copper (Cu) is an essential metal micronutrient, and a fungal pathogens' ability to thrive in diverse niches across a broad range of bioavailable copper levels is vital for host-colonization and fungal-propagation. Recent transcriptomic studies have implemented that trace metal acquisition is important for the propagation of the white nose syndrome (WNS) causing fungus, Pseudogymnoascus destructans , on bat hosts. This report characterizes the P. destructans transcriptional response to Cu-withholding and Cu-overload stress. We identify 583 differently expressed genes (DEGs) that respond to Cu-withholding stress and 667 DEGs that respond to Cu-overload stress. We find that the P. destructans Cu-transporter genes CTR 1a and CTR1 b, as well as two homologs to Cryptococcus neoformans Cbi1/BIM1 VC83_03095 (BLP2) and VC83_07867 (BLP3) are highly regulated by Cu-withholding stress. We identify a cluster of genes, VC83_01834 - VC83_01837, that are regulated by copper bioavailability, which we identify as the Cu Responsive gene Cluster (CRC). We find that chronic exposure to elevated copper levels leads to an increase in genes associated with DNA repair and DNA replication fidelity. A comparison of our transcriptomic data sets with P. destructans at WNS fungal infection sites reveals several putative fungal virulence factors that respond to environmental copper stress.
Collapse
|
13
|
Sheneman KR, Cummins TD, Merchant ML, Hood JL, Uriarte SM, Lawrenz MB. Yersinia pestis Actively Inhibits the Production of Extracellular Vesicles by Human Neutrophils. J Extracell Vesicles 2025; 14:e70074. [PMID: 40240908 PMCID: PMC12003101 DOI: 10.1002/jev2.70074] [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: 12/17/2024] [Accepted: 03/23/2025] [Indexed: 04/18/2025] Open
Abstract
Yersinia pestis is the etiologic agent of the plague. A hallmark of plague is subversion of the host immune response by disrupting host signalling pathways required for inflammation. This non-inflammatory environment permits bacterial colonization and has been shown to be essential for disease manifestation. Previous work has shown that Y. pestis inhibits phagocytosis and degranulation by neutrophils. Manipulation of these key vesicular trafficking pathways suggests that Y. pestis influences extracellular vesicle (EV) secretion, cargo selection, trafficking and/or maturation. Our goals were to define the EV population produced by neutrophils in response to Y. pestis and determine how these vesicles might influence inflammation. Towards these goals, EVs were isolated from human neutrophils infected with Y. pestis or a mutant lacking bacterial effector proteins known to manipulate host cell signalling. Mass spectrometry data revealed that cargoes packaged in EVs isolated from mutant infected cells were enriched with antimicrobial and cytotoxic proteins, contents which differed from uninfected and Y. pestis infected cells. Further, EVs produced in response to Y. pestis lacked inflammatory properties observed in those isolated from neutrophils responding to the mutant. Together, these data demonstrate that Y. pestis actively inhibits the production of antimicrobial EVs produced by neutrophils, likely contributing to immune evasion.
Collapse
Affiliation(s)
- Katelyn R. Sheneman
- Department of Microbiology and ImmunologyUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Timothy D. Cummins
- Department of Medicine and Proteomics Technology CenterUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Michael L. Merchant
- Department of Pharmacology and ToxicologyUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Joshua L. Hood
- Department of Pharmacology and ToxicologyUniversity of LouisvilleLouisvilleKentuckyUSA
- Hepatobiology and Toxicology COBREUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Silvia M. Uriarte
- Department of Oral Immunology & Infectious DiseaseUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Matthew B. Lawrenz
- Department of Microbiology and ImmunologyUniversity of LouisvilleLouisvilleKentuckyUSA
- Center for Predictive Medicine for Biodefense and Emerging Infectious DiseasesUniversity of LouisvilleLouisvilleKentuckyUSA
| |
Collapse
|
14
|
Kim S, Lee W, Kim HI, Kim MK, Choi TS. Recent advances and future challenges in predictive modeling of metalloproteins by artificial intelligence. Mol Cells 2025; 48:100191. [PMID: 39938866 PMCID: PMC11919430 DOI: 10.1016/j.mocell.2025.100191] [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: 12/03/2024] [Revised: 01/31/2025] [Accepted: 02/01/2025] [Indexed: 02/14/2025] Open
Abstract
Metal coordination is essential for structural/catalytic functions of metalloproteins that mediate a wide range of biological processes in living organisms. Advances in bioinformatics have significantly enhanced our understanding of metal-binding sites and their functional roles in metalloproteins. State-of-the-art computational models developed for metal-binding sites seamlessly integrate protein sequence and structural data to unravel the complexities of metal coordination environments. Our goal in this mini-review is to give an overview of these tools and highlight the current challenges (predicting dynamic metal-binding sites, determining functional metalation states, and designing intricate coordination networks) remaining in the predictive models of metal-binding sites. Addressing these challenges will not only deepen our knowledge of natural metalloproteins but also accelerate the development of artificial metalloproteins with novel and precisely engineered functionalities.
Collapse
Affiliation(s)
- Soohyeong Kim
- Departments of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Wonseok Lee
- Division of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Hugh I Kim
- Departments of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Min Kyung Kim
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Tae Su Choi
- Division of Life Sciences, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
15
|
Ma H, Wang M, Yao Y, Zhang S, Wang M, Zhu D, Jia R, Chen S, Zhao X, Yang Q, Wu Y, Zhang S, Huang J, Tian B, Ou X, Sun D, He Y, Wu Z, Zhang L, Yu Y, Cheng A, Liu M. ZntR is a critical regulator for zinc homeostasis and involved in pathogenicity in Riemerella anatipestifer. Microbiol Spectr 2025; 13:e0317824. [PMID: 40035565 PMCID: PMC11960050 DOI: 10.1128/spectrum.03178-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 01/31/2025] [Indexed: 03/05/2025] Open
Abstract
Zinc (Zn2+) is essential for all bacteria, but excessive Zn2+ levels are toxic. Bacteria maintain zinc homeostasis through regulators, such as Zur, AdcR, and ZntR. Riemerella anatipestifer is a significant Flavobacteriales pathogen causing acute serositis in ducks and other birds. In this study, we identified a homolog of ZntR, a regulator for zinc homeostasis, and demonstrated its contribution to the pathogenicity of R. anatipestifer. Deletion of zntR makes the bacteria hypersensitive to excess Zn2+ but not to other metals like manganese (Mn2+), copper (Cu2+), cobalt (Co2+), and nickel (Ni2+). Deletion of zntR also leads to intracellular zinc accumulation but not of other metals. Additionally, compared to the wild type, the deletion of zntR increases resistance to oxidants hydrogen peroxide (H2O2) and sodium hypochlorite (NaOCl), respectively. The deletion of zntR causes significant changes in transcriptional and protein expression levels, revealing 35 genes with potential zinc metabolism functions. Among them, zupT, which is inhibited by ZntR, is required for zinc transport and resistance to oxidative stress. Finally, deletion of zntR leads to attenuation of colonization in ducklings. In summary, ZntR is a crucial regulator for zinc homeostasis and contributes to the pathogenicity of R. anatipestifer.IMPORTANCEZinc homeostasis plays a critical role in the environmental adaptability of bacteria. Riemerella anatipestifer is a significant pathogen in poultry with the potential to encounter zinc-deficient or zinc-excess environment. The mechanism of zinc homeostasis in this bacterium remains largely unexplored. In this study, we showed that the transcriptional regulator ZntR of R. anatipestifer is critical for zinc homeostasis by altering the transcription and expression of a number of genes. Importantly, ZntR inhibits the transcription of zinc transporter ZupT and contributes to colonization in R. anatipestifer. The results are significant for understanding zinc homeostasis and the pathogenic mechanisms in R. anatipestifer.
Collapse
Affiliation(s)
- Hongmeng Ma
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mengying Wang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yizhou Yao
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shutong Zhang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mingshu Wang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Dekang Zhu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Renyong Jia
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shun Chen
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xinxin Zhao
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qiao Yang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ying Wu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shaqiu Zhang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Juan Huang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Bin Tian
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xumin Ou
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Di Sun
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yu He
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhen Wu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ling Zhang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yanling Yu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Anchun Cheng
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mafeng Liu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
16
|
Araújo-Pereira M, Andrade BB. Oxidative battles in tuberculosis: walking the ferroptotic tightrope. Trends Immunol 2025; 46:338-351. [PMID: 40122726 DOI: 10.1016/j.it.2025.02.009] [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: 12/20/2024] [Revised: 02/12/2025] [Accepted: 02/18/2025] [Indexed: 03/25/2025]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains one of the leading causes of death worldwide. TB pathogenesis is shaped by a complex interaction between the pathogen and host immune responses, particularly through mechanisms such as oxidative stress and ferroptosis; a form of regulated necrotic cell death driven by iron-dependent lipid peroxidation. This Review highlights recent insights into how Mtb modulates oxidative stress pathways and thus triggers ferroptosis in host cells. Understanding the interplay between oxidative stress responses and cellular and tissue necrosis opens new avenues for therapeutic interventions of TB by controlling bacterial growth and preventing host tissue damage.
Collapse
Affiliation(s)
- Mariana Araújo-Pereira
- Laboratório de Pesquisa Clínica e Translacional (LPCT), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia 40296-710, Brazil; Instituto de Pesquisa Clínica e Translacional (IPCT), Medicina Zarns, Clariens Educação, Salvador, Bahia 41720-200, Brazil; Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Institute, Salvador, Bahia 41810-710, Brazil.
| | - Bruno B Andrade
- Laboratório de Pesquisa Clínica e Translacional (LPCT), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia 40296-710, Brazil; Instituto de Pesquisa Clínica e Translacional (IPCT), Medicina Zarns, Clariens Educação, Salvador, Bahia 41720-200, Brazil; Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Institute, Salvador, Bahia 41810-710, Brazil.
| |
Collapse
|
17
|
Perfect JR, Kronstad JW. Cryptococcal nutrient acquisition and pathogenesis: dining on the host. Microbiol Mol Biol Rev 2025; 89:e0001523. [PMID: 39927764 PMCID: PMC11948494 DOI: 10.1128/mmbr.00015-23] [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] [Indexed: 02/11/2025] Open
Abstract
SUMMARYPathogens must acquire essential nutrients to successfully colonize and proliferate in host tissue. Additionally, nutrients provide signals that condition pathogen deployment of factors that promote disease. A series of transcriptomics experiments over the last 20 years, primarily with Cryptococcus neoformans and to a lesser extent with Cryptococcus gattii, provide insights into the nutritional requirements for proliferation in host tissues. Notably, the identified functions include a number of transporters for key nutrients including sugars, amino acids, metals, and phosphate. Here, we first summarize the in vivo gene expression studies and then discuss the follow-up analyses that specifically test the relevance of the identified transporters for the ability of the pathogens to cause disease. The conclusion is that predictions based on transcriptional profiling of cryptococcal cells in infected tissue are well supported by subsequent investigations using targeted mutations. Overall, the combination of transcriptomic and genetic approaches provides substantial insights into the nutritional requirements that underpin proliferation in the host.
Collapse
Affiliation(s)
- John R. Perfect
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - James W. Kronstad
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
18
|
Sharifian Gh M, Norouzi F, Sorci M, Zaidi TS, Pier GB, Achimovich A, Ongwae GM, Liang B, Ryan M, Lemke M, Belfort G, Gadjeva M, Gahlmann A, Pires MM, Venter H, Harris TE, Laurie GW. Lacritin Cleavage-Potentiated Targeting of Iron - Respiratory Reciprocity Promotes Bacterial Death. J Biol Chem 2025:108455. [PMID: 40154612 DOI: 10.1016/j.jbc.2025.108455] [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: 01/24/2025] [Revised: 03/15/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025] Open
Abstract
Discovering new bacterial signaling pathways offers unique antibiotic strategies. With current antibiotic classes targeting cell wall synthesis or depolarizing the inner-membrane or altering the bacterial metabolome or inhibiting replication or transcription pathways, manipulation of transporters to limit bacterial respiration and thereby pathogenesis has been a decades long quest. Here we report an inhibitor of multiple bacterial transporters. The inhibitor is the bactericidal N-104 endogenous cleavage fragment of the prosecretory mitogen lacritin. Lacritin is now known to be widely distributed in plasma, cerebral spinal fluid, tears and saliva. With the bactericidal mechanism determined to be nonlytic by surface plasmon resonance as confirmed by lack of SYTOX Orange entry, we performed an unbiased resistance screen of 3,884 E. coli gene knockout strains revealing a complex N-104 polypharmacology. Validation in the virulent P. aeruginosa strain PA14 - one of three WHO Priority 1: Critical list species focused on an approach that sequentially couples three transporters and downstream transcription to lethally suppress respiration. By targeting outer membrane YaiW, cationic N-104 translocates into the periplasm where it ligates inner membrane transporters FeoB and PotH, respectively, to suppress both ferrous iron and polyamine uptake. With FeoB favoring an anaerobic environment, N-104 promotes the expression of genes regulating anaerobic respiration while largely suppressing those involved in aerobic respiration - a strategy counterproductive under aerobic conditions. This mechanism is innate to the surface of the eye and is enhanced by synergistic coupling with tear thrombin fragment GKY20 as tested on antibiotic resistant clinical isolates.
Collapse
Affiliation(s)
| | - Fatemeh Norouzi
- Departments of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - Mirco Sorci
- Howard P. Isermann Department of Chemical and Biological Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Tanweer S Zaidi
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Gerald B Pier
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | | | - Binyong Liang
- Physiology, University of Virginia, Charlottesville, VA, USA
| | - Margaret Ryan
- Departments of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - Michael Lemke
- Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Georges Belfort
- Howard P. Isermann Department of Chemical and Biological Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Mihaela Gadjeva
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Marcos M Pires
- Chemistry, University of Virginia, Charlottesville, VA, USA
| | - Henrietta Venter
- Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
| | - Thurl E Harris
- Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Gordon W Laurie
- Departments of Cell Biology, University of Virginia, Charlottesville, VA, USA; Biomedical Engineering, University of Virginia, Charlottesville, VA, USA; Ophthalmology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
19
|
Zimmerman TJ, Carabeo RA. Chlamydia trachomatis invasion: a duet of effectors. Biochem Soc Trans 2025; 0:BST20240800. [PMID: 40131835 DOI: 10.1042/bst20240800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025]
Abstract
Members of the genus Chlamydia require an intracellular niche for growth and replication, thus highlighting the extreme significance of its ability to invade epithelial cells-the favored host cell in vivo. Because epithelial cells are not phagocytic, the uptake of Chlamydia must be driven by the pathogen. To this end, two bacterial proteins, translocated actin-recruiting protein (TarP) and translocated membrane effector A (TmeA), identified in Chlamydia trachomatis are translocated from the infectious chlamydial elementary bodies to the host cell cytosol to facilitate extensive remodeling of the cortical actin network to produce protrusive structures designed for pathogen engulfment. Notably, both effectors act by promoting highly localized actin nucleation at sites of bacterial adhesion. However, they have non-redundant functions, with both required for optimal actin remodeling dynamics and efficient invasion. Finally, these effectors also mediate the latter stages of the invasion process, specifically by modulating host dynamin 2, a large GTPase critical to closure and scission of invaginating vesicles harboring elementary bodies. In summary, TarP and TmeA orchestrate major aspects of C. trachomatis invasion.
Collapse
Affiliation(s)
- Tyler J Zimmerman
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska 68198, U.S.A
| | - Rey A Carabeo
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska 68198, U.S.A
| |
Collapse
|
20
|
Pawlik K, Ostrowska M, Gumienna-Kontecka E. Systematic Model Peptide Studies: A Crucial Step To Understand the Coordination Chemistry of Mn(II) and Fe(II) in Proteins. Inorg Chem 2025; 64:5472-5486. [PMID: 40067133 PMCID: PMC11938343 DOI: 10.1021/acs.inorgchem.4c05380] [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: 12/17/2024] [Revised: 02/12/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025]
Abstract
Pathogenic bacteria and all other species require Mn(II) and Fe(II) ions for proper growth. Microbes use a variety of assimilation pathways to obtain the necessary metal ions, and their metal homeostasis mechanisms are still not fully uncovered. The knowledge of the poorly discovered complexation chemistry of Mn(II) and Fe(II) ions could help us to understand the basis of those processes better. We have designed six model peptides (L1 - Ac-HHHHHH-NH2, L2 - Ac-HHHHHHHHH-NH2, L3 - Ac-HAHAHAHAH-NH2, L4 - Ac-HHAAAAAAAAAHHHH-NH2, L5 - Ac-HDHDHDHDH-NH2, and L6 - Ac-HEHEHEHEH-NH2) inspired by Mn(II) and Fe(II) binding motifs that are prevalent in nature, in order to clarify their coordination preferences. Spectrometric, spectroscopic, and potentiometric techniques were used to determine the thermodynamic and structural properties of the studied systems. All of the investigated ligands possess efficient Mn(II), Fe(II), and Zn(II) binding sites. Complex stability and metal affinity are significantly influenced by the length of the peptide sequences, as well as the location and quantity of coordinating amino acid residues like His, Asp, and Glu.
Collapse
Affiliation(s)
- Karolina Pawlik
- Faculty of Chemistry, University of Wrocław, Wrocław 50-383, Poland
| | | | | |
Collapse
|
21
|
Germon P, Foucras G, Smith DGE, Rainard P. INVITED REVIEW: Mastitis Escherichia coli strains: Mastitis-Associated or Mammo-Pathogenic ? J Dairy Sci 2025:S0022-0302(25)00158-4. [PMID: 40139360 DOI: 10.3168/jds.2024-26109] [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: 12/04/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025]
Abstract
Bovine mastitis remains a major concern for dairy farmers, mainly because of its impact on the economy of their activity and on animal welfare. Because Escherichia coli is considered a major mastitis pathogen, the diversity of E. coli strains isolated from mastitis cases has been studied for decades, with the aim to discover new ways to fight this infection. With the recent advances in whole-genome sequencing, a detailed view of the peculiarities of mastitis E. coli strains has emerged. This review aims to bring together the knowledge garnered over the years with the more recent results of whole-genome analyses. While the concept of a Mammary Pathogenic E. coli has been proposed, because a common set of virulence genes cannot be identified among mastitis E. coli strains, we prefer the use of Mastitis-associated E. coli (MAEC) with MAEC being more an "ecotype" rather than a "pathotype." Indeed, data available so far suggest that a common feature of MAEC would rather be an enrichment in fitness capabilities that makes them well-suited for survival and rapid adaptation to changing biotopes in the mammary gland which we qualify as intramammary ecotopes.
Collapse
Affiliation(s)
- Pierre Germon
- ISP UMR 1282, INRAE, Université François Rabelais de Tours, Nouzilly, France.
| | - Gilles Foucras
- IHAP, Université de Toulouse, INRAE, ENVT, Toulouse, France
| | | | - Pascal Rainard
- ISP UMR 1282, INRAE, Université François Rabelais de Tours, Nouzilly, France
| |
Collapse
|
22
|
Montes N, Wilson T, Krug SA, Mouriño S, Kane MA, Deredge D, Wilks A. The Pseudomonas aeruginosa PhuS proximal ligand His-209 triggers a conformational switch in function from DNA binding to heme transfer. J Biol Chem 2025; 301:108440. [PMID: 40122173 DOI: 10.1016/j.jbc.2025.108440] [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: 11/16/2024] [Revised: 02/22/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
Pseudomonas aeruginosa can acquire iron from heme via the heme assimilation system and Pseudomonas heme uptake (Phu) systems. Heme uptake is regulated at the metabolic level by the cytoplasmic protein PhuS that controls heme flux through a heme oxygenase HemO, releasing iron and biliverdin IXβ and IXδ. We have shown PhuS regulates extracellular heme flux, and in its apo-form transcriptionally regulates the iron and heme-dependent small RNAs (sRNAs) PrrF/PrrH. This mutual exclusivity of function is driven by conformational rearrangement of PhuS on heme binding. Herein, we show through a combination of EMSA and fluorescence anisotropy that mutation of the His-209 proximal ligand allows both apo- and holo-PhuS H209A to bind to the prrF1 promoter with significantly lower affinity when compared to PAO1 WT. Hydrogen deuterium exchange coupled to mass spectrometry revealed the apo- and holo-PhuS H209A structures are closer to each other than their WT counterparts and sample a conformational landscape between the apo- and holo-PhuS WT conformations, that is neither optimal for heme transfer nor DNA-binding. Furthermore, quantitative PCR and Western blot analysis of the phuSH209A allelic strain compared to PAO1 WT revealed an uncoupling of the PhuS-HemO dependent regulation of heme flux into the cell that abrogates the heme dependent regulation of the PrrF/PrrH sRNAs. The data supports a model where heme coordination through His-209 drives the conformational switch that determines mutual exclusivity in function of apo- and holo-PhuS. This dual function of PhuS is central to integrating extracellular heme utilization into the PrrF/PrrH sRNA regulatory network critical for P. aeruginosa adaptation within the host.
Collapse
Affiliation(s)
- Nicholas Montes
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Tyree Wilson
- Molecular Pathogenesis and Biomarkers Section, National Institute of Allergy and Infectious Disease, Rockville, Maryland, USA
| | - Samuel A Krug
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Susana Mouriño
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Daniel Deredge
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Angela Wilks
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA.
| |
Collapse
|
23
|
Li S, Jiang G, Wang S, Wang M, Wu Y, Zhang J, Liu X, Zhong L, Zhou M, Xie S, Ren Y, He P, Lou Y, Li H, Du J, Zhou Z. Emergence and global spread of a dominant multidrug-resistant clade within Acinetobacter baumannii. Nat Commun 2025; 16:2787. [PMID: 40118837 PMCID: PMC11928498 DOI: 10.1038/s41467-025-58106-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 03/12/2025] [Indexed: 03/24/2025] Open
Abstract
The proliferation of multi-drug resistant (MDR) bacteria is driven by the global spread of epidemic lineages that accumulate antimicrobial resistance genes (ARGs). Acinetobacter baumannii, a leading cause of nosocomial infections, displays resistance to most frontline antimicrobials and represents a significant challenge to public health. In this study, we conduct a comprehensive genomic analysis of over 15,000 A. baumannii genomes to identify a predominant epidemic super-lineage (ESL) accounting for approximately 70% of global isolates. Through hierarchical classification of the ESL into distinct lineages, clusters, and clades, we identified a stepwise evolutionary trajectory responsible for the worldwide expansion and transmission of A. baumannii over the last eight decades. We observed the rise and global spread of a previously unrecognized Clade 2.5.6, which emerged in East Asia in 2006. The epidemic of the clade is linked to the ongoing acquisition of ARGs and virulence factors facilitated by genetic recombination. Our results highlight the necessity for One Health-oriented research and interventions to address the spread of this MDR pathogen.
Collapse
Affiliation(s)
- Shengkai Li
- MOE Key Laboratory of Geriatric Diseases and Immunology, Cancer Institute, Suzhou Medical College, Soochow University, Suzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Guilai Jiang
- MOE Key Laboratory of Geriatric Diseases and Immunology, Cancer Institute, Suzhou Medical College, Soochow University, Suzhou, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| | - Shengke Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Min Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yilei Wu
- MOE Key Laboratory of Geriatric Diseases and Immunology, Cancer Institute, Suzhou Medical College, Soochow University, Suzhou, China
- Department of Life Sciences, Imperial College London, London, UK
| | - Jinzhi Zhang
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Xiao Liu
- MOE Key Laboratory of Geriatric Diseases and Immunology, Cancer Institute, Suzhou Medical College, Soochow University, Suzhou, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| | - Ling Zhong
- MOE Key Laboratory of Geriatric Diseases and Immunology, Cancer Institute, Suzhou Medical College, Soochow University, Suzhou, China
| | - Min Zhou
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shichang Xie
- MOE Key Laboratory of Geriatric Diseases and Immunology, Cancer Institute, Suzhou Medical College, Soochow University, Suzhou, China
- Iotabiome Biotechnology Inc., Suzhou, China
| | - Yi Ren
- Iotabiome Biotechnology Inc., Suzhou, China
| | - Ping He
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongliang Lou
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Heng Li
- MOE Key Laboratory of Geriatric Diseases and Immunology, Cancer Institute, Suzhou Medical College, Soochow University, Suzhou, China.
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China.
| | - Jimei Du
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Zhemin Zhou
- MOE Key Laboratory of Geriatric Diseases and Immunology, Cancer Institute, Suzhou Medical College, Soochow University, Suzhou, China.
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China.
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China.
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| |
Collapse
|
24
|
He S, Zheng S, Hu Y, Yang Z, Zhu H, Yu B, Wei J, Pan G, Zhou Z, Li C. The unique transmembrane protein NbTMP2 (NBO_555g0004) of Nosema bombycis contributes to host infection. J Invertebr Pathol 2025; 211:108320. [PMID: 40120668 DOI: 10.1016/j.jip.2025.108320] [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: 12/05/2024] [Revised: 02/18/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Nosema bombycis is one of the earliest discovered microsporidia and can infect silkworms through both horizontal and transovarial transmission, posing a significant threat to the sericulture industry. Microsporidia can form mature dormant spores with a thick proteinaceous and chitin-rich wall that shields them from environmental stress, leading to difficulties in prevention and control. However, the intracellular proliferative phase is a particularly active life stage for the pathogen. At this stage, the plasma membrane of pathogen is exposed to the cytoplasm of the host cells without the protection of the spore walls, which make it an ideal phase for intervention. In this study, based on transcriptomic data, we identified that a transmembrane protein of N. bombycis, NbTMP2 (NBO_555g0004), was highly expressed after infection with microsporidia. Sequence analysis of the cloned NbTMP2 gene revealed that the protein consists of 253 amino acids, including a signal peptide and a transmembrane domain. Indirect immunofluorescence analysis (IFA) showed that NbTMP2 is localized to the plasma membrane. Furthermore, IFA, RT-qPCR, and western blotting indicated that NbTMP2 is expressed at all developmental stages, with a significant upregulation during the early proliferative phase of N. bombycis. The interference of NbTMP2 confirmed that downregulation of NbTMP2 expression significantly inhibit the proliferation of N. bombycis. In conclusion, NbTMP2 is a membrane protein that plays a role during the development of N. bombycis. This study provides a potential target for inhibiting the proliferation of N. bombycis and lays a foundation for future research on breeding N. bombycis-resistant silkworms.
Collapse
Affiliation(s)
- Shaogang He
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Shiyi Zheng
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Affiliated Jinhua Hospital, Zhejiang University of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang 321000, China
| | - Yuanke Hu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Zhonghua Yang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Honglin Zhu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Bin Yu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China; Laboratory of Tissue and Cell Biology, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, China
| | - Junhong Wei
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Guoqing Pan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
| | - Zeyang Zhou
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China; College of Life Sciences, Chongqing Normal University, Chongqing 401331, China
| | - Chunfeng Li
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China.
| |
Collapse
|
25
|
Heckart A, Cocuron JC, Ray SC, Rappleye CA, Alonso AP. Revealing pathogenesis-associated metabolites in Histoplasma capsulatum through comprehensive metabolic profiling. mSystems 2025; 10:e0018625. [PMID: 40013825 PMCID: PMC11915866 DOI: 10.1128/msystems.00186-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
During infection, Histoplasma capsulatum yeasts interact with a variety of phagocytic cells, where macrophages represent an important niche for long-term intracellular fungal survival and replication. In the phagosomes of macrophages, a hostile environment where most microorganisms are killed, Histoplasma not only survives but overcomes several biological challenges and proliferates intracellularly. To better understand the characteristics of intracellular Histoplasma and the phagosomal environment, a metabolomic platform was used to analyze Histoplasma yeasts cultured on different carbon sources and yeasts extracted from macrophages, identifying metabolites associated with pathogenesis. Metabolomic results of in vitro-grown yeasts were further characterized with available transcriptomic data, informing underlying gene expression patterns in response to contrasting milieus. These approaches revealed that Histoplasma yeasts, unlike many other yeasts, do not ferment sugars to ethanol, and, when cultivated on glycolytic versus gluconeogenic carbon sources, produce distinct metabolomes with altered intracellular amino acid, lipid, and sugar contents. Furthermore, analysis of Histoplasma-inoculated media illustrated that Histoplasma secretes mannitol and anthranilates. Lastly, a comparison of the metabolomes derived from in vitro cultivation versus intracellular growth highlighted leucine and cysteine/cystine as amino acids, which may serve as sources of carbon, nitrogen, and sulfur to yeasts within macrophages. These results detail metabolites linked to Histoplasma metabolism during macrophage infection, identifying potential candidates to target for novel histoplasmosis therapeutics.IMPORTANCEIntracellular pathogens reside within host cells, surviving against innate immune responses while exploiting host resources to proliferate. Understanding the mechanisms that underlie their survival and proliferation is critical for developing novel treatments and therapeutics for the diseases these pathogens cause. While Histoplasma is a unique example of a true intra-phagosomal pathogen, insights into its pathogenesis may still inform the study of other intracellular pathogens.
Collapse
Affiliation(s)
- Adrian Heckart
- Department of Biological Sciences & BioDiscovery Institute, University of North Texas, Denton, Texas, USA
| | | | - Stephanie C Ray
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Chad A Rappleye
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Ana P Alonso
- Department of Biological Sciences & BioDiscovery Institute, University of North Texas, Denton, Texas, USA
- BioAnalytical Facility, University of North Texas, Denton, Texas, USA
| |
Collapse
|
26
|
Yu Y, Xie B, Wang J, Luo W, Yang M, Xiong Z, Huang G, Yang J, Tang Z, Qiao R, Yuan Z, He L, Chen T. Translational Selenium Nanoparticles Promotes Clinical Non-small-cell Lung Cancer Chemotherapy via Activating Selenoprotein-driven Immune Manipulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2415818. [PMID: 40095246 DOI: 10.1002/adma.202415818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Reconstructing the tumor immune microenvironment is an effective strategy to enhance therapeutic efficacy limited by immunosuppression in non-small-cell lung cancer (NSCLC). In this study, it is found that selenium (Se) depletion and immune dysfunction are present in patients with advanced NSCLC compared with healthy volunteers. Surprisingly, Se deficiency resulted in decreased immunity and accelerated rapid tumor growth in the mice model, which further reveals that the correlation between micronutrient Se and lung cancer progression. This pioneering work achieves 500-L scale production of Se nanoparticles (SeNPs) at GMP level and utilizes it to reveal how and why the trace element Se can enhance clinical immune-mediated treatment efficacy against NSCLC. The results found that translational SeNPs can promote the proliferation of NK cells and enhance its cytotoxicity against cancer cells by activating mTOR signaling pathway driven by GPXs to regulate the secretion of cytokines to achieve an antitumor response. Moreover, a clinical study of an Investigator-initiated Trial shows that translational SeNPs supplementation in combination with bevacizumab/cisplatin/pemetrexed exhibits enhanced therapeutic efficacy with an objective response rate of 83.3% and a disease control rate of 100%, through potentiating selenoprotein-driven antitumor immunity. Taken together, this study, for the first time, highlights the translational SeNPs-enhanced therapeutic efficacy against clinical advanced NSCLC.
Collapse
Affiliation(s)
- Yanzi Yu
- College of Chemistry and Materials Science, Department of Oncology and Nano-therapeutics Institute of The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Bin Xie
- College of Chemistry and Materials Science, Department of Oncology and Nano-therapeutics Institute of The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Jinlin Wang
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Weizhan Luo
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Meijin Yang
- College of Chemistry and Materials Science, Department of Oncology and Nano-therapeutics Institute of The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Zushuang Xiong
- College of Chemistry and Materials Science, Department of Oncology and Nano-therapeutics Institute of The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Guanning Huang
- College of Chemistry and Materials Science, Department of Oncology and Nano-therapeutics Institute of The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Jianwei Yang
- College of Chemistry and Materials Science, Department of Oncology and Nano-therapeutics Institute of The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Zhiying Tang
- College of Chemistry and Materials Science, Department of Oncology and Nano-therapeutics Institute of The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Rui Qiao
- College of Chemistry and Materials Science, Department of Oncology and Nano-therapeutics Institute of The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Zhongwen Yuan
- College of Chemistry and Materials Science, Department of Oncology and Nano-therapeutics Institute of The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Lizhen He
- College of Chemistry and Materials Science, Department of Oncology and Nano-therapeutics Institute of The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Tianfeng Chen
- College of Chemistry and Materials Science, Department of Oncology and Nano-therapeutics Institute of The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
27
|
Orzel B, Ostrowska M, Potocki S, Zoroddu MA, Kozlowski H, Peana M, Gumienna-Kontecka E. The Coordination Chemistry of Two Peptidic Models of NFeoB and Core CFeoB Regions of FeoB Protein: Complexes of Fe(II), Mn(II), and Zn(II). Inorg Chem 2025; 64:5038-5052. [PMID: 40048504 PMCID: PMC11920956 DOI: 10.1021/acs.inorgchem.4c05111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Often necessary for efficient Fe(II) trafficking into bacterial cell, the Feo system is a vital transporter for many pathogenic bacteria and indispensable for proper development and survival in the host organism during infection. In this work, we present the metal-binding characteristics of the peptidic models of two putative Fe(II)-binding sites of E. coliFeoB: L1 (Ac-477IMRGEATPFVMELPVYHVPH496-CONH2) being a fragment of the Core CFeoB region located between the transmembrane helices and L2 (Ac-38VERKEG43-CONH2), which represents the ExxE motif found within the NFeoB domain. With a variety of physicochemical methods, such as potentiometry, mass spectrometry, NMR, and EPR spectroscopy, we have determined the stability constants and metal-binding residues for the complexes of Fe(II), Mn(II), and Zn(II) with two ligands, L1 and L2, acting as models for the Core CFeoB and ExxE motif. We compare their affinities toward the studied metal ions with the previously studied C-terminal part of the protein and discuss a possible role in metal trafficking by the whole protein.
Collapse
Affiliation(s)
- Bartosz Orzel
- Faculty of Chemistry, University of Wrocław, Wrocław 50-383, Poland
| | | | - Slawomir Potocki
- Faculty of Chemistry, University of Wrocław, Wrocław 50-383, Poland
| | - Maria Antonietta Zoroddu
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Sassari 07100, Italy
| | - Henryk Kozlowski
- Faculty of Chemistry, University of Wrocław, Wrocław 50-383, Poland
- Faculty of Health Sciences, University of Opole, Katowicka, Opole 68 45-060, Poland
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Sassari 07100, Italy
| | | |
Collapse
|
28
|
Cherrak Y, Younes AA, Perez-Molphe-Montoya E, Maurer L, Yilmaz K, Enz U, Zeder C, Kiefer P, Christen P, Gül E, Vorholt JA, von Mering C, Hardt WD. Neutrophil recruitment during intestinal inflammation primes Salmonella elimination by commensal E. coli in a context-dependent manner. Cell Host Microbe 2025; 33:358-372.e4. [PMID: 40023150 DOI: 10.1016/j.chom.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/19/2024] [Accepted: 02/05/2025] [Indexed: 03/04/2025]
Abstract
Foodborne bacterial diarrhea involves complex pathogen-microbiota-host interactions. Pathogen-displacing probiotics are increasingly popular, but heterogeneous patient outcomes highlighted the need to understand individualized host-probiotic activity. Using the mouse gut commensal Escherichia coli 8178 and the human probiotic E. coli Nissle 1917, we found that the degree of protection against the enteric pathogen Salmonella enterica serovar Typhimurium (S. Tm) varies across mice with distinct gut microbiotas. Pathogen clearance is linked to enteropathy severity and subsequent recruitment of intraluminal neutrophils, which differs in a microbiota-dependent manner. By combining mouse knockout and antibody-mediated depletion models with bacterial genetics, we show that neutrophils and host-derived reactive oxygen species directly influence E. coli-mediated S. Tm displacement by potentiating siderophore-bound toxin killing. Our work demonstrates how host immune factors shape pathogen-displacing probiotic efficiency while also revealing an unconventional antagonistic interaction where a gut commensal and the host synergize to displace an enteric pathogen.
Collapse
Affiliation(s)
- Yassine Cherrak
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland.
| | - Andrew Abi Younes
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Eugenio Perez-Molphe-Montoya
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, 8057 Zurich, Switzerland
| | - Luca Maurer
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Koray Yilmaz
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Ursina Enz
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Christophe Zeder
- Laboratory of Nutrition and Metabolic Epigenetics, Department of Health Science and Technology, 8092 Zurich, Switzerland
| | - Patrick Kiefer
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Philipp Christen
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Ersin Gül
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Julia A Vorholt
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Christian von Mering
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, 8057 Zurich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
29
|
Stow PR, Forsch KO, Thomsen E, Naka H, Haygood MG, Barbeau KA, Butler A. Stereospecific control of microbial growth by a combinatoric suite of chiral siderophores. Proc Natl Acad Sci U S A 2025; 122:e2423730122. [PMID: 40030022 PMCID: PMC11912440 DOI: 10.1073/pnas.2423730122] [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: 11/14/2024] [Accepted: 01/17/2025] [Indexed: 03/19/2025] Open
Abstract
Bacteria compete for iron by producing small-molecule chelators known as siderophores. The triscatechol siderophores trivanchrobactin and ruckerbactin, produced by Vibrio campbellii DS40M4 and Yersinia ruckeri YRB, respectively, are naturally occurring diastereomers that form chiral ferric complexes in opposing enantiomeric configurations. Chiral recognition is a hallmark of specificity in biological systems, yet the biological consequences of chiral coordination compounds are relatively unexplored. We demonstrate stereoselective discrimination of microbial growth and iron uptake by chiral Fe(III)-siderophores. The siderophore utilization pathway in V. campbellii DS40M4 is stereoselective for Λ-Fe(III)-trivanchrobactin, but not the mismatched Δ-Fe(III)-ruckerbactin diastereomer. Chiral recognition is likely conferred by the stereospecificity of both the outer membrane receptor (OMR) protein FvtA and the periplasmic binding protein (PBP) FvtB, both of which must interact preferentially with the Λ-configured Fe(III)-coordination complexes.
Collapse
Affiliation(s)
- Parker R. Stow
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA93106
| | - Kiefer O. Forsch
- Geosciences Research Division, Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA92093
| | - Emil Thomsen
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA93106
| | - Hiroaki Naka
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT84112
| | - Margo G. Haygood
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT84112
| | - Katherine A. Barbeau
- Geosciences Research Division, Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA92093
| | - Alison Butler
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA93106
| |
Collapse
|
30
|
Buglino JA, Ozakman Y, Hatch CE, Benjamin A, Tan DS, Glickman MS. Chalkophore mediated respiratory oxidase flexibility controls M. tuberculosis virulence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.12.589290. [PMID: 38645185 PMCID: PMC11030325 DOI: 10.1101/2024.04.12.589290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Oxidative phosphorylation has emerged as a critical therapeutic vulnerability of M. tuberculosis (Mtb). However, it is unknown how intracellular bacterial pathogens such as Mtb maintain respiration during infection despite the chemical effectors of host immunity. Mtb synthesizes diisonitrile lipopeptides that tightly chelate copper, but the role of these chalkophores in host-pathogen interactions is also unknown. We demonstrate that M. tuberculosis chalkophores maintain the function of the heme-copper bcc:aa 3 respiratory supercomplex under copper limitation. Chalkophore deficiency impairs Mtb survival, respiration to oxygen, and ATP production under copper deprivation in culture, effects that are exacerbated by loss of the heme dependent Cytochrome BD respiratory oxidase. Our genetic analyses indicate that maintenance of respiration is the only cellular target of chalkophore mediated copper acquisition. M. tuberculosis lacking chalkophore biosynthesis is attenuated in mice, a phenotype that is also severely exacerbated by loss of the CytBD respiratory oxidase. We find that the host immune pressure that attenuates chalkophore deficient Mtb is independent of adaptive immunity and neutrophils. These data demonstrate that chalkophores counter host inflicted copper deprivation and highlight a multilayered system by which M. tuberculosis maintains respiration during infection.
Collapse
Affiliation(s)
- John A. Buglino
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065 USA
| | - Yaprak Ozakman
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065 USA
| | - Chad E. Hatch
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065 USA
| | - Anna Benjamin
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065 USA
| | - Derek S. Tan
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065 USA
- Tri-Institutional Research Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065 USA
| | - Michael S. Glickman
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065 USA
| |
Collapse
|
31
|
Alves e Silva TL, Kanatani S, Barletta Ferreira AB, Schwartz C, Talyuli OA, Olivas J, Nagata BM, Pala ZR, Pascini T, Alves DA, Zhao M, Suzuki M, Dorner LP, Frischknecht F, Coppens I, Barillas-Mury C, Ribeiro JM, Sinnis P, Vega-Rodriguez J. High-Resolution Proteomics Unveils Salivary Gland Disruption and Saliva-Hemolymph Protein Exchange in Plasmodium-Infected Mosquitoes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640873. [PMID: 40060675 PMCID: PMC11888397 DOI: 10.1101/2025.02.28.640873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Plasmodium sporozoites, the stage that initiates a malaria infection, must invade the mosquito salivary glands (SGs) before transmitting to a vertebrate host. However, the effects of sporozoite invasion on salivary gland physiology and saliva composition remain largely unexplored. We examined the impact of Plasmodium infection on Anopheles gambiae salivary glands using high-resolution proteomics, gene expression, and morphological analysis. The data revealed differential expression of various proteins, including the enrichment of humoral proteins in infected salivary glands originating from the hemolymph. These proteins diffused into the SGs due to structural damage caused by the sporozoites during invasion. Conversely, saliva proteins diffused out into the circulation of infected mosquitoes. Moreover, infection altered saliva protein composition, as shown by proteomes from saliva collected from mosquitoes infected by P. berghei or P. falciparum, revealing a significant reduction of immune proteins compared to uninfected mosquitoes. This reduction is likely due to the association of these proteins with the surface of sporozoites within the mosquito salivary secretory cavities. The saliva protein profiles from mosquitoes infected with both Plasmodium species were remarkably similar, suggesting a conserved interaction between sporozoites and salivary glands. Our results provide a foundation for understanding the molecular interactions between Plasmodium sporozoites and mosquito salivary glands.
Collapse
Affiliation(s)
- Thiago Luiz Alves e Silva
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Sachi Kanatani
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Ana Beatriz Barletta Ferreira
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Cindi Schwartz
- Microscopy Unit, Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Octavio A.C. Talyuli
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Janet Olivas
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
- Present address: Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Bianca M. Nagata
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Zarna Rajeshkumar Pala
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
- Present address: Biological Sciences Graduate Program, University of Maryland, College Park, MD 20742
| | - Tales Pascini
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
- Present address: Sanaria Inc., 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Derron A. Alves
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Ming Zhao
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA
| | - Motoshi Suzuki
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Ln, Rockville, MD, 20852, USA
| | - Lilian P. Dorner
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
- German Center for Infection Research, partner site Heidelberg
| | - Isabelle Coppens
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Jose M.C. Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Photini Sinnis
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Joel Vega-Rodriguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
32
|
De A, Hoang CV, Escudero V, Armas AM, Echavarri‐Erasun C, González‐Guerrero M, Jordá L. Combating plant diseases through transition metal allocation. THE NEW PHYTOLOGIST 2025; 245:1833-1842. [PMID: 39707630 PMCID: PMC11798897 DOI: 10.1111/nph.20366] [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: 10/09/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
Understanding how plants fend-off invading microbes is essential for food security and the economy of large parts of the world. Consequently, a sustained and dedicated effort has been directed at unveiling how plants protect themselves from invading microbes. Major defense hormone signaling pathways have been characterized, the identity of many immune response-triggering molecules as well as many of their receptors have been determined, and the mechanisms of pathogen-host arms race are being studied. In recent years, evidence for a new layer of plant innate immunity involving transition metals has been brought forward. This would link plant metal nutrition with plant immune responses and open up possible new strategies for pathogen control involving metal fertilizers instead of pesticides. In this review, we outline our current understanding of metal-mediated plant immune response and indicate the future avenues of exploration of this topic.
Collapse
Affiliation(s)
- Aishee De
- Centro de Biotecnología y Genómica de Plantas (UPM‐INIA/CSIC), Universidad Politécnica de Madrid‐Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA)/Consejo Superior de Investigaciones Científicas (CSIC)28223Pozuelo de Alarcón (Madrid)Spain
| | - Cuong V. Hoang
- Centro de Biotecnología y Genómica de Plantas (UPM‐INIA/CSIC), Universidad Politécnica de Madrid‐Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA)/Consejo Superior de Investigaciones Científicas (CSIC)28223Pozuelo de Alarcón (Madrid)Spain
| | - Viviana Escudero
- Centro de Biotecnología y Genómica de Plantas (UPM‐INIA/CSIC), Universidad Politécnica de Madrid‐Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA)/Consejo Superior de Investigaciones Científicas (CSIC)28223Pozuelo de Alarcón (Madrid)Spain
| | - Alejandro M. Armas
- Centro de Biotecnología y Genómica de Plantas (UPM‐INIA/CSIC), Universidad Politécnica de Madrid‐Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA)/Consejo Superior de Investigaciones Científicas (CSIC)28223Pozuelo de Alarcón (Madrid)Spain
| | - Carlos Echavarri‐Erasun
- Centro de Biotecnología y Genómica de Plantas (UPM‐INIA/CSIC), Universidad Politécnica de Madrid‐Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA)/Consejo Superior de Investigaciones Científicas (CSIC)28223Pozuelo de Alarcón (Madrid)Spain
| | - Manuel González‐Guerrero
- Centro de Biotecnología y Genómica de Plantas (UPM‐INIA/CSIC), Universidad Politécnica de Madrid‐Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA)/Consejo Superior de Investigaciones Científicas (CSIC)28223Pozuelo de Alarcón (Madrid)Spain
- Departamento de Biotecnología‐Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de BiosistemasUniversidad Politécnica de Madrid28040MadridSpain
| | - Lucía Jordá
- Centro de Biotecnología y Genómica de Plantas (UPM‐INIA/CSIC), Universidad Politécnica de Madrid‐Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA)/Consejo Superior de Investigaciones Científicas (CSIC)28223Pozuelo de Alarcón (Madrid)Spain
- Departamento de Biotecnología‐Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de BiosistemasUniversidad Politécnica de Madrid28040MadridSpain
| |
Collapse
|
33
|
Shen Y, Li Y, Quan Y, Jin W, Wang Y, Liu B, Wang Y. Effects of environment regulating T4SS on virulence and adaptability of Streptococcus suis. ENVIRONMENTAL RESEARCH 2025; 268:120751. [PMID: 39778619 DOI: 10.1016/j.envres.2025.120751] [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: 10/21/2024] [Revised: 12/30/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025]
Abstract
Streptococcus suis (S. suis) represents a significant bacterial pathogen, with its zoonotic transmission from infected or deceased pigs to humans posing a serious threat to public health. The type IV secretion system (T4SS), a critical virulence factor of S. suis, is tightly regulated by diverse environmental conditions. This study explores the influence of environmental variables, including temperature, incubation duration, monosaccharides, and metal ions, on the regulation of T4SS in S. suis and its associated pathogenicity. Results revealed that T4SS expression peaked during the stabilization phase at 37 °C, with galactose markedly enhancing T4SS expression relative to glucose. Zinc ions specifically enhanced the expression of the T4SS effector SspA among various metal ions. Moreover, zinc exposure significantly augmented both T4SS and virulence gene expression capabilities in S. suis. Zinc-treated S. suis exhibited enhanced adhesion, invasion, and colonization capacity in Hep-2 cells, Raw264.7 cells, and mouse models. These findings provide a deeper comprehension of the environmental modulation of T4SS in S. suis, paving the way for advanced studies into its mechanisms of pathogenicity.
Collapse
Affiliation(s)
- Yamin Shen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China
| | - Yue Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China
| | - Yingying Quan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China
| | - Wenjie Jin
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China
| | - Baobao Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China.
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China.
| |
Collapse
|
34
|
Liu W, Yang T, Kong Y, Xie X, Ruan Z. Ureaplasma infections: update on epidemiology, antimicrobial resistance, and pathogenesis. Crit Rev Microbiol 2025; 51:317-347. [PMID: 38794781 DOI: 10.1080/1040841x.2024.2349556] [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: 12/06/2023] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024]
Abstract
Human Ureaplasma species are being increasingly recognized as opportunistic pathogens in human genitourinary tract infections, infertility, adverse pregnancy, neonatal morbidities, and other adult invasive infections. Although some general reviews have focused on the detection and clinical manifestations of Ureaplasma spp., the molecular epidemiology, antimicrobial resistance, and pathogenesis of Ureaplasma spp. have not been adequately explained. The purpose of this review is to offer valuable insights into the current understanding and future research perspectives of the molecular epidemiology, antimicrobial resistance, and pathogenesis of human Ureaplasma infections. This review summarizes the conventional culture and detection methods and the latest molecular identification technologies for Ureaplasma spp. We also reviewed the global prevalence and mechanisms of antibiotic resistance for Ureaplasma spp. Aside from regular antibiotics, novel antibiotics with outstanding in vitro antimicrobial activity against Ureaplasma spp. are described. Furthermore, we discussed the pathogenic mechanisms of Ureaplasma spp., including adhesion, proinflammatory effects, cytotoxicity, and immune escape effects, from the perspectives of pathology, related molecules, and genetics.
Collapse
Affiliation(s)
- Wenwen Liu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Ting Yang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Yingying Kong
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Xinyou Xie
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Zhi Ruan
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
35
|
Ahmad M, Aduru SV, Smith RP, Zhao Z, Lopatkin AJ. The role of bacterial metabolism in antimicrobial resistance. Nat Rev Microbiol 2025:10.1038/s41579-025-01155-0. [PMID: 39979446 DOI: 10.1038/s41579-025-01155-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 02/22/2025]
Abstract
The relationship between bacterial metabolism and antibiotic treatment is complex. On the one hand, antibiotics leverage cell metabolism to function. On the other hand, increasing research has highlighted that the metabolic state of the cell also impacts all aspects of antibiotic biology, from drug efficacy to the evolution of antimicrobial resistance (AMR). Given that AMR is a growing threat to the current global antibiotic arsenal and ability to treat infectious diseases, understanding these relationships is key to improving both public and human health. However, quantifying the contribution of metabolism to antibiotic activity and subsequent bacterial evolution has often proven challenging. In this Review, we discuss the complex and often bidirectional relationships between metabolism and the various facets of antibiotic treatment and response. We first summarize how antibiotics leverage metabolism for their function. We then focus on the converse of this relationship by specifically delineating the unique contribution of metabolism to three distinct but related arms of antibiotic biology: antibiotic efficacy, AMR evolution and AMR mechanisms. Finally, we note the relevance of metabolism in clinical contexts and explore the future of metabolic-based strategies for personalized antimicrobial therapies. A deeper understanding of these connections is crucial for the broader scientific community to address the growing crisis of AMR and develop future effective therapeutics.
Collapse
Affiliation(s)
- Mehrose Ahmad
- Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Sai Varun Aduru
- Department of Chemical Engineering, University of Rochester, Rochester, NY, USA
| | - Robert P Smith
- Cell Therapy Institute, Kiran Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
- Department of Medical Education, Kiran Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Zirui Zhao
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Allison J Lopatkin
- Department of Chemical Engineering, University of Rochester, Rochester, NY, USA.
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
36
|
Pan C, Jiang X, Wei J, Liu C, Zhang M, Gao C, Chen R, Yang C, Wang B, Yu M, Gan Y. Ameba-inspired strategy enhances probiotic efficacy via prebound nutrient supply. Nat Commun 2025; 16:1827. [PMID: 39979278 PMCID: PMC11842784 DOI: 10.1038/s41467-025-57071-7] [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: 08/10/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
Nutrient competition with indigenous microbes or pathogens presents a significant challenge for oral probiotic efficacy. To address this issue, we develop an ameba-inspired food-carrying strategy (AIFS) by prebinding ginger-derived exosome-like nanoparticles (GELNs) onto probiotics as food depots. AIFS enables probiotics to efficiently and exclusively consume GELNs in situ, even in the presence of competing bacteria. This results in up to 21 times higher uptake efficiency compared to unengineered probiotics, dramatically accelerating probiotic proliferation. Meanwhile, AIFS potentiates probiotics' resistance to multiple GI stressors. In a murine model of colitis, AIFS can improve the abundance of probiotics and inhibit pathogens, maintaining intestinal flora homeostasis. Additionally, it can upregulate the anti-inflammatory IL-10, reduce the proinflammatory IL-1β, and repair damaged intestinal mucus. Thereby, AIFS displays potently elevated prophylactic and therapeutic efficacy for colitis mice. This work provides a method for microbial engineering, with broad implications for microbiotherapy and gut health.
Collapse
Affiliation(s)
- Chao Pan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Xiuxian Jiang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Junchao Wei
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- School of Pharmacy, Henan University, Kaifeng, PR China
| | - Chang Liu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Min Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Chuan Gao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Rongrong Chen
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - Canyu Yang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Bingqi Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Miaorong Yu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Yong Gan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China.
- University of Chinese Academy of Sciences, Beijing, PR China.
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China.
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, Beijing, PR China.
| |
Collapse
|
37
|
Critchlow JM, Barraza JP, Munneke MJ, Krystofiak E, Green ER, Skaar EP. The interplay between Acinetobacter baumannii ZigA and SltB promotes zinc homeostasis and cell envelope integrity. Infect Immun 2025; 93:e0042224. [PMID: 39846731 PMCID: PMC11834433 DOI: 10.1128/iai.00422-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
Acinetobacter baumannii is an opportunistic human pathogen that acquires nutrient metals from the vertebrate host amid infection. During zinc (Zn) scarcity, A. baumannii upregulates the expression of the predicted Zn metallochaperone, zigA. Loss of zigA compromises fitness during Zn deficiency, highlighting its role in this condition. To assess the contribution of ZigA to Zn-deficient A. baumannii, a multiparallel transposon sequencing and genetic interaction mapping approach was used. Transposon insertions in A1S_3027, encoding a predicted soluble lytic transglycosylase that tailors the bacterial cell wall, were enriched in the Zn-starved ΔzigA transposon library. Based on previous studies as well as structural and sequence homology, we designated A1S_3027 as soluble lytic transglycosylase B (SltB). Further analyses revealed that inactivating sltB rescued ΔzigA fitness defects during Zn starvation. An A. baumannii ΔzigAΔsltB mutant demonstrated altered cell envelope structures and increased cellular permeability, highlighting the roles of ZigA and SltB in maintaining cell envelope integrity. Furthermore, these mutants exhibited heightened resistance to β-lactam antibiotics and other cell wall-targeting agents. Alterations in cell envelope integrity in the ΔzigAΔsltB mutant improved fitness in a murine pneumonia infection model, emphasizing the contribution of ZigA and SltB to A. baumannii pathogenesis. This study elucidates how functional interactions between ZigA and SltB modulate cell envelope integrity and pathogenesis of A. baumannii during Zn depletion. These findings reveal an interplay between metal homeostasis and cell envelope integrity, offering insights into how A. baumannii ZigA contributes to these critical cellular processes.
Collapse
Affiliation(s)
- Jeanette M. Critchlow
- Microbe-Host Interactions Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Juan P. Barraza
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Matthew J. Munneke
- Microbe-Host Interactions Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Evan Krystofiak
- Cell Imaging Shared Resource, Vanderbilt University, Nashville, Tennessee, USA
| | - Erin R. Green
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Eric P. Skaar
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
38
|
Bright R, Sivanantha S, Hayles A, Phuoc Ton T, Ninan N, Luo X, Vasilev K, Truong VK. In Vitro Assessment of Gallium Nanoalloy Hydrogels for Antimicrobial and Wound Healing Applications. ACS APPLIED BIO MATERIALS 2025; 8:1017-1026. [PMID: 39433303 DOI: 10.1021/acsabm.4c01182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Chronic and recurring wounds pose a significant challenge in modern healthcare, leading to substantial morbidity. These wounds allow pathogens to colonize, potentially resulting in local and systemic infections. Current interventions need to be revised and become increasingly less reliable due to the emergence of antibiotic resistance. In the present study, we aim to address these issues by fabricating hydrogels impregnated with gallium-based nanoalloys for their antimicrobial activity. Gallium liquid metal nanoparticles (approximately 100 nm in diameter) were alloyed in different combinations with bismuth and silver ions through a galvanic replacement reaction. These multimetallic hydrogels showed favorable antibacterial activity against the Gram-positive Staphylococcus aureus and the Gram-negative Pseudomonas aeruginosa, as observed with fluorescence microscopy and inhibition assays. The multimetallic hydrogels showed no toxicity against murine macrophages or human dermal fibroblasts and enhanced in vitro wound healing. The development of these innovative gallium-based hydrogels represents a promising strategy to combat chronic wounds and their associated complications, offering an effective alternative to current antimicrobial treatments amidst rising antibiotic resistance.
Collapse
Affiliation(s)
- Richard Bright
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Soroopan Sivanantha
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Andrew Hayles
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Tan Phuoc Ton
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Neethu Ninan
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Xuan Luo
- College of Science and Engineering, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Krasimir Vasilev
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Vi Khanh Truong
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| |
Collapse
|
39
|
Kumar R, Singh A, Srivastava A. Xenosiderophores: bridging the gap in microbial iron acquisition strategies. World J Microbiol Biotechnol 2025; 41:69. [PMID: 39939429 DOI: 10.1007/s11274-025-04287-w] [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: 12/03/2024] [Accepted: 02/04/2025] [Indexed: 02/14/2025]
Abstract
Microorganisms acquire iron from surrounding environment through specific iron chelators known as siderophores that can be of self-origin or synthesized by neighboring microbes. The latter are termed as xenosiderophores. The acquired iron supports their growth, survival, and pathogenesis. Various microorganisms possess the ability to utilize xenosiderophores, a mechanism popularly termed as 'siderophore piracy' besides synthesizing their own siderophores. This adaptability allows microorganisms to conserve energy by reducing the load of siderogenesis. Owing to the presence of xenosiderophore transport machinery, these microbial systems can be used for targeting antibiotics-siderophore conjugates to control pathogenesis and combat antimicrobial resistance. This review outlines the significance of xenosiderophore utilization for growth, stress management and virulence. Siderogenesis and the molecular mechanism of its uptake by related organisms have been discussed vividly. It focuses on potential applications like disease diagnostics, drug delivery, and combating antibiotic resistance. In brief, this review highlights the importance of xenosiderophores projecting them beyond their role as mere iron chelators.
Collapse
Affiliation(s)
- Ravinsh Kumar
- Department of Life Science, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya, Bihar, India
| | - Ashutosh Singh
- Department of Life Science, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya, Bihar, India
| | - Amrita Srivastava
- Department of Life Science, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya, Bihar, India.
| |
Collapse
|
40
|
LeGrand EK. Beyond nutritional immunity: immune-stressing challenges basic paradigms of immunometabolism and immunology. Front Nutr 2025; 12:1508767. [PMID: 40013164 PMCID: PMC11860096 DOI: 10.3389/fnut.2025.1508767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/31/2025] [Indexed: 02/28/2025] Open
Abstract
Pathogens have the well-known advantage of rapid evolution due to short generation times and large populations. However, pathogens have the rarely noted disadvantage of the vulnerability to stress involved in proliferation as well as being localized. Presented here are numerous new paradigms in immunology, and especially immunometabolism, which are derived from examining how hosts capitalize on pathogen vulnerabilities to stress. Universally, proliferation requires both resources and synthesis, which are vulnerable to resource-limiting stress and damaging/noxious stress, respectively. Pathogens are particularly vulnerable to stress at the time when they are most threatening-when they are proliferating. Since immune cells actively controlling pathogens (effector cells) typically do not proliferate at infected sites, there is a "stress vulnerability gap" wherein proliferating pathogens are more vulnerable to any type of stress than are the attacking effector cells. Hosts actively stress vulnerable proliferating pathogens by restricting resources (resource-limiting stress) and generating noxious waste products (damaging/disruptive stress) in a fundamental defense here-in termed "immune-stressing." While nutritional immunity emphasizes denying pathogens micronutrients, immune-stressing extends the concept to restricting all resources, especially glucose and oxygen, coupled with the generation of noxious metabolic products such as lactic acid, reactive oxygen species (ROS), and heat to further harm or stress the pathogens. At present much of the field of immunometabolism centers on how nutrition and metabolism regulate immune function, a central feature being the inefficient use of glucose via aerobic glycolysis (with much lactate/lactic acid production) by effector immune cells. In contrast, immune-stressing emphasizes how the immune system uses nutrition and metabolism to control infections. Immune-stressing addresses effector cell glycolysis at the infected site by noting that the high uptake of glucose linked with high output of lactic acid is an ideal double-pronged stressor targeting proliferating pathogens. Once the basic vulnerability of pathogen proliferation is recognized, numerous other paradigms of immunometabolism, and immunology as a whole, are challenged.
Collapse
Affiliation(s)
- Edmund K. LeGrand
- Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, United States
| |
Collapse
|
41
|
Rios-Delgado G, McReynolds AKG, Pagella EA, Norambuena J, Briaud P, Zheng V, Munneke MJ, Kim J, Racine H, Carroll RK, Zelzion E, Skaar E, Bose JL, Parker D, Lalaouna D, Boyd JM. The Staphylococcus aureus non-coding RNA IsrR regulates TCA cycle activity and virulence. Nucleic Acids Res 2025; 53:gkae1243. [PMID: 39704109 DOI: 10.1093/nar/gkae1243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 11/08/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024] Open
Abstract
Staphylococcus aureus has evolved mechanisms to cope with low iron (Fe) availability in host tissues. Staphylococcus aureus uses the ferric uptake transcriptional regulator (Fur) to sense titers of cytosolic Fe. Upon Fe depletion, apo-Fur relieves transcriptional repression of genes utilized for Fe uptake. We demonstrate that an S. aureus Δfur mutant has decreased expression of acnA, which codes for the Fe-dependent enzyme aconitase. This prevents the Δfur mutant from growing with amino acids as sole carbon and energy sources. We used a suppressor screen to exploit this phenotype and determined that a mutation that decreases the transcription of isrR, which produces a regulatory RNA, increased acnA expression, thereby enabling growth. Directed mutation of bases predicted to facilitate the interaction between the acnA transcript and IsrR, decreased the ability of IsrR to control acnA expression in vivo and IsrR bound to the acnA transcript in vitro. IsrR also bound transcripts coding the alternate tricarboxylic acid cycle proteins sdhC, mqo, citZ and citM. Whole-cell metal analyses suggest that IsrR promotes Fe uptake and increases intracellular Fe not ligated by macromolecules. Lastly, we determined that Fur and IsrR promote infection using murine skin and acute pneumonia models.
Collapse
Affiliation(s)
- Gustavo Rios-Delgado
- Department of Biochemistry and Microbiology, Rutgers, the State University of New Jersey, 76 Lipman Dr, New Brunswick, NJ 08901, USA
| | - Aubrey K G McReynolds
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Emma A Pagella
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Javiera Norambuena
- Department of Biochemistry and Microbiology, Rutgers, the State University of New Jersey, 76 Lipman Dr, New Brunswick, NJ 08901, USA
| | - Paul Briaud
- Department of Biological Sciences, Ohio University, 7 Depot St, Athens, OH 45701, USA
| | - Vincent Zheng
- Department of Biochemistry and Microbiology, Rutgers, the State University of New Jersey, 76 Lipman Dr, New Brunswick, NJ 08901, USA
| | - Matthew J Munneke
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, USA
| | - Jisun Kim
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Hugo Racine
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR9002, 15 rue René Descartes, Strasbourg 67000, France
| | - Ronan K Carroll
- Department of Biological Sciences, Ohio University, 7 Depot St, Athens, OH 45701, USA
| | - Ehud Zelzion
- Office of Advanced Research Computing, Rutgers University, 96 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Eric Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, USA
| | - Jeffrey L Bose
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Dane Parker
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - David Lalaouna
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR9002, 15 rue René Descartes, Strasbourg 67000, France
| | - Jeffrey M Boyd
- Department of Biochemistry and Microbiology, Rutgers, the State University of New Jersey, 76 Lipman Dr, New Brunswick, NJ 08901, USA
| |
Collapse
|
42
|
Zeng W, Wang F, Cui Z, Zhang Y, Li Y, Li N, Mao Z, Zhang H, Liu Y, Miao Y, Sun S, Cai Y, Xiong B. Inhibition of ferroptosis counteracts the advanced maternal age-induced oocyte deterioration. Cell Death Differ 2025:10.1038/s41418-025-01456-0. [PMID: 39910323 DOI: 10.1038/s41418-025-01456-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 01/13/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025] Open
Abstract
Ferroptosis, a recently discovered form of programmed cell death triggered by the excessive accumulation of iron-dependent lipid peroxidation products, plays a critical role in the development of various diseases. However, whether it is involved in the age-related decline in oocyte quality remains unexplored. Here, we took advantage of nano-proteomics to uncover that reduced ferritin heavy chain (Fth1) level is a major cause leading to the occurrence of ferroptosis in aged oocytes. Specifically, induction of ferroptosis in young oocytes by its activators RSL3 and FAC, or knockdown of Fth1 all phenocopied the meiotic defects observed in aged oocytes, including failed oocyte meiotic maturation, aberrant cytoskeleton dynamics, as well as impaired mitochondrial function. Transcriptome analysis showed that knockdown of Fth1 affected meiosis-related and aging-related pathways in oocytes. Conversely, inhibition of ferroptosis by its inhibitors or expression of Fth1 improved the quality of aged oocytes. We also validated the effects of ferroptosis on the porcine oocyte quality in vitro. Altogether, we demonstrate the contribution of ferroptosis to the age-induced oocyte defects and evidence that inhibition of ferroptosis might be a feasible strategy to ameliorate the reproductive outcomes of female animals at an advanced age.
Collapse
Affiliation(s)
- Wenjun Zeng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Feixue Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zhaokang Cui
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yu Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yu Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Na Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zipeng Mao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Hanwen Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yiting Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yilong Miao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Shaochen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yafei Cai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Bo Xiong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.
- College of Animal Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
43
|
Bhowmik S, Pathak A, Pandey S, Devnath K, Sett A, Jyoti N, Bhando T, Akhter J, Chugh S, Singh R, Sharma TK, Pathania R. Acinetobacter baumannii represses type VI secretion system through a manganese-dependent small RNA-mediated regulation. mBio 2025; 16:e0302524. [PMID: 39704509 PMCID: PMC11796373 DOI: 10.1128/mbio.03025-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024] Open
Abstract
Type VI secretion system (T6SS) is utilized by many Gram-negative bacteria to eliminate competing bacterial species and manipulate host cells. Acinetobacter baumannii ATCC 17978 utilizes T6SS at the expense of losing pAB3 plasmid to induce contact-dependent killing of competitor microbes, resulting in the loss of antibiotic resistance carried by pAB3. However, the regulatory network associated with T6SS in A. baumannii remains poorly understood. Here, we identified an Mn2+-dependent post-transcriptional regulation of T6SS mediated by a bonafide small RNA, AbsR28. A. baumannii utilizes MumT, an Mn2+-uptake inner membrane transporter, for the uptake of extracellular Mn2+ during oxidative stress. We demonstrate that the abundance of intracellular Mn2+ enables complementary base pairing of AbsR28-tssM mRNA (that translates to TssM, one of the vital inner membrane components of T6SS), inducing RNase E-mediated degradation of tssM mRNA and resulting in T6SS repression. Thus, AbsR28 mediates a crosstalk between MumT and T6SS in A. baumannii.IMPORTANCESmall RNAs (sRNAs) are identified as critical components within the bacterial regulatory networks involved in fine regulation of virulence-associated factors. The sRNA-mediated regulation of type VI secretion system (T6SS) in Acinetobacter baumannii was unchartered. Previously, it was demonstrated that A. baumannii ATCC 17978 cells switch from T6- to T6+ phenotype, resulting in the loss of antibiotic resistance conferred by plasmid pAB3. Furthermore, the derivatives of pAB3 found in recent clinical isolates of A. baumannii harbor expanded antibiotic resistance genes and multiple determinants for virulence factors. Hence, the loss of this plasmid for T6SS activity renders A. baumannii T6+ cells susceptible to antibiotics and compromises their virulence. Our findings show how A. baumannii tends to inactivate T6SS through an sRNA-mediated regulation that relies on Mn2+ and retains pAB3 during infection to retain antibiotic resistance genes carried on the plasmid.
Collapse
Affiliation(s)
- Somok Bhowmik
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Avik Pathak
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Shivam Pandey
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Kuldip Devnath
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Abhiroop Sett
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Nishant Jyoti
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Timsy Bhando
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Jawed Akhter
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Saurabh Chugh
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Ramandeep Singh
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Tarun Kumar Sharma
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Ranjana Pathania
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
- Center of Excellence in Disaster Mitigation and Management, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| |
Collapse
|
44
|
Duman O, Kuznetsova A, Levanon NL, Grupper M, Ersoy AA, Acar B, Kessel A, Ben‐Tal N, Lewinson O, Haliloglu T. Computational and experimental mapping of the allosteric network of two manganese ABC transporters. Protein Sci 2025; 34:e70039. [PMID: 39887508 PMCID: PMC11779740 DOI: 10.1002/pro.70039] [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: 04/20/2024] [Revised: 12/27/2024] [Accepted: 01/07/2025] [Indexed: 02/01/2025]
Abstract
Transition metals (e.g., Fe2/3+, Zn2+, Mn2+) are essential enzymatic cofactors in all organisms. Their environmental scarcity led to the evolution of high-affinity uptake systems. Our research focuses on two bacterial manganese ABC importers, Streptococcus pneumoniae PsaBC and Bacillus anthracis MntBC, both critical for virulence. Both importers share a similar homodimeric structure, where each protomer comprises a transmembrane domain (TMD) linked to a cytoplasmic nucleotide-binding domain (NBD). Due to their size and slow turnover rates, the utility of conventional molecular simulation approaches to reveal functional dynamics is limited. Thus, we employed a novel, computationally efficient method integrating Gaussian Network Models (GNM) with information theory Transfer Entropy (TE) calculations. Our calculations are in remarkable agreement with previous functional studies. Furthermore, based on the calculations, we generated 10 point-mutations and experimentally tested their effects, finding excellent concordance between computational predictions and experimental results. We identified "allosteric hotspots" in both transporters, in the transmembrane translocation pathway, at the coupling helices linking the TMDs and NBDs, and in the ATP binding sites. In both PsaBC and MntBC, we observed bi-directional information flow between the two TMDs, with minimal allosteric transmission to the NBDs. Conversely, the NBDs exhibited almost no NBD-NBD allosteric crosstalk but showed pronounced information flow from the NBD of one protomer towards the TMD of the other protomer. This unique allosteric "footprint" distinguishes ABC importers of transition metals from other members of the ABC transporter superfamily establishing them as a distinct functional class. This study offers the first comprehensive insight into the conformational dynamics of these vital virulence determinants, providing potential avenues for developing urgently needed novel antibacterial agents.
Collapse
Affiliation(s)
- Ozge Duman
- Department of Chemical EngineeringBogazici UniversityIstanbulTurkey
- Polymer Research CenterBogazici UniversityIstanbulTurkey
| | - Anastasiya Kuznetsova
- Department of Molecular MicrobiologyBruce and Ruth Rappaport Faculty of Medicine, Technion‐Israel Institute of TechnologyHaifaIsrael
| | - Nurit Livnat Levanon
- Department of Molecular MicrobiologyBruce and Ruth Rappaport Faculty of Medicine, Technion‐Israel Institute of TechnologyHaifaIsrael
| | - Moti Grupper
- Infectious Disease UnitRambam Health Care CampusHaifaIsrael
| | - Akarun Ayca Ersoy
- Department of Chemical EngineeringBogazici UniversityIstanbulTurkey
- Polymer Research CenterBogazici UniversityIstanbulTurkey
| | - Burcin Acar
- Polymer Research CenterBogazici UniversityIstanbulTurkey
| | - Amit Kessel
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life SciencesTel‐Aviv UniversityTel‐AvivIsrael
| | - Nir Ben‐Tal
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life SciencesTel‐Aviv UniversityTel‐AvivIsrael
| | - Oded Lewinson
- Department of Molecular MicrobiologyBruce and Ruth Rappaport Faculty of Medicine, Technion‐Israel Institute of TechnologyHaifaIsrael
| | - Turkan Haliloglu
- Department of Chemical EngineeringBogazici UniversityIstanbulTurkey
- Polymer Research CenterBogazici UniversityIstanbulTurkey
| |
Collapse
|
45
|
Sweet MJ, Ramnath D, Singhal A, Kapetanovic R. Inducible antibacterial responses in macrophages. Nat Rev Immunol 2025; 25:92-107. [PMID: 39294278 DOI: 10.1038/s41577-024-01080-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/20/2024]
Abstract
Macrophages destroy bacteria and other microorganisms through phagocytosis-coupled antimicrobial responses, such as the generation of reactive oxygen species and the delivery of hydrolytic enzymes from lysosomes to the phagosome. However, many intracellular bacteria subvert these responses, escaping to other cellular compartments to survive and/or replicate. Such bacterial subversion strategies are countered by a range of additional direct antibacterial responses that are switched on by pattern-recognition receptors and/or host-derived cytokines and other factors, often through inducible gene expression and/or metabolic reprogramming. Our understanding of these inducible antibacterial defence strategies in macrophages is rapidly evolving. In this Review, we provide an overview of the broad repertoire of antibacterial responses that can be engaged in macrophages, including LC3-associated phagocytosis, metabolic reprogramming and antimicrobial metabolites, lipid droplets, guanylate-binding proteins, antimicrobial peptides, metal ion toxicity, nutrient depletion, autophagy and nitric oxide production. We also highlight key inducers, signalling pathways and transcription factors involved in driving these different antibacterial responses. Finally, we discuss how a detailed understanding of the molecular mechanisms of antibacterial responses in macrophages might be exploited for developing host-directed therapies to combat antibiotic-resistant bacterial infections.
Collapse
Affiliation(s)
- Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
| | - Divya Ramnath
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Amit Singhal
- Infectious Diseases Labs (ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Ronan Kapetanovic
- INRAE, Université de Tours, Infectiologie et Santé Publique (ISP), Nouzilly, France
| |
Collapse
|
46
|
Stark FG, Torii-Karch M, Yuvaraj S, Bonometti L, Gladieux P, Glass NL, Krasileva K. Molecular Insights into Fungal Innate Immunity Using the Neurospora crassa - Pseudomonas syringae Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.633611. [PMID: 39896647 PMCID: PMC11785063 DOI: 10.1101/2025.01.22.633611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Recent comparative genomics and mechanistic analyses support the existence of a fungal immune system. Fungi encode genes with features similar to non-self recognition systems in plants, animals, and bacteria. However, limited functional or mechanistic evidence exists for the surveillance-system recognition of heterologous microbes in fungi. We found that Neurospora species coexist with Pseudomonas in their natural environment. We leveraged two model organisms, Neurospora crassa and Pseudomonas syringae DC3000 (PSTDC3000) to observe immediate fungal responses to bacteria. PSTDC3000 preferentially surrounds N. crassa cells on a solid surface, causing environmental dependent growth responses, bacterial proliferation and varying fungal fitness. Specifically, the Type III secretion system (T3SS) ΔhrcC mutant of PSTDC3000 colonized N. crassa hyphae less well. To dissect initial cellular signaling events within the population of germinated asexual spores (germlings), we performed transcriptomics on N. crassa after PSTDC3000 inoculation. Upon contact with live bacteria, a subpopulation of fungal germlings initiate a response as early as ten minutes post-contact revealing transcriptional differentiation of Reactive Oxygen Species (ROS) mechanisms, trace metal warfare, cell wall remodeling dynamics, multidrug-efflux transporters, secondary metabolite synthesis, and excretion. We dissected mutants of plausible receptors, signaling pathways, and responses that N. crassa uses to detect and mount a defense against PSTDC3000 and found seven genes that influence resistant and susceptibility phenotypes of N. crassa to bacterial colonization. Mutants in genes encoding a ctr copper transporter ( tcu-1 ), ferric reductase ( fer-1 ), superoxide reductase ( sod-2 ), multidrug resistance transporter ( mdr-6 ), a secreted lysozyme-Glycoside hydrolase ( lyz ) and the Woronin body tether leashin (NCU02793, lah-1 and lah-2 ) showed a significant reduction of growth in the presence of bacteria, allowing the bacteria to fully take over the fungal mycelium faster than wildtype. In this study we provide a bacterial-fungal model system within Dikarya that allows us to begin to dissect signaling pathways of the putative fungal immune system.
Collapse
|
47
|
Helmann JD. Metals in Motion: Understanding Labile Metal Pools in Bacteria. Biochemistry 2025; 64:329-345. [PMID: 39755956 PMCID: PMC11755726 DOI: 10.1021/acs.biochem.4c00726] [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: 10/28/2024] [Revised: 12/02/2024] [Accepted: 12/13/2024] [Indexed: 01/07/2025]
Abstract
Metal ions are essential for all life. In microbial cells, potassium (K+) is the most abundant cation and plays a key role in maintaining osmotic balance. Magnesium (Mg2+) is the dominant divalent cation and is required for nucleic acid structure and as an enzyme cofactor. Microbes typically require the transition metals manganese (Mn), iron (Fe), copper (Cu), and zinc (Zn), although the precise set of metal ions needed to sustain life is variable. Intracellular metal pools can be conceptualized as a chemically complex mixture of rapidly exchanging (labile) ions, complemented by those reservoirs that exchange slowly relative to cell metabolism (sequestered). Labile metal pools are buffered by transient interactions with anionic metabolites and macromolecules, with the ribosome playing a major role. Sequestered metal pools include many metalloproteins, cofactors, and storage depots, with some pools redeployed upon metal depletion. Here, I review the size, composition, and dynamics of intracellular metal pools and highlight the major gaps in understanding.
Collapse
Affiliation(s)
- John D. Helmann
- Department of Microbiology, Cornell University, Ithaca, New York 14853-8101, United States
| |
Collapse
|
48
|
Brunson DN, Manzer H, Smith AB, Zackular JP, Kitten T, Lemos JA. A Novel Heme-Degrading Enzyme that Regulates Heme and Iron Homeostasis and Promotes Virulence in Enterococcus faecalis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.20.633879. [PMID: 39896487 PMCID: PMC11785130 DOI: 10.1101/2025.01.20.633879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Enterococcus faecalis, a gut commensal, is a leading cause of opportunistic infections. Its virulence is linked to its ability to thrive in hostile environments, which includes host-imposed metal starvation. We recently showed that E. faecalis evades iron starvation using five dedicated transporters that collectively scavenge iron from host tissues and other iron-deprived conditions. Interestingly, heme, the most abundant source of iron in the human body, supported growth of a strain lacking all five iron transporters (Δ5Fe). To release iron from heme, many bacterial pathogens utilize heme oxygenase enzymes to degrade the porphyrin that coordinates the iron ion of heme. Although E. faecalis lacks these enzymes, bioinformatics revealed a potential ortholog of the anaerobic heme-degrading enzyme anaerobilin synthase, found in Escherichia coli and a few other Gram-negative bacteria. Here, we demonstrated that deletion of OG1RF_RS05575 in E. faecalis (ΔRS05575) or in the Δ5Fe background (Δ5FeΔRS05575) led to intracellular heme accumulation and hypersensitivity under anaerobic conditions, suggesting RS05575 encodes an anaerobilin synthase, the first of its kind described in Gram-positive bacteria. Additionally, deletion of RS05575, either alone or in the Δ5Fe background, impaired E. faecalis colonization in the mouse gastrointestinal tract and virulence in mouse peritonitis and rabbit infective endocarditis models. These results reveal that RS05575 is responsible for anaerobic degradation of heme and identify this relatively new enzyme class as a novel factor in bacterial pathogenesis. Findings from this study are likely to have broad implications, as homologues of RS05575 are found in other Gram-positive facultative anaerobes.
Collapse
Affiliation(s)
- Debra N. Brunson
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Hader Manzer
- Division of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Alexander B. Smith
- Division of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Joseph P. Zackular
- Division of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Microbial Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Todd Kitten
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298-0566, USA
| | - José A. Lemos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| |
Collapse
|
49
|
Jiao M, He W, Ouyang Z, Yu Q, Zhang J, Qin Q, Wang R, Guo X, Liu R, He X, Hwang PM, Zheng F, Wen Y. Molybdate uptake interplay with ROS tolerance modulates bacterial pathogenesis. SCIENCE ADVANCES 2025; 11:eadq9686. [PMID: 39813328 PMCID: PMC11734730 DOI: 10.1126/sciadv.adq9686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
The rare metal element molybdenum functions as a cofactor in molybdoenzymes that are essential to life in almost all living things. Molybdate can be captured by the periplasmic substrate-binding protein ModA of ModABC transport system in bacteria. We demonstrate that ModA plays crucial roles in growth, multiple metabolic pathways, and ROS tolerance in Acinetobacter baumannii. Crystal structures of molybdate-coordinated A. baumannii ModA show a noncanonical disulfide bond with a conformational change between reduced and oxidized states. Disulfide bond formation reduced binding affinity to molybdate by two orders of magnitude and contributes to its substrate preference. ModA-mediated molybdate binding was important for A. baumannii infection in a murine pneumonia model. Together, our study sheds light on the structural and functional diversity of molybdate uptake and highlights a potential target for antibacterial development.
Collapse
Affiliation(s)
- Min Jiao
- Center for Microbiome Research of Med-X Institute, Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China
| | - Wenbo He
- Center for Microbiome Research of Med-X Institute, Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China
| | - Zhenlin Ouyang
- Center for Microbiome Research of Med-X Institute, Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China
| | - Qinyue Yu
- Center for Microbiome Research of Med-X Institute, Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China
| | - Jiaxin Zhang
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Qian Qin
- Center for Microbiome Research of Med-X Institute, Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China
| | - Ruochen Wang
- Center for Microbiome Research of Med-X Institute, Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China
| | - Xiaolong Guo
- Center for Microbiome Research of Med-X Institute, Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China
| | - Ruihan Liu
- Center for Microbiome Research of Med-X Institute, Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China
| | - Xiaoyu He
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Peter M. Hwang
- Departments of Medicine and Biochemistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Fang Zheng
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Yurong Wen
- Center for Microbiome Research of Med-X Institute, Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
50
|
Jones K, de Brito CB, Byndloss MX. Metabolic tug-of-war: Microbial metabolism shapes colonization resistance against enteric pathogens. Cell Chem Biol 2025; 32:46-60. [PMID: 39824157 DOI: 10.1016/j.chembiol.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 11/06/2024] [Accepted: 12/11/2024] [Indexed: 01/20/2025]
Abstract
A widely recognized benefit of gut microbiota is that it provides colonization resistance against enteric pathogens. The gut microbiota and their products can protect the host from invading microbes directly via microbe-pathogen interactions and indirectly by host-microbiota interactions, which regulate immune system function. In contrast, enteric pathogens have evolved mechanisms to utilize microbiota-derived metabolites to overcome colonization resistance and increase their pathogenic potential. This review will focus on recent studies of metabolism-mediated mechanisms of colonization resistance and virulence strategies enteric pathogens use to overcome them, along with how induction of inflammation by pathogenic bacteria changes the landscape of the gut and enables alternative metabolic pathways. We will focus on how intestinal pathogens counteract the protective effects of microbiota-derived metabolites to illustrate the growing appreciation of how metabolic factors may serve as crucial virulence determinants and overcome colonization resistance.
Collapse
Affiliation(s)
- Katerina Jones
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Camila Bernardo de Brito
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mariana Xavier Byndloss
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Howard Hughes Medical Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute of Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|