1
|
Swain J, Askenasy I, Rudland Nazeer R, Ho PM, Labrini E, Mancini L, Xu Q, Hollendung F, Sheldon I, Dickson C, Welch A, Agbamu A, Godlee C, Welch M. Pathogenicity and virulence of Pseudomonas aeruginosa: Recent advances and under-investigated topics. Virulence 2025; 16:2503430. [PMID: 40353451 PMCID: PMC12087490 DOI: 10.1080/21505594.2025.2503430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/23/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025] Open
Abstract
Pseudomonas aeruginosa is a model for the study of quorum sensing, protein secretion, and biofilm formation. Consequently, it has become one of the most intensely reviewed pathogens, with many excellent articles in the current literature focusing on these aspects of the organism's biology. Here, though, we aim to take a slightly different approach and consider some less well appreciated (but nonetheless important) factors that affect P. aeruginosa virulence. We start by reminding the reader of the global importance of P. aeruginosa infection and that the "virulome" is very niche-specific. Overlooked but obvious questions such as "what prevents secreted protein products from being digested by co-secreted proteases?" are discussed, and we suggest how the nutritional preference(s) of the organism might dictate its environmental reservoirs. Recent studies identifying host genes associated with genetic predisposition towards P. aeruginosa infection (and even infection by specific P. aeruginosa strains) and the role(s) of intracellular P. aeruginosa are introduced. We also discuss the fact that virulence is a high-risk strategy and touch on how expression of the two main classes of virulence factors is regulated. A particular focus is on recent findings highlighting how nutritional status and metabolism are as important as quorum sensing in terms of their impact on virulence, and how co-habiting microbial species at the infection site impact on P. aeruginosa virulence (and vice versa). It is our view that investigation of these issues is likely to dominate many aspects of research into this WHO-designated priority pathogen over the next decade.
Collapse
Affiliation(s)
- Jemima Swain
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Isabel Askenasy
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | - Pok-Man Ho
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Edoardo Labrini
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | - Qingqing Xu
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | | | - Camilla Dickson
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Amelie Welch
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Adam Agbamu
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Camilla Godlee
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Martin Welch
- Department of Biochemistry, Cambridge University, Cambridge, UK
| |
Collapse
|
2
|
Chen YT, Lohia GK, Chen S, Liu Z, Wong Fok Lung T, Wang C, Riquelme SA. A host-pathogen metabolic synchrony that facilitates disease tolerance. Nat Commun 2025; 16:3729. [PMID: 40253414 PMCID: PMC12009439 DOI: 10.1038/s41467-025-59134-1] [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: 10/10/2024] [Accepted: 04/10/2025] [Indexed: 04/21/2025] Open
Abstract
Disease tolerance mitigates organ damage from non-resolving inflammation during persistent infections, yet its underlying mechanisms remain unclear. Here we show, in a Pseudomonas aeruginosa pneumonia mouse model, that disease tolerance depends on the mitochondrial metabolite itaconate, which mediates cooperative host-pathogen interactions. In P. aeruginosa, itaconate modifies key cysteine residues in TCA cycle enzymes critical for succinate metabolism, inducing bioenergetic stress and promoting the formation biofilms that are less immunostimulatory and allow the bacteria to integrate into the local microbiome. Itaconate incorporates into the central metabolism of the biofilm, driving exopolysaccharide production-particularly alginate-which amplifies airway itaconate signaling. This itaconate-alginate interplay limits host immunopathology by enabling pulmonary glutamine assimilation, activating glutaminolysis, and thereby restrain detrimental inflammation caused by the inflammasome. Clinical sample analysis reveals that P. aeruginosa adapts to this metabolic environment through compensatory mutations in the anti-sigma-factor mucA, which restore the succinate-driven bioenergetics and disrupt the metabolic synchrony essential for sustaining disease tolerance.
Collapse
Affiliation(s)
- Ying-Tsun Chen
- Department of Pediatrics, Columbia University, New York, NY, USA
| | | | - Samantha Chen
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Zihua Liu
- Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | | | - Chu Wang
- Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | | |
Collapse
|
3
|
Lohia GK, Riquelme SA. Influence of cell bioenergetics on host-pathogen interaction in the lung. Front Immunol 2025; 16:1549293. [PMID: 40248701 PMCID: PMC12003392 DOI: 10.3389/fimmu.2025.1549293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
Pulmonary diseases, arising from infections caused by bacteria, fungi, and viruses, or stemming from underlying genetic factors are one of the leading causes of mortality in humans, accounting for millions of deaths every year. At the onset of pulmonary diseases, crucial roles are played by phagocytic immune cells, particularly tissue-resident macrophages, in regulating the immune response at the mucosal barrier. Recent strides have illuminated the pivotal role of host bioenergetics modulated by metabolites derived from both pathogens and hosts in influencing the pathophysiology of major organs. Their influence extends to processes such as the infiltration of immune cells, activation of macrophages, and the polarization phenomenon. Furthermore, host-derived metabolites, such as itaconate, contribute to the promotion of anti-inflammatory responses, thereby preventing immunopathology and facilitating the preservation of mucosal niches to thrive for the long-term. This review explores recent advancements in the field of immunometabolism, with a particular emphasis on the intricacies of disease progression in pulmonary infections caused by bacteria such as P. aeruginosa, M. tuberculosis and S. aureus and fungi like C. albicans.
Collapse
|
4
|
Qu J, Yin L, Qin S, Sun X, Gong X, Li S, Pan X, Jin Y, Cheng Z, Jin S, Wu W. Identification of the Pseudomonas aeruginosa AgtR-CspC-RsaL pathway that controls Las quorum sensing in response to metabolic perturbation and Staphylococcus aureus. PLoS Pathog 2025; 21:e1013054. [PMID: 40198682 PMCID: PMC12051497 DOI: 10.1371/journal.ppat.1013054] [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: 10/17/2024] [Revised: 04/21/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025] Open
Abstract
Environmental metabolites and metabolic pathways significantly influence bacterial pathogenesis and interspecies competition. We previously discovered that a mutation in the triosephosphate isomerase gene, tpiA, in Pseudomonas aeruginosa led to defective type III secretion and increased susceptibility to aminoglycoside antibiotics. In this study, we found that the tpiA mutation enhances the Las quorum sensing system due to reduced translation of the negative regulator RsaL. Further investigations demonstrated an upregulation of CspC, a CspA family protein that represses rsaL translation. DNA pull-down assay, along with genetic studies, revealed the role of AgtR in regulating cspC transcription. AgtR is known to regulate pyocyanin production in response to N-acetylglucosamine (GlcNAc), contributing to competition against Staphylococcus aureus. We demonstrated that CspC activates the Las quorum sensing system and subsequent pyocyanin production in response to GlcNAc and S. aureus. Overall, our results elucidate the AgtR-CspC-RsaL-LasI pathway that regulates bacterial virulence factors and its role in competition against S. aureus.
Collapse
Affiliation(s)
- Junze Qu
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Liwen Yin
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Shanhua Qin
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaomeng Sun
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Xuetao Gong
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Shouyi Li
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, National Key laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key laboratory of Digestive Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, People’s Republic of China
| | - Xiaolei Pan
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yongxin Jin
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhihui Cheng
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Shouguang Jin
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Weihui Wu
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
5
|
Dimitriew W, Schuster S. Dynamic optimization elucidates higher-level pathogenicity strategies of Pseudomonas aeruginosa. MICROLIFE 2025; 6:uqaf005. [PMID: 40182079 PMCID: PMC11967335 DOI: 10.1093/femsml/uqaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 04/05/2025]
Abstract
Multiple dangerous pathogens from the World Health Organization's priority list possess a plethora of virulence components, including the ability to survive inside macrophages. Often, the pathogens rely on a multi-layered defence strategy in order to defend themselves against the immune system. Here, a minimal model is proposed to study such a strategy. By way of example, we consider the interaction between Pseudomonas aeruginosa and the human host, in which the host and the pathogen counter each other in a back-and-forth interaction. In particular, the pathogen attacks the host, macrophages of the host engulf the pathogen and reduce its access to glucose, the pathogen activates the glyoxylate shunt, which is started by the enzyme isocitrate lyase (Icl), the host inhibits it by itaconic acid, and the pathogen metabolizes itaconic acid using the enzyme succinyl-CoA:itaconate CoA transferase (Ict). The flux through the glyoxylate shunt allows the pathogen to avoid carbon loss and oxidative stress. These functions are of utmost importance inside a phagolysosome. Therefore, the pathogen needs to allocate its limited protein resource between the enzymes Icl and Ict in order to maximize the time integral of a flux through the enzyme Icl. We use both random search and dynamic optimization to identify the enzyme Ict as a cost-effective means of counter-counter-counter-defence and as a possible drug target during the early phase of infection.
Collapse
Affiliation(s)
- Wassili Dimitriew
- Department of Bioinformatics, Friedrich Schiller University of Jena, 07743 Jena, Germany
| | - Stefan Schuster
- Department of Bioinformatics, Friedrich Schiller University of Jena, 07743 Jena, Germany
| |
Collapse
|
6
|
Wu S, Bu X, Chen D, Wu X, Wu H, Caiyin Q, Qiao J. Molecules-mediated bidirectional interactions between microbes and human cells. NPJ Biofilms Microbiomes 2025; 11:38. [PMID: 40038292 PMCID: PMC11880406 DOI: 10.1038/s41522-025-00657-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
Complex molecules-mediated interactions, which are based on the bidirectional information exchange between microbes and human cells, enable the defense against diseases and health maintenance. Recently, diverse single-direction interactions based on active metabolites, immunity factors, and quorum sensing signals have largely been summarized separately. In this review, according to a simplified timeline, we proposed the framework of Molecules-mediated Bidirectional Interactions (MBI) between microbe and humans to decipher and understand their intricate interactions systematically. About the microbe-derived interactions, we summarized various molecules, such as short-chain fatty acids, bile acids, tryptophan catabolites, and quorum sensing molecules, and their corresponding human receptors. Concerning the human-derived interactions, we reviewed the effect of human molecules, including hormones, cytokines, and other circulatory metabolites on microbial characteristics and phenotypes. Finally, we discussed the challenges and trends for developing and deciphering molecule-mediated bidirectional interactions and their potential applications in the guard of human health.
Collapse
Affiliation(s)
- Shengbo Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China
| | - Xueying Bu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
| | - Danlei Chen
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China
| | - Xueyan Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
| | - Hao Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China.
| | - Qinggele Caiyin
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China.
- Key Laboratory of Systems Bioengineering, Ministry of Education (Tianjin University), Tianjin, 300072, China.
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300072, China.
| | - Jianjun Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China.
- Key Laboratory of Systems Bioengineering, Ministry of Education (Tianjin University), Tianjin, 300072, China.
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300072, China.
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
7
|
McBride MA, Caja KR, Patil TK, Owen AM, Luan L, Bohannon JK, Hernandez A, Stothers CL, Trenary IA, Rahim M, Young JD, Calcutt MW, Stephens VR, Davis X, Oliver MA, Hao D, Si C, McRae M, Nguyen KK, Davis NS, Wang J, Patil NK, Sherwood ER. Immunoresponsive gene 1 facilitates TLR4 agonist-induced augmentation of innate antimicrobial immunity. J Leukoc Biol 2025; 117:qiae198. [PMID: 39351765 PMCID: PMC11879002 DOI: 10.1093/jleuko/qiae198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/10/2024] [Indexed: 11/09/2024] Open
Abstract
Treatment with the toll-like receptor 4 agonist monophosphoryl lipid A conditions innate immunocytes to respond robustly to subsequent infection, a phenotype termed innate immune memory. Our published studies show that metabolic reprogramming of macrophages is a prominent feature of the memory phenotype. We undertook studies to define the functional contributions of tricarboxylic acid cycle reprogramming to innate immune memory. We observed that priming of wild-type mice with monophosphoryl lipid A potently facilitated accumulation of the tricarboxylic acid cycle metabolite itaconate at sites of infection and enhanced microbial clearance. Augmentation of itaconate accumulation and microbial clearance was ablated in Irg1-deficient mice. We further observed that monophosphoryl lipid A potently induces expression of Irg1 and accumulation of itaconate in macrophages. Compared to wild-type macrophages, the ability of Irg1-deficient macrophages to kill Pseudomonas aeruginosa was impaired. We further observed that itaconate is directly antimicrobial against P. aeruginosa at pH 5, which is characteristic of the phagolysosome, and is facilitated by reactive oxygen species. Monophosphoryl lipid A-induced augmentation of glycolysis, oxidative phosphorylation, and accumulation of the tricarboxylic acid cycle metabolites succinate and malate was decreased in Irg1 knockout macrophages compared to wild-type controls. RNA sequencing revealed suppressed transcription of genes associated with phagolysosome function and increased expression of genes associated with cytokine production and chemotaxis in Irg1-deficient macrophages. This study identifies a contribution of itaconate to monophosphoryl lipid A-induced augmentation of innate antimicrobial immunity via facilitation of microbial killing as well as impact on metabolic and transcriptional adaptations.
Collapse
Affiliation(s)
- Margaret A McBride
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Katherine R Caja
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Tazeen K Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Allison M Owen
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Liming Luan
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Julia K Bohannon
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Cody L Stothers
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Irina A Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Mohsin Rahim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - M Wade Calcutt
- Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University School of Medicine, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Victoria R Stephens
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Xenia Davis
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Mary A Oliver
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Dan Hao
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Clara Si
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Malik McRae
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Kenny K Nguyen
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Nicholas S Davis
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Jingbin Wang
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Naeem K Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Edward R Sherwood
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
- Department of Surgery, East Tennessee State University, Quillen College of Medicine, PO Box 70575, Johnson City, TN 37614, United States
- Center of Excellence for Inflammation, Infectious Disease and Immunity, East Tennessee State University, Quillen College of Medicine, Johnson City, TN 37614, United States
| |
Collapse
|
8
|
Liu Z, Wang C. Dissecting S-itaconation at host-pathogen interactions with chemical proteomics tools. Curr Opin Microbiol 2025; 83:102579. [PMID: 39842211 DOI: 10.1016/j.mib.2025.102579] [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: 12/04/2024] [Revised: 01/01/2025] [Accepted: 01/02/2025] [Indexed: 01/24/2025]
Abstract
The molecular essence of the battle between host and pathogens lies in the protein-protein or protein-metabolite interactions. Itaconate is one of the most upregulated immunometabolites, regulating immune responses through either noncovalent binding or covalent modification in the host. We herein briefly review recent progresses in the discoveries of physiological and pathological roles of itaconate and applications of chemical proteomic technologies in exploring itaconate modifications on cysteines (S-itaconation) at the interface of host-pathogen interactions. Key challenges are also proposed as future outlook.
Collapse
Affiliation(s)
- Zihua Liu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Chu Wang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| |
Collapse
|
9
|
Yin S, Tao Y, Li T, Li C, Cui Y, Zhang Y, Yin S, Zhao L, Hu P, Cui L, Wu Y, He Y, Yu S, Chen J, Lu S, Qiu G, Song M, Hou Q, Qian C, Zou Z, Xu S, Yu Y. Itaconate facilitates viral infection via alkylating GDI2 and retaining Rab GTPase on the membrane. Signal Transduct Target Ther 2024; 9:371. [PMID: 39730330 DOI: 10.1038/s41392-024-02077-8] [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: 03/19/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 12/29/2024] Open
Abstract
Metabolic reprogramming of host cells plays critical roles during viral infection. Itaconate, a metabolite produced from cis-aconitate in the tricarboxylic acid cycle (TCA) by immune responsive gene 1 (IRG1), is involved in regulating innate immune response and pathogen infection. However, its involvement in viral infection and underlying mechanisms remain incompletely understood. Here, we demonstrate that the IRG1-itaconate axis facilitates the infections of VSV and IAV in macrophages and epithelial cells via Rab GTPases redistribution. Mechanistically, itaconate promotes the retention of Rab GTPases on the membrane via directly alkylating Rab GDP dissociation inhibitor beta (GDI2), the latter of which extracts Rab GTPases from the membrane to the cytoplasm. Multiple alkylated residues by itaconate, including cysteines 203, 335, and 414 on GDI2, were found to be important during viral infection. Additionally, this effect of itaconate needs an adequate distribution of Rab GTPases on the membrane, which relies on Rab geranylgeranyl transferase (GGTase-II)-mediated geranylgeranylation of Rab GTPases. The single-cell RNA sequencing data revealed high expression of IRG1 primarily in neutrophils during viral infection. Co-cultured and in vivo animal experiments demonstrated that itaconate produced by neutrophils plays a dominant role in promoting viral infection. Overall, our study reveals that neutrophils-derived itaconate facilitates viral infection via redistribution of Rab GTPases, suggesting potential targets for antiviral therapy.
Collapse
Affiliation(s)
- Shulei Yin
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Yijie Tao
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Tianliang Li
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Chunzhen Li
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Yani Cui
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Yunyan Zhang
- Department of Respiratory and Critical Care Medicine, Changzheng Hospital, Naval Medical University, Shanghai, 200433, China
| | - Shenhui Yin
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Liyuan Zhao
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Panpan Hu
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Likun Cui
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Yunyang Wu
- Department of Traditional Chinese Medicine, Naval Medical University, Shanghai, 200433, China
| | - Yixian He
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Shu Yu
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Jie Chen
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Shaoteng Lu
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Guifang Qiu
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Mengqi Song
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Qianshan Hou
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Cheng Qian
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Zui Zou
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China.
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China.
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Sheng Xu
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China.
| | - Yizhi Yu
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
10
|
Zhang X, Sun B, Xi T, Zhao J, Yang K, Sun S, Yang C. Quorum quenching as a mechanism for Cu-bearing stainless steel to conduct nonbiocidal retardation of biofilm development. CHEMICAL ENGINEERING JOURNAL 2024; 499:155996. [DOI: 10.1016/j.cej.2024.155996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
|
11
|
Licea-Herrera JI, Guerrero A, Mireles-Martínez M, Rodríguez-González Y, Aguilera-Arreola G, Contreras-Rodríguez A, Fernandez-Davila S, Requena-Castro R, Rivera G, Bocanegra-García V, Martínez-Vázquez AV. Agricultural Soil as a Reservoir of Pseudomonas aeruginosa with Potential Risk to Public Health. Microorganisms 2024; 12:2181. [PMID: 39597570 PMCID: PMC11596188 DOI: 10.3390/microorganisms12112181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen with a high capacity to adapt to different factors. The aim of this study is to analyze the pathogenicity in P. aeruginosa strains and their resistance to heavy metals and antibiotics, in agricultural soil of the state of Tamaulipas, Mexico. Susceptibility to 16 antibiotics was tested using the Kirby-Bauer method (CLSI). Eight virulence factors (FV) and six genes associated with heavy metal resistance were detected by PCR. As a result, P. aeruginosa was detected in 55% of the samples. The eight virulence factors were identified in ≥80% of the strains. The strains showed some level of resistance to only three antibiotics: 32.8% to ticarcillin, 40.8% to ticarcillin/clavulanic acid and 2.4% to aztreonam. The most frequent heavy metal resistance genes were arsC (92.8%) and copA (90.4%). However, copB and arsB genes were also identified in a percentage greater than 80%, and the least frequent genes were merA in 14.4% and czcA in 7.2%. Although P. aeruginosa strains showed a high percentage of factor virulence (potential ability to cause infections), their high levels of susceptibility to antibiotics lead to the assumption that infections are easily curable.
Collapse
Affiliation(s)
- Jessica I. Licea-Herrera
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Tamaulipas, Mexico; (J.I.L.-H.); (M.M.-M.); (Y.R.-G.); (S.F.-D.); (R.R.-C.); (G.R.); (V.B.-G.)
| | - Abraham Guerrero
- Consejo Nacional de Ciencia y Tecnología (CONAHCyT), Centro de Investigación en Alimentación y Desarrollo (CIAD), Mazatlán 82100, Sinaloa, Mexico;
| | - Maribel Mireles-Martínez
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Tamaulipas, Mexico; (J.I.L.-H.); (M.M.-M.); (Y.R.-G.); (S.F.-D.); (R.R.-C.); (G.R.); (V.B.-G.)
| | - Yuridia Rodríguez-González
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Tamaulipas, Mexico; (J.I.L.-H.); (M.M.-M.); (Y.R.-G.); (S.F.-D.); (R.R.-C.); (G.R.); (V.B.-G.)
| | - Guadalupe Aguilera-Arreola
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City 11340, Mexico; (G.A.-A.); (A.C.-R.)
| | - Araceli Contreras-Rodríguez
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City 11340, Mexico; (G.A.-A.); (A.C.-R.)
| | - Susana Fernandez-Davila
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Tamaulipas, Mexico; (J.I.L.-H.); (M.M.-M.); (Y.R.-G.); (S.F.-D.); (R.R.-C.); (G.R.); (V.B.-G.)
| | - Rocío Requena-Castro
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Tamaulipas, Mexico; (J.I.L.-H.); (M.M.-M.); (Y.R.-G.); (S.F.-D.); (R.R.-C.); (G.R.); (V.B.-G.)
| | - Gildardo Rivera
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Tamaulipas, Mexico; (J.I.L.-H.); (M.M.-M.); (Y.R.-G.); (S.F.-D.); (R.R.-C.); (G.R.); (V.B.-G.)
| | - Virgilio Bocanegra-García
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Tamaulipas, Mexico; (J.I.L.-H.); (M.M.-M.); (Y.R.-G.); (S.F.-D.); (R.R.-C.); (G.R.); (V.B.-G.)
| | - Ana Verónica Martínez-Vázquez
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Tamaulipas, Mexico; (J.I.L.-H.); (M.M.-M.); (Y.R.-G.); (S.F.-D.); (R.R.-C.); (G.R.); (V.B.-G.)
| |
Collapse
|
12
|
El Hafi B, Jean-Pierre F, O’Toole GA. Pseudomonas aeruginosa supports the survival of Prevotella melaninogenica in a cystic fibrosis lung polymicrobial community through metabolic cross-feeding. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.619475. [PMID: 39484496 PMCID: PMC11527032 DOI: 10.1101/2024.10.21.619475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Cystic fibrosis (CF) is a multi-organ genetic disorder that affects more than 100,000 individuals worldwide. Chronic respiratory infections are among the hallmark complications associated with CF lung disease, and these infections are often due to polymicrobial communities that colonize the airways of persons with CF (pwCF). Such infections are a significant cause of morbidity and mortality, with studies indicating that pwCF who are co-infected with more than one organism experience more frequent pulmonary exacerbations, leading to a faster decline in lung function. Previous work established an in vitro CF-relevant polymicrobial community model composed of P. aeruginosa, S. aureus, S. sanguinis, and P. melaninogenica. P. melaninogenica cannot survive in monoculture in this model. In this study, we leverage this model to investigate the interactions between P. aeruginosa and P. melaninogenica, allowing us to understand the mechanisms by which the two microbes interact to support the growth of P. melaninogenica specifically in the context of the polymicrobial community. We demonstrate a cross-feeding mechanism whereby P. melaninogenica metabolizes mucin into short-chain fatty acids that are in turn utilized by P. aeruginosa and converted into metabolites (succinate, acetate) that are cross-fed to P. melaninogenica, supporting the survival of this anaerobe in the CF lung-relevant model.
Collapse
Affiliation(s)
- Bassam El Hafi
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Fabrice Jean-Pierre
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - George A. O’Toole
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
13
|
Moulding PB, El-Halfawy OM. Chemical-mediated virulence: the effects of host chemicals on microbial virulence and potential new antivirulence strategies. Can J Microbiol 2024; 70:405-425. [PMID: 38905704 DOI: 10.1139/cjm-2024-0017] [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: 06/23/2024]
Abstract
The rising antimicrobial resistance rates and declining antimicrobial discovery necessitate alternative strategies to combat multidrug-resistant pathogens. Targeting microbial virulence is an emerging area of interest. Traditionally, virulence factors were largely restricted to bacteria-derived toxins, adhesins, capsules, quorum sensing systems, secretion systems, factors required to sense, respond to, acquire, or synthesize, and utilize trace elements (such as iron and other metals) and micronutrients (such as vitamins), and other factors bacteria use to establish infection, form biofilms, or damage the host tissues and regulatory elements thereof. However, this traditional definition overlooks bacterial virulence that may be induced or influenced by host-produced metabolites or other chemicals that bacteria may encounter at the infection site. This review will discuss virulence from a non-traditional perspective, shedding light on chemical-mediated host-pathogen interactions and outlining currently available mechanistic insight into increased bacterial virulence in response to host factors. This review aims to define a possibly underestimated theme of chemically mediated host-pathogen interactions and encourage future validation and characterization of the contribution of host chemicals to microbial virulence in vivo. From this perspective, we discuss proposed antivirulence compounds and suggest new potential targets for antimicrobials that prevent chemical-mediated virulence. We also explore proposed host-targeting therapeutics reducing the level of host chemicals that induce microbial virulence, serving as virulence attenuators. Understanding the host chemical-mediated virulence may enable new antimicrobial solutions to fight multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Peri B Moulding
- Department of Chemistry and Biochemistry, Faculty of Science, University of Regina, Regina, SK S4S 0A2, Canada
| | - Omar M El-Halfawy
- Department of Chemistry and Biochemistry, Faculty of Science, University of Regina, Regina, SK S4S 0A2, Canada
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| |
Collapse
|
14
|
He R, Zuo Y, Yi K, Liu B, Song C, Li N, Geng Q. The role and therapeutic potential of itaconate in lung disease. Cell Mol Biol Lett 2024; 29:129. [PMID: 39354366 PMCID: PMC11445945 DOI: 10.1186/s11658-024-00642-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 09/04/2024] [Indexed: 10/03/2024] Open
Abstract
Lung diseases triggered by endogenous or exogenous factors have become a major concern, with high morbidity and mortality rates, especially after the coronavirus disease 2019 (COVID-19) pandemic. Inflammation and an over-activated immune system can lead to a cytokine cascade, resulting in lung dysfunction and injury. Itaconate, a metabolite produced by macrophages, has been reported as an effective anti-inflammatory and anti-oxidative stress agent with significant potential in regulating immunometabolism. As a naturally occurring metabolite in immune cells, itaconate has been identified as a potential therapeutic target in lung diseases through its role in regulating inflammation and immunometabolism. This review focuses on the origin, regulation, and function of itaconate in lung diseases, and briefly discusses its therapeutic potential.
Collapse
Affiliation(s)
- Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Hubei Province, 99 Zhangzhidong Road, Wuhan, 430060, China
| | - Yifan Zuo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Hubei Province, 99 Zhangzhidong Road, Wuhan, 430060, China
| | - Ke Yi
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Hubei Province, 99 Zhangzhidong Road, Wuhan, 430060, China
| | - Bohao Liu
- Department of Thoracic Surgery, Jilin University, Changchun, China
| | - Congkuan Song
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Hubei Province, 99 Zhangzhidong Road, Wuhan, 430060, China.
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Hubei Province, 99 Zhangzhidong Road, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Hubei Province, 99 Zhangzhidong Road, Wuhan, 430060, China.
| |
Collapse
|
15
|
Thom RE, D’Elia RV. Future applications of host direct therapies for infectious disease treatment. Front Immunol 2024; 15:1436557. [PMID: 39411713 PMCID: PMC11473292 DOI: 10.3389/fimmu.2024.1436557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
New and emerging pathogens, such as SARS-CoV2 have highlighted the requirement for threat agnostic therapies. Some antibiotics or antivirals can demonstrate broad-spectrum activity against pathogens in the same family or genus but efficacy can quickly reduce due to their specific mechanism of action and for the ability of the disease causing agent to evolve. This has led to the generation of antimicrobial resistant strains, making infectious diseases more difficult to treat. Alternative approaches therefore need to be considered, which include exploring the utility of Host-Directed Therapies (HDTs). This is a growing area with huge potential but difficulties arise due to the complexity of disease profiles. For example, a HDT given early during infection may not be appropriate or as effective when the disease has become chronic or when a patient is in intensive care. With the growing understanding of immune function, a new generation of HDT for the treatment of disease could allow targeting specific pathways to augment or diminish the host response, dependent upon disease profile, and allow for bespoke therapeutic management plans. This review highlights promising and approved HDTs that can manipulate the immune system throughout the spectrum of disease, in particular to viral and bacterial pathogens, and demonstrates how the advantages of HDT will soon outweigh the potential side effects.
Collapse
Affiliation(s)
- Ruth E. Thom
- Chemical, Biological and Radiological Division, Defence Science and Technology Laboratory, Porton Down, Salisbury, United Kingdom
| | - R V. D’Elia
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
16
|
Zhao R, Xu L, Chen J, Yang Y, Guo X, Dai M, Tian GB, Qin LN. Itaconate induces tolerance of Staphylococcus aureus to aminoglycoside antibiotics. Front Microbiol 2024; 15:1450085. [PMID: 39403084 PMCID: PMC11471559 DOI: 10.3389/fmicb.2024.1450085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/09/2024] [Indexed: 01/04/2025] Open
Abstract
INTRODUCTION Staphylococcus aureus is one of the chief pathogens that cause chronic and recurrent infections. Failure of the antibiotics to curb the infections contributes to relapse and is an important reason for the high mortality rate. Treatment failure may also be due to antibiotic tolerance. Accumulating evidence suggests that t the host immune environment plays an important role in inducing antibiotic tolerance of S. aureus, but research in this area has been limited. METHODS In this study,the minimum inhibitory concentration (MIC) of the antibiotics against S. aureus was determined using the standard broth microdilution method.The study evaluated whether itaconate induces antibiotic tolerance in S. aureus through an antibiotic bactericidal activity assay.The effect of itaconate on the growth of S. aureus was evaluated by monitoring the growth of S. aureus in medium supplemented with itaconate. Additionally, RNA sequencing and metabolomics analyses were used to determine transcriptional and metabolic changes in S. aureus when exposed to itaconate. RESULTS AND DISCUSSION According to the study,we found that the immune metabolite itaconate can induce tolerance in both methicillin-resistant and -susceptible S. aureus to aminoglycosides. When S. aureus was exposed to itaconate, its growth slowed down and transcriptomic and metabolomic alterations associated with decreased energy metabolism, including the tricarboxylate cycle, glycolysis, pyruvate metabolism, and arginine biosynthesis, were observed. These changes are associated with aminoglycoside tolerance. This study highlights the role of immune signaling metabolites in bacterial antibiotic tolerance and suggests new strategies to improve antibiotic treatment by modulating the host immune response and stimulating the metabolism of bacteria.
Collapse
Affiliation(s)
- Runping Zhao
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Lei Xu
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Jieyun Chen
- Zhongshan School of Medicine, Advanced Medical Technology Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Immunology, School of Medicine, Sun Yat-Sen University, Shenzhen, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pharmacy, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Key Laboratory of Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Yanxian Yang
- Zhongshan School of Medicine, Advanced Medical Technology Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Immunology, School of Medicine, Sun Yat-Sen University, Shenzhen, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pharmacy, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Key Laboratory of Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Xilong Guo
- Zhongshan School of Medicine, Advanced Medical Technology Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Immunology, School of Medicine, Sun Yat-Sen University, Shenzhen, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pharmacy, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Key Laboratory of Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Min Dai
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Guo-Bao Tian
- Zhongshan School of Medicine, Advanced Medical Technology Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Immunology, School of Medicine, Sun Yat-Sen University, Shenzhen, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pharmacy, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Key Laboratory of Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Li-Na Qin
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
17
|
Liu Z, Liu D, Wang C. In situ chemoproteomic profiling reveals itaconate inhibits de novo purine biosynthesis in pathogens. Cell Rep 2024; 43:114737. [PMID: 39277862 DOI: 10.1016/j.celrep.2024.114737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/23/2024] [Accepted: 08/23/2024] [Indexed: 09/17/2024] Open
Abstract
Itaconate serves as an immune-specific metabolite that regulates gene transcription and metabolism in both host and pathogens. S-itaconation is a post-translational modification that regulates immune response; however, its antimicrobial mechanism under the physiological condition remains unclear. Here, we apply a bioorthogonal itaconate probe to perform global profiling of S-itaconation in living pathogens, including S. Typhimurium, S. aureus, and P. aeruginosa. Some functional enzymes are covalently modified by itaconate, including those involved in the de novo purine biosynthesis pathway. Further biochemical studies demonstrate that itaconate suppresses this specific pathway to limit Salmonella growth by inhibiting the initiator purF to lower de novo purine biosynthesis and simultaneously targeting the guaABC cluster to block the salvage route. Our chemoproteomic study provides a global portrait of S-itaconation in multiple pathogens and offers a valuable resource for finding susceptible targets to combat drug-resistant pathogens in the future.
Collapse
Affiliation(s)
- Zihua Liu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Dongyang Liu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Chu Wang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| |
Collapse
|
18
|
Jeong GJ, Khan F, Tabassum N, Kim YM. Natural and synthetic molecules with potential to enhance biofilm formation and virulence properties in Pseudomonas aeruginosa. Crit Rev Microbiol 2024; 50:830-858. [PMID: 37968960 DOI: 10.1080/1040841x.2023.2282459] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 10/06/2023] [Accepted: 11/01/2023] [Indexed: 11/17/2023]
Abstract
Pseudomonas aeruginosa can efficiently adapt to changing environmental conditions due to its ubiquitous nature, intrinsic/acquired/adaptive resistance mechanisms, high metabolic versatility, and the production of numerous virulence factors. As a result, P. aeruginosa becomes an opportunistic pathogen, causing chronic infection in the lungs and several organs of patients suffering from cystic fibrosis. Biofilm established by P. aeruginosa in host tissues and medical device surfaces has been identified as a major obstruction to antimicrobial therapy. P. aeruginosa is very likely to be closely associated with the various microorganisms in the host tissues or organs in a pathogenic or nonpathogenic behavior. Aside from host-derived molecules, other beneficial and pathogenic microorganisms produce a diverse range of secondary metabolites that either directly or indirectly favor the persistence of P. aeruginosa. Thus, it is critical to understand how P. aeruginosa interacts with different molecules and ions in the host and abiotic environment to produce extracellular polymeric substances and virulence factors. Thus, the current review discusses how various natural and synthetic molecules in the environment induce biofilm formation and the production of multiple virulence factors.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan, Republic of Korea
| | - Fazlurrahman Khan
- Institute of Fisheries Sciences, Pukyong National University, Busan, Republic of Korea
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, Republic of Korea
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, Republic of Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan, Republic of Korea
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, Republic of Korea
| |
Collapse
|
19
|
Zhang H, Tsui CK, Garcia G, Joe LK, Wu H, Maruichi A, Fan W, Pandovski S, Yoon PH, Webster BM, Durieux J, Frankino PA, Higuchi-Sanabria R, Dillin A. The extracellular matrix integrates mitochondrial homeostasis. Cell 2024; 187:4289-4304.e26. [PMID: 38942015 DOI: 10.1016/j.cell.2024.05.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/22/2024] [Accepted: 05/31/2024] [Indexed: 06/30/2024]
Abstract
Cellular homeostasis is intricately influenced by stimuli from the microenvironment, including signaling molecules, metabolites, and pathogens. Functioning as a signaling hub within the cell, mitochondria integrate information from various intracellular compartments to regulate cellular signaling and metabolism. Multiple studies have shown that mitochondria may respond to various extracellular signaling events. However, it is less clear how changes in the extracellular matrix (ECM) can impact mitochondrial homeostasis to regulate animal physiology. We find that ECM remodeling alters mitochondrial homeostasis in an evolutionarily conserved manner. Mechanistically, ECM remodeling triggers a TGF-β response to induce mitochondrial fission and the unfolded protein response of the mitochondria (UPRMT). At the organismal level, ECM remodeling promotes defense of animals against pathogens through enhanced mitochondrial stress responses. We postulate that this ECM-mitochondria crosstalk represents an ancient immune pathway, which detects infection- or mechanical-stress-induced ECM damage, thereby initiating adaptive mitochondria-based immune and metabolic responses.
Collapse
Affiliation(s)
- Hanlin Zhang
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - C Kimberly Tsui
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gilberto Garcia
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Larry K Joe
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Haolun Wu
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ayane Maruichi
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Wudi Fan
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sentibel Pandovski
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Peter H Yoon
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Brant M Webster
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jenni Durieux
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Phillip A Frankino
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ryo Higuchi-Sanabria
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Andrew Dillin
- Department of Molecular & Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
20
|
Hofstaedter CE, O’Keefe IP, Met CM, Wu L, Vanderwoude J, Shin S, Diggle SP, Riquelme SA, Rasko DA, Doi Y, Harro JM, Kopp BT, Ernst RK. Pseudomonas aeruginosa Lipid A Structural Variants Induce Altered Immune Responses. Am J Respir Cell Mol Biol 2024; 71:207-218. [PMID: 38656811 PMCID: PMC11299085 DOI: 10.1165/rcmb.2024-0059oc] [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: 02/09/2024] [Accepted: 04/23/2024] [Indexed: 04/26/2024] Open
Abstract
Pseudomonas aeruginosa causes chronic lung infection in cystic fibrosis (CF), resulting in structural lung damage and progressive pulmonary decline. P. aeruginosa in the CF lung undergoes numerous changes, adapting to host-specific airway pressures while establishing chronic infection. P. aeruginosa undergoes lipid A structural modification during CF chronic infection that is not seen in any other disease state. Lipid A, the membrane anchor of LPS (i.e., endotoxin), comprises the majority of the outer membrane of Gram-negative bacteria and is a potent Toll-like receptor 4 (TLR4) agonist. The structure of P. aeruginosa lipid A is intimately linked with its recognition by TLR4 and subsequent immune response. Prior work has identified P. aeruginosa strains with altered lipid A structures that arise during chronic CF lung infection; however, the impact of the P. aeruginosa lipid A structure on airway disease has not been investigated. Here, we show that P. aeruginosa lipid A lacks PagL-mediated deacylation during human airway infection using a direct-from-sample mass spectrometry approach on human BAL fluid. This structure triggers increased proinflammatory cytokine production by primary human macrophages. Furthermore, alterations in lipid A 2-hydroxylation impact cytokine response in a site-specific manner, independent of CF transmembrane conductance regulator function. It is interesting that there is a CF-specific reduction in IL-8 secretion within the epithelial-cell compartment that only occurs in CF bronchial epithelial cells when infected with CF-adapted P. aeruginosa that lacks PagL-mediated lipid A deacylation. Taken together, we show that P. aeruginosa alters its lipid A structure during acute lung infection and that this lipid A structure induces stronger signaling through TLR4.
Collapse
Affiliation(s)
| | | | | | - Ling Wu
- Department of Microbiology and
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jelly Vanderwoude
- Center for Microbial Dynamics and Infection, School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia
| | | | - Stephen P. Diggle
- Center for Microbial Dynamics and Infection, School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia
| | | | - David A. Rasko
- Institute for Genome Sciences
- Department of Microbiology and Immunology, and
- Center for Pathogen Research, University of Maryland, Baltimore, Baltimore, Maryland
| | - Yohei Doi
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | | | - Benjamin T. Kopp
- Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Emory University, Atlanta, Georgia
| | | |
Collapse
|
21
|
Pedersen BH, Simões FB, Pogrebnyakov I, Welch M, Johansen HK, Molin S, La Rosa R. Metabolic specialization drives reduced pathogenicity in Pseudomonas aeruginosa isolates from cystic fibrosis patients. PLoS Biol 2024; 22:e3002781. [PMID: 39178315 PMCID: PMC11376529 DOI: 10.1371/journal.pbio.3002781] [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: 01/17/2024] [Revised: 09/05/2024] [Accepted: 08/01/2024] [Indexed: 08/25/2024] Open
Abstract
Metabolism provides the foundation for all cellular functions. During persistent infections, in adapted pathogenic bacteria metabolism functions radically differently compared with more naïve strains. Whether this is simply a necessary accommodation to the persistence phenotype or if metabolism plays a direct role in achieving persistence in the host is still unclear. Here, we characterize a convergent shift in metabolic function(s) linked with the persistence phenotype during Pseudomonas aeruginosa colonization in the airways of people with cystic fibrosis. We show that clinically relevant mutations in the key metabolic enzyme, pyruvate dehydrogenase, lead to a host-specialized metabolism together with a lower virulence and immune response recruitment. These changes in infection phenotype are mediated by impaired type III secretion system activity and by secretion of the antioxidant metabolite, pyruvate, respectively. Our results show how metabolic adaptations directly impinge on persistence and pathogenicity in this organism.
Collapse
Affiliation(s)
- Bjarke Haldrup Pedersen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Filipa Bica Simões
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
| | - Ivan Pogrebnyakov
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Martin Welch
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Helle Krogh Johansen
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Molin
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Ruggero La Rosa
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
22
|
Huang Q, Duan C, Ma H, Nong C, Zheng Q, Zhou J, Zhao N, Mou X, Liu T, Zou S, Yang N, Tong A, Qin W, Bao R. Structural and functional characterization of itaconyl-CoA hydratase and citramalyl-CoA lyase involved in itaconate metabolism of Pseudomonas aeruginosa. Structure 2024; 32:941-952.e3. [PMID: 38677288 DOI: 10.1016/j.str.2024.04.004] [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: 10/25/2023] [Revised: 02/23/2024] [Accepted: 04/02/2024] [Indexed: 04/29/2024]
Abstract
Itaconate is a key anti-inflammatory/antibacterial metabolite in pathogen-macrophage interactions that induces adaptive changes in Pseudomonas aeruginosa-exposed airways. However, the impact and mechanisms underlying itaconate metabolism remain unclear. Our study reveals that itaconate significantly upregulates the expression of pyoverdine in P. aeruginosa and enhances its tolerance to tobramycin. Notably, the enzymes responsible for efficient itaconate metabolism, PaIch and PaCcl, play crucial roles in both utilizing itaconate and clearing its toxic metabolic intermediates. By using protein crystallography and molecular dynamics simulations analyses, we have elucidated the unique catalytic center and substrate-binding pocket of PaIch, which contribute to its highly efficient catalysis. Meanwhile, analysis of PaCcl has revealed how interactions between domains regulate the conformational changes of the active sites and binding pockets, influencing the catalytic process. Overall, our research uncovers the significance and mechanisms of PaIch and PaCcl in the efficient metabolism of itaconate by P. aeruginosa.
Collapse
Affiliation(s)
- Qin Huang
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chenxi Duan
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Haichuan Ma
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Cheng Nong
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qianqian Zheng
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jun Zhou
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ninglin Zhao
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingyu Mou
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tonggen Liu
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shuang Zou
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ningchuan Yang
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Aiping Tong
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wenming Qin
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, CAS, Shanghai, China.
| | - Rui Bao
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
23
|
Ye D, Wang P, Chen LL, Guan KL, Xiong Y. Itaconate in host inflammation and defense. Trends Endocrinol Metab 2024; 35:586-606. [PMID: 38448252 DOI: 10.1016/j.tem.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 03/08/2024]
Abstract
Immune cells undergo rapid and extensive metabolic changes during inflammation. In addition to contributing to energetic and biosynthetic demands, metabolites can also function as signaling molecules. Itaconate (ITA) rapidly accumulates to high levels in myeloid cells under infectious and sterile inflammatory conditions. This metabolite binds to and regulates the function of diverse proteins intracellularly to influence metabolism, oxidative response, epigenetic modification, and gene expression and to signal extracellularly through binding the G protein-coupled receptor (GPCR). Administration of ITA protects against inflammatory diseases and blockade of ITA production enhances antitumor immunity in preclinical models. In this article, we review ITA metabolism and its regulation, discuss its target proteins and mechanisms, and conjecture a rationale for developing ITA-based therapeutics to treat inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Dan Ye
- Molecular and Cell Biology Laboratory, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China.
| | - Pu Wang
- Molecular and Cell Biology Laboratory, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lei-Lei Chen
- Molecular and Cell Biology Laboratory, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Kun-Liang Guan
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Yue Xiong
- Cullgen Inc., 12730 High Bluff Drive, San Diego, CA 92130, USA.
| |
Collapse
|
24
|
Ramachandran RA, Abdallah JT, Rehman M, Baniasadi H, Blanton AM, Vizcaino S, Robertson DM. Pseudomonas aeruginosa impairs mitochondrial function and metabolism during infection of corneal epithelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600521. [PMID: 38979356 PMCID: PMC11230238 DOI: 10.1101/2024.06.24.600521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Pseudomonas aeruginosa (PA) is a gram-negative opportunistic pathogen that can infect the cornea as a result of trauma or contact lens wear. In addition to their known energy producing role, mitochondria are important mediators of immune signaling and host defense. While certain pathogens have developed strategies to evade host defenses by modulating host mitochondrial dynamics and metabolism, the ability of PA to harness host cell mitochondria during corneal infection is unknown. Using a combination of biochemical and imaging techniques, we show that PA infection of corneal epithelial cells induced mitochondrial fission in a DRP1-dependent manner that preceded PINK1/Parkin and FUNDC1-mediated mitophagy. PA also impaired NADH-linked respiration through a reduction in complex 1. This corresponded to a decrease in metabolic pathways related to glycolysis and the TCA cycle. Metabolomics analysis further demonstrated an upregulation of the pentose phosphate pathway, arginine, purine, and pyrimidine metabolism in PA infected cells. These pathways may provide a key source of nucleotides, amino acids, and nitrogen for both the host cell and PA, in addition to antioxidant functions. Following treatment with gentamicin to kill all extracellular bacteria, metabolic flux analysis showed that corneal epithelial cells were able to restore mitochondrial function despite the continued presence of intracellular PA. Taken together, these data demonstrate that mitochondrial dysfunction and metabolic rewiring in host cells is triggered by extracellular PA, but once inside, PA requires healthy mitochondria to ensure host cell survival.
Collapse
|
25
|
Gao Q, Chu X, Yang J, Guo Y, Guo H, Qian S, Yang Y, Wang B. An Antibiotic Nanobomb Constructed from pH-Responsive Chemical Bonds in Metal-Phenolic Network Nanoparticles for Biofilm Eradication and Corneal Ulcer Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309086. [PMID: 38488341 PMCID: PMC11165475 DOI: 10.1002/advs.202309086] [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: 12/24/2023] [Revised: 02/13/2024] [Indexed: 06/12/2024]
Abstract
In the treatment of refractory corneal ulcers caused by Pseudomonas aeruginosa, antibacterial drugs delivery faces the drawbacks of low permeability and short ocular surface retention time. Hence, novel positively-charged modular nanoparticles (NPs) are developed to load tobramycin (TOB) through a one-step self-assembly method based on metal-phenolic network and Schiff base reaction using 3,4,5-trihydroxybenzaldehyde (THBA), ε-poly-ʟ-lysine (EPL), and Cu2+ as matrix components. In vitro antibacterial test demonstrates that THBA-Cu-TOB NPs exhibit efficient instantaneous sterilization owing to the rapid pH responsiveness to bacterial infections. Notably, only 2.6 µg mL-1 TOP is needed to eradicate P. aeruginosa biofilm in the nano-formed THBA-Cu-TOB owing to the greatly enhanced penetration, which is only 1.6% the concentration of free TOB (160 µg mL-1). In animal experiments, THBA-Cu-TOB NPs show significant advantages in ocular surface retention, corneal permeability, rapid sterilization, and inflammation elimination. Based on molecular biology analysis, the toll-like receptor 4 and nuclear factor kappa B signaling pathways are greatly downregulated as well as the reduction of inflammatory cytokines secretions. Such a simple and modular strategy in constructing nano-drug delivery platform offers a new idea for toxicity reduction, physiological barrier penetration, and intelligent drug delivery.
Collapse
Affiliation(s)
- Qiang Gao
- National Engineering Research Center of Ophthalmology and OptometryEye HospitalWenzhou Medical UniversityWenzhou325000P. R. China
- State Key Laboratory of Ophthalmology, Optometry and Visual ScienceWenzhou Medical UniversityWenzhou325027P. R. China
| | - Xiaoying Chu
- National Engineering Research Center of Ophthalmology and OptometryEye HospitalWenzhou Medical UniversityWenzhou325000P. R. China
| | - Jie Yang
- School of Life SciencesJilin University2699 Qianjin StreetChangchun130012P. R. China
| | - Yishun Guo
- National Engineering Research Center of Ophthalmology and OptometryEye HospitalWenzhou Medical UniversityWenzhou325000P. R. China
| | - Hanwen Guo
- National Engineering Research Center of Ophthalmology and OptometryEye HospitalWenzhou Medical UniversityWenzhou325000P. R. China
| | - Siyuan Qian
- National Engineering Research Center of Ophthalmology and OptometryEye HospitalWenzhou Medical UniversityWenzhou325000P. R. China
| | - Ying‐Wei Yang
- College of ChemistryJilin University2699 Qianjin StreetChangchun130012P. R. China
| | - Bailiang Wang
- National Engineering Research Center of Ophthalmology and OptometryEye HospitalWenzhou Medical UniversityWenzhou325000P. R. China
- State Key Laboratory of Ophthalmology, Optometry and Visual ScienceWenzhou Medical UniversityWenzhou325027P. R. China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic DiseasesWenzhou325027P. R. China
| |
Collapse
|
26
|
Breen P, Zimbric M, Caverly LJ. Itaconic acid inhibits nontuberculous mycobacterial growth in pH dependent manner while 4-octyl-itaconic acid enhances THP-1 clearance of nontuberculous mycobacteria in vitro. PLoS One 2024; 19:e0303516. [PMID: 38728330 PMCID: PMC11086914 DOI: 10.1371/journal.pone.0303516] [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: 11/26/2023] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
Increasingly prevalent, nontuberculous mycobacteria (NTM) infections affect approximately 20% of people with cystic fibrosis (CF). Previous studies of CF sputum identified lower levels of the host metabolite itaconate in those infected with NTM. Itaconate can inhibit the growth of M. tuberculosis (MTB) in vitro via the inhibition of the glyoxylate cycle enzyme (ICL), but its impact on NTM is unclear. To test itaconic acid's (IA) effect on NTM growth, laboratory and CF clinical strains of Mycobacterium abscessus and Mycobacterium avium were cultured in 7H9 minimal media supplemented with 1-10 mM of IA and short-chain fatty acids (SCFA). M. avium and M. abscessus grew when supplemented with SCFAs, whereas the addition of IA (≥ 10 mM) completely inhibited NTM growth. NTM supplemented with acetate or propionate and 5 mM IA displayed slower growth than NTM cultured with SCFA and ≤ 1 mM of IA. However, IA's inhibition of NTM was pH dependent; as similar and higher quantities (100 mM) of pH adjusted IA (pH 7) did not inhibit growth in vitro, while in an acidic minimal media (pH 6.1), 1 to 5 mM of non-pH adjusted IA inhibited growth. None of the examined isolates displayed the ability to utilize IA as a carbon source, and IA added to M. abscessus isocitrate lyase (ICL) decreased enzymatic activity. Lastly, the addition of cell-permeable 4-octyl itaconate (4-OI) to THP-1 cells enhanced NTM clearance, demonstrating a potential role for IA/itaconate in host defense against NTM infections.
Collapse
Affiliation(s)
- Paul Breen
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Madsen Zimbric
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Lindsay J. Caverly
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States of America
| |
Collapse
|
27
|
Luo Z, Sheng Z, Hu L, Shi L, Tian Y, Zhao X, Yang W, Xiao Z, Shen D, Wu W, Lan T, Zhao B, Wang X, Zhuang N, Zhang JN, Wang Y, Lu Y, Wang L, Zhang C, Wang P, An J, Yang F, Li Q. Targeted macrophage phagocytosis by Irg1/itaconate axis improves the prognosis of intracerebral hemorrhagic stroke and peritonitis. EBioMedicine 2024; 101:104993. [PMID: 38324982 PMCID: PMC10862510 DOI: 10.1016/j.ebiom.2024.104993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Macrophages are innate immune cells whose phagocytosis function is critical to the prognosis of stroke and peritonitis. cis-aconitic decarboxylase immune-responsive gene 1 (Irg1) and its metabolic product itaconate inhibit bacterial infection, intracellular viral replication, and inflammation in macrophages. Here we explore whether itaconate regulates phagocytosis. METHODS Phagocytosis of macrophages was investigated by time-lapse video recording, flow cytometry, and immunofluorescence staining in macrophage/microglia cultures isolated from mouse tissue. Unbiased RNA-sequencing and ChIP-sequencing assays were used to explore the underlying mechanisms. The effects of Irg1/itaconate axis on the prognosis of intracerebral hemorrhagic stroke (ICH) and peritonitis was observed in transgenic (Irg1flox/flox; Cx3cr1creERT/+, cKO) mice or control mice in vivo. FINDINGS In a mouse model of ICH, depletion of Irg1 in macrophage/microglia decreased its phagocytosis of erythrocytes, thereby exacerbating outcomes (n = 10 animals/group, p < 0.05). Administration of sodium itaconate/4-octyl itaconate (4-OI) promoted macrophage phagocytosis (n = 7 animals/group, p < 0.05). In addition, in a mouse model of peritonitis, Irg1 deficiency in macrophages also inhibited phagocytosis of Staphylococcus aureus (n = 5 animals/group, p < 0.05) and aggravated outcomes (n = 9 animals/group, p < 0.05). Mechanistically, 4-OI alkylated cysteine 155 on the Kelch-like ECH-associated protein 1 (Keap1), consequent in nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and transcriptional activation of Cd36 gene. Blocking the function of CD36 completely abolished the phagocytosis-promoting effects of Irg1/itaconate axis in vitro and in vivo. INTERPRETATION Our findings provide a potential therapeutic target for phagocytosis-deficiency disorders, supporting further development towards clinical application for the benefit of stroke and peritonitis patients. FUNDING The National Natural Science Foundation of China (32070735, 82371321 to Q. Li, 82271240 to F. Yang) and the Beijing Natural Science Foundation Program and Scientific Research Key Program of Beijing Municipal Commission of Education (KZ202010025033 to Q. Li).
Collapse
Affiliation(s)
- Zhaoli Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Ziyang Sheng
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Liye Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Lei Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yichen Tian
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xiaochu Zhao
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Wei Yang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Zhongnan Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Danmin Shen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Weihua Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Ting Lan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Boqian Zhao
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xiaogang Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Nan Zhuang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Jian-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yamei Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yabin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Liyong Wang
- Core Facilities for Molecular Biology, Capital Medical University, Beijing 100069, China
| | - Chenguang Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Peipei Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Fei Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing 100069, China.
| | - Qian Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
28
|
Almeida L, Dhillon-LaBrooy A, Sparwasser T. The evolutionary tug-of-war of macrophage metabolism during bacterial infection. Trends Endocrinol Metab 2024; 35:235-248. [PMID: 38040578 DOI: 10.1016/j.tem.2023.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 12/03/2023]
Abstract
The function and phenotype of macrophages are intimately linked with pathogen detection. On sensing pathogen-derived signals and molecules, macrophages undergo a carefully orchestrated process of polarization to acquire pathogen-clearing properties. This phenotypic change must be adequately supported by metabolic reprogramming that is now known to support the acquisition of effector function, but also generates secondary metabolites with direct microbicidal activity. At the same time, bacteria themselves have adapted to both manipulate and take advantage of macrophage-specific metabolic adaptations. Here, we summarize the current knowledge on macrophage metabolism during infection, with a particular focus on understanding the 'arms race' between host immune cells and bacteria during immune responses.
Collapse
Affiliation(s)
- Luís Almeida
- Institute of Medical Microbiology and Hygiene, University Medical Center of Johannes Gutenberg University, Mainz 55131, Germany.
| | - Ayesha Dhillon-LaBrooy
- Institute of Medical Microbiology and Hygiene, University Medical Center of Johannes Gutenberg University, Mainz 55131, Germany
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Center of Johannes Gutenberg University, Mainz 55131, Germany.
| |
Collapse
|
29
|
Panahi Z, Owrang M, Goli HR. Significant role of pyocyanin and exotoxin A in the pathogenesis of Pseudomonas aeruginosa isolated from hospitalized patients. Folia Med (Plovdiv) 2024; 66:88-96. [PMID: 38426470 DOI: 10.3897/folmed.66.e111038] [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: 08/11/2023] [Accepted: 11/27/2023] [Indexed: 03/02/2024] Open
Abstract
AIM Due to the importance of exotoxin A and pyocyanin in the pathogenicity of this bacterium, we decided to evaluate the prevalence of genes encoding these virulence factors in clinical isolates of P.aeruginosa.
Collapse
|
30
|
Liu Y, Zhou M, Bu Y, Qin L, Zhang Y, Shao S, Wang Q. Lysine acetylation regulates the AT-rich DNA possession ability of H-NS. Nucleic Acids Res 2024; 52:1645-1660. [PMID: 38059366 PMCID: PMC10899749 DOI: 10.1093/nar/gkad1172] [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: 08/23/2023] [Revised: 11/01/2023] [Accepted: 11/23/2023] [Indexed: 12/08/2023] Open
Abstract
H-NS, the histone-like nucleoid-structuring protein in bacteria, regulates the stability of the bacterial genome by inhibiting the transcription of horizontally transferred genes, such as the type III and type VI secretion systems (T3/T6SS). While eukaryotic histone posttranslational modifications (PTMs) have been extensively studied, little is known about prokaryotic H-NS PTMs. Here, we report that the acetylation of H-NS attenuates its ability to silence horizontally transferred genes in response to amino acid nutrition and immune metabolites. Moreover, LC-MS/MS profiling showed that the acetyllysine sites of H-NS and K120 are indispensable for its DNA-binding ability. Acetylation of K120 leads to a low binding affinity for DNA and enhances T3/T6SS expression. Furthermore, acetylation of K120 impairs the AT-rich DNA recognition ability of H-NS. In addition, lysine acetylation in H-NS modulates in vivo bacterial virulence. These findings reveal the mechanism underlying H-NS PTMs and propose a novel mechanism by which bacteria counteract the xenogeneic silencing of H-NS.
Collapse
Affiliation(s)
- Yabo Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Mengqing Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yifan Bu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Liang Qin
- New Product R&D, GenScript Biotech Corporation, Nanjing 211100, China
| | - Yuanxing Zhang
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China
- Laboratory of Aquatic Animal Diseases of MOA, Shanghai 200237, China
| | - Shuai Shao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China
- Laboratory of Aquatic Animal Diseases of MOA, Shanghai 200237, China
| | - Qiyao Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China
- Laboratory of Aquatic Animal Diseases of MOA, Shanghai 200237, China
| |
Collapse
|
31
|
Mirzaei R, Campoccia D, Ravaioli S, Arciola CR. Emerging Issues and Initial Insights into Bacterial Biofilms: From Orthopedic Infection to Metabolomics. Antibiotics (Basel) 2024; 13:184. [PMID: 38391570 PMCID: PMC10885942 DOI: 10.3390/antibiotics13020184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024] Open
Abstract
Bacterial biofilms, enigmatic communities of microorganisms enclosed in an extracellular matrix, still represent an open challenge in many clinical contexts, including orthopedics, where biofilm-associated bone and joint infections remain the main cause of implant failure. This study explores the scenario of biofilm infections, with a focus on those related to orthopedic implants, highlighting recently emerged substantial aspects of the pathogenesis and their potential repercussions on the clinic, as well as the progress and gaps that still exist in the diagnostics and management of these infections. The classic mechanisms through which biofilms form and the more recently proposed new ones are depicted. The ways in which bacteria hide, become impenetrable to antibiotics, and evade the immune defenses, creating reservoirs of bacteria difficult to detect and reach, are delineated, such as bacterial dormancy within biofilms, entry into host cells, and penetration into bone canaliculi. New findings on biofilm formation with host components are presented. The article also delves into the emerging and critical concept of immunometabolism, a key function of immune cells that biofilm interferes with. The growing potential of biofilm metabolomics in the diagnosis and therapy of biofilm infections is highlighted, referring to the latest research.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Davide Campoccia
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (D.C.); (S.R.)
| | - Stefano Ravaioli
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (D.C.); (S.R.)
| | - Carla Renata Arciola
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| |
Collapse
|
32
|
Michalaki C, Albers GJ, Byrne AJ. Itaconate as a key regulator of respiratory disease. Clin Exp Immunol 2024; 215:120-125. [PMID: 38018224 PMCID: PMC10847819 DOI: 10.1093/cei/uxad127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/21/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023] Open
Abstract
Macrophage activation results in the accumulation of endogenous metabolites capable of adopting immunomodulatory roles; one such bioactive metabolite is itaconate. After macrophage stimulation, the TCA-cycle intermediate cis-aconitate is converted to itaconate (by aconitate decarboxylase-1, ACOD1) in the mitochondrial matrix. Recent studies have highlighted the potential of targeting itaconate as a therapeutic strategy for lung diseases such as asthma, idiopathic pulmonary fibrosis (IPF), and respiratory infections. This review aims to bring together evidence which highlights a role for itaconate in chronic lung diseases (such as asthma and pulmonary fibrosis) and respiratory infections (such as SARS-CoV-2, influenza and Mycobacterium tuberculosis infection). A better understanding of the role of itaconate in lung disease could pave the way for novel therapeutic interventions and improve patient outcomes in respiratory disorders.
Collapse
Affiliation(s)
- Christina Michalaki
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Gesa J Albers
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Adam J Byrne
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- School of Medicine and Conway Institute of Biomedical Sciences, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
33
|
Chen YT, Lohia GK, Chen S, Riquelme SA. Immunometabolic Regulation of Bacterial Infection, Biofilms, and Antibiotic Susceptibility. J Innate Immun 2024; 16:143-158. [PMID: 38310854 PMCID: PMC10914382 DOI: 10.1159/000536649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 02/01/2024] [Indexed: 02/06/2024] Open
Abstract
BACKGROUND Upon infection, mucosal tissues activate a brisk inflammatory response to clear the pathogen, i.e., resistance to disease. Resistance to disease is orchestrated by tissue-resident macrophages, which undergo profound metabolic reprogramming after sensing the pathogen. These metabolically activated macrophages release many inflammatory factors, which promote their bactericidal function. However, in immunocompetent individuals, pathogens like Pseudomonas aeruginosa, Staphylococcus aureus, and Salmonella evade this type of immunity, generating communities that thrive for the long term. SUMMARY These organisms develop features that render them less susceptible to eradication, such as biofilms and increased tolerance to antibiotics. Furthermore, after antibiotic therapy withdrawal, "persister" cells rapidly upsurge, triggering inflammatory relapses that worsen host health. How these pathogens persisted in inflamed tissues replete with activated macrophages remains poorly understood. KEY MESSAGES In this review, we discuss recent findings indicating that the ability of P. aeruginosa, S. aureus, and Salmonella to evolve biofilms and antibiotic tolerance is promoted by the similar metabolic routes that regulate macrophage metabolic reprogramming.
Collapse
Affiliation(s)
- Ying-Tsun Chen
- Department of Pediatrics, Division of Infectious Diseases, Columbia University, New York, New York, USA
| | - Gaurav Kumar Lohia
- Department of Pediatrics, Division of Infectious Diseases, Columbia University, New York, New York, USA
| | - Samantha Chen
- Department of Pediatrics, Division of Infectious Diseases, Columbia University, New York, New York, USA
| | - Sebastián A Riquelme
- Department of Pediatrics, Division of Infectious Diseases, Columbia University, New York, New York, USA
| |
Collapse
|
34
|
Lewis IA. Boundary flux analysis: an emerging strategy for investigating metabolic pathway activity in large cohorts. Curr Opin Biotechnol 2024; 85:103027. [PMID: 38061263 DOI: 10.1016/j.copbio.2023.103027] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 11/02/2023] [Accepted: 11/15/2023] [Indexed: 02/09/2024]
Abstract
Many biological phenotypes are rooted in metabolic pathway activity rather than the concentrations of individual metabolites. Despite this, most metabolomics studies only capture steady-state metabolism - not metabolic flux. Although sophisticated metabolic flux analysis strategies have been developed, these methods are technically challenging and difficult to implement in large-cohort studies. Recently, a new boundary flux analysis (BFA) approach has emerged that captures large-scale metabolic flux phenotypes by quantifying changes in metabolite levels in the media of cultured cells. This approach is advantageous because it is relatively easy to implement yet captures complex metabolic flux phenotypes. We describe the opportunities and challenges of BFA and illustrate how it can be harnessed to investigate a wide transect of biological phenomena.
Collapse
Affiliation(s)
- Ian A Lewis
- Alberta Centre for Advanced Diagnostics, Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada.
| |
Collapse
|
35
|
Eberhart T, Stanley FU, Ricci L, Chirico T, Ferrarese R, Sisti S, Scagliola A, Baj A, Badurek S, Sommer A, Culp-Hill R, Dzieciatkowska M, Shokry E, Sumpton D, D'Alessandro A, Clementi N, Mancini N, Cardaci S. ACOD1 deficiency offers protection in a mouse model of diet-induced obesity by maintaining a healthy gut microbiota. Cell Death Dis 2024; 15:105. [PMID: 38302438 PMCID: PMC10834593 DOI: 10.1038/s41419-024-06483-2] [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: 09/07/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/03/2024]
Abstract
Aconitate decarboxylase 1 (ACOD1) is the enzyme synthesizing itaconate, an immuno-regulatory metabolite tuning host-pathogen interactions. Such functions are achieved by affecting metabolic pathways regulating inflammation and microbe survival. However, at the whole-body level, metabolic roles of itaconate remain largely unresolved. By using multiomics-integrated approaches, here we show that ACOD1 responds to high-fat diet consumption in mice by promoting gut microbiota alterations supporting metabolic disease. Genetic disruption of itaconate biosynthesis protects mice against obesity, alterations in glucose homeostasis and liver metabolic dysfunctions by decreasing meta-inflammatory responses to dietary lipid overload. Mechanistically, fecal metagenomics and microbiota transplantation experiments demonstrate such effects are dependent on an amelioration of the intestinal ecosystem composition, skewed by high-fat diet feeding towards obesogenic phenotype. In particular, unbiased fecal microbiota profiling and axenic culture experiments point towards a primary role for itaconate in inhibiting growth of Bacteroidaceae and Bacteroides, family and genus of Bacteroidetes phylum, the major gut microbial taxon associated with metabolic health. Specularly to the effects imposed by Acod1 deficiency on fecal microbiota, oral itaconate consumption enhances diet-induced gut dysbiosis and associated obesogenic responses in mice. Unveiling an unrecognized role of itaconate, either endogenously produced or exogenously administered, in supporting microbiota alterations underlying diet-induced obesity in mice, our study points ACOD1 as a target against inflammatory consequences of overnutrition.
Collapse
Affiliation(s)
- Tanja Eberhart
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Federico Uchenna Stanley
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Luisa Ricci
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Tiziana Chirico
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Roberto Ferrarese
- Laboratory of Medical Microbiology and Virology, Vita-Salute San Raffaele University, Milan, 20100, Italy
- IRCCS San Raffaele Hospital, Milan, 20100, Italy
- Synlab Italia, Castenedolo, BS, Italy
| | - Sofia Sisti
- Laboratory of Medical Microbiology and Virology, Vita-Salute San Raffaele University, Milan, 20100, Italy
- IRCCS San Raffaele Hospital, Milan, 20100, Italy
| | - Alessandra Scagliola
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- Istituto Nazionale di Genetica Molecolare, INGM, "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Andreina Baj
- Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Sylvia Badurek
- Preclinical Phenotyping Facility, Vienna BioCenter Core Facilities (VBCF), member of the Vienna BioCenter (VBC), Vienna, Austria
| | - Andreas Sommer
- Next Generation Sequencing Facility, Vienna BioCenter Core Facilities (VBCF), member of the Vienna BioCenter (VBC), Vienna, Austria
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | | | | | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Nicola Clementi
- Laboratory of Medical Microbiology and Virology, Vita-Salute San Raffaele University, Milan, 20100, Italy
- IRCCS San Raffaele Hospital, Milan, 20100, Italy
| | - Nicasio Mancini
- Laboratory of Medical Microbiology and Virology, Vita-Salute San Raffaele University, Milan, 20100, Italy
- IRCCS San Raffaele Hospital, Milan, 20100, Italy
- Laboratory of Medical Microbiology and Virology, Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
- Laboratory of Medical Microbiology and Virology, Fondazione Macchi University Hospital, Varese, Italy
| | - Simone Cardaci
- Cancer Metabolism Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
| |
Collapse
|
36
|
Yang L, Wang L, Wang M, Bajinka O, Wu G, Qin L, Tan Y. Oligoribonuclease mediates high adaptability of P. aeruginosa through metabolic conversion. BMC Microbiol 2024; 24:25. [PMID: 38238663 PMCID: PMC10797966 DOI: 10.1186/s12866-023-03175-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 12/26/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Oligoribonuclease (orn) of P. aeruginosa is a highly conserved exonuclease, which can regulate the global gene expression levels of bacteria through regulation of both the nanoRNA and c-di-GMP. NanoRNA can regulate the expression of the bacterial global genome as a transcription initiator, and c-di-GMP is the most widely second messenger in bacterial cells. OBJECTIVE This study seeks to elucidate on the regulation by orn on pathogenicity of P. aeruginosa. METHODS P. aeruginosa with orn deletion was constructed by suicide plasmid homologous recombination method. The possible regulatory process of orn was analyzed by TMT quantitative labeling proteomics. Then experiments were conducted to verify the changes of Δorn on bacterial motility, virulence and biofilm formation. Bacterial pathogenicity was further detected in cell and animal skin trauma models. ELISA detection c-di-GMP concentration and colony aggregation and biofilm formation were observed by scanning electron microscope. RESULTS orn deletion changed the global metabolism of P. aeruginosa and reduced intracellular energy metabolism. It leads to the disorder of the quorum sensing system, the reduction of bacterial motility and virulence factors pyocyanin and rhamnolipids. But, orn deletion enhanced pathogenicity in vitro and in vivo, a high level of c-di-GMP and biofilm development of P. aeruginosa. CONCLUSION orn regulates the ability of P. aeruginosa to adapt to the external environment.
Collapse
Affiliation(s)
- Lulu Yang
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Lili Wang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Mengyu Wang
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Ousman Bajinka
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Guojun Wu
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Ling Qin
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China.
| | - Yurong Tan
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China.
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China.
| |
Collapse
|
37
|
Maldarelli ME, Noto MJ. The emerging role for neutrophil mitochondrial metabolism in lung inflammation. IMMUNOMETABOLISM (COBHAM, SURREY) 2024; 6:e00036. [PMID: 38283697 PMCID: PMC10810349 DOI: 10.1097/in9.0000000000000036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 12/15/2023] [Indexed: 01/30/2024]
Abstract
Recent advances shed light on the importance of mitochondrial metabolism in supporting essential neutrophil functions such as trafficking, NETosis, bacterial killing, and modulating inflammatory responses. Mitochondrial metabolism is now recognized to contribute to a number of lung diseases marked by neutrophilic inflammation, including bacterial pneumonia, acute lung injury, and chronic obstructive pulmonary disease. In this mini review, we provide an overview of neutrophil metabolism focusing on the role of mitochondrial programs, discuss select neutrophil effector functions that are directly influenced by mitochondrial metabolism, and present what is known about the role for mitochondrial metabolism in lung diseases marked by neutrophilic inflammation.
Collapse
Affiliation(s)
- Mary E. Maldarelli
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michael J. Noto
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
38
|
Arumugam P, Kielian T. Metabolism Shapes Immune Responses to Staphylococcus aureus. J Innate Immun 2023; 16:12-30. [PMID: 38016430 PMCID: PMC10766399 DOI: 10.1159/000535482] [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: 10/04/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Staphylococcus aureus (S. aureus) is a common cause of hospital- and community-acquired infections that can result in various clinical manifestations ranging from mild to severe disease. The bacterium utilizes different combinations of virulence factors and biofilm formation to establish a successful infection, and the emergence of methicillin- and vancomycin-resistant strains introduces additional challenges for infection management and treatment. SUMMARY Metabolic programming of immune cells regulates the balance of energy requirements for activation and dictates pro- versus anti-inflammatory function. Recent investigations into metabolic adaptations of leukocytes and S. aureus during infection indicate that metabolic crosstalk plays a crucial role in pathogenesis. Furthermore, S. aureus can modify its metabolic profile to fit an array of niches for commensal or invasive growth. KEY MESSAGES Here we focus on the current understanding of immunometabolism during S. aureus infection and explore how metabolic crosstalk between the host and S. aureus influences disease outcome. We also discuss how key metabolic pathways influence leukocyte responses to other bacterial pathogens when information for S. aureus is not available. A better understanding of how S. aureus and leukocytes adapt their metabolic profiles in distinct tissue niches may reveal novel therapeutic targets to prevent or control invasive infections.
Collapse
Affiliation(s)
- Prabhakar Arumugam
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
39
|
Loi VV, Busche T, Kuropka B, Müller S, Methling K, Lalk M, Kalinowski J, Antelmann H. Staphylococcus aureus adapts to the immunometabolite itaconic acid by inducing acid and oxidative stress responses including S-bacillithiolations and S-itaconations. Free Radic Biol Med 2023; 208:859-876. [PMID: 37793500 DOI: 10.1016/j.freeradbiomed.2023.09.031] [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: 08/14/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023]
Abstract
Staphylococcus aureus is a major pathogen, which has to defend against reactive oxygen and electrophilic species encountered during infections. Activated macrophages produce the immunometabolite itaconate as potent electrophile and antimicrobial upon pathogen infection. In this work, we used transcriptomics, metabolomics and shotgun redox proteomics to investigate the specific stress responses, metabolic changes and redox modifications caused by sublethal concentrations of itaconic acid in S. aureus. In the RNA-seq transcriptome, itaconic acid caused the induction of the GlnR, KdpDE, CidR, SigB, GraRS, PerR, CtsR and HrcA regulons and the urease-encoding operon, revealing an acid and oxidative stress response and impaired proteostasis. Neutralization using external urea as ammonium source improved the growth and decreased the expression of the glutamine synthetase-controlling GlnR regulon, indicating that S. aureus experienced ammonium starvation upon itaconic acid stress. In the extracellular metabolome, the amounts of acetate and formate were decreased, while secretion of pyruvate and the neutral product acetoin were strongly enhanced to avoid intracellular acidification. Exposure to itaconic acid affected the amino acid uptake and metabolism as revealed by the strong intracellular accumulation of lysine, threonine, histidine, aspartate, alanine, valine, leucine, isoleucine, cysteine and methionine. In the proteome, itaconic acid caused widespread S-bacillithiolation and S-itaconation of redox-sensitive antioxidant and metabolic enzymes, ribosomal proteins and translation factors in S. aureus, supporting its oxidative and electrophilic mode of action in S. aureus. In phenotype analyses, the catalase KatA, the low molecular weight thiol bacillithiol and the urease provided protection against itaconic acid-induced oxidative and acid stress in S. aureus. Altogether, our results revealed that under physiological infection conditions, such as in the acidic phagolysome, itaconic acid is a highly effective antimicrobial against multi-resistant S. aureus isolates, which acts as weak acid causing an acid, oxidative and electrophilic stress response, leading to S-bacillithiolation and itaconation.
Collapse
Affiliation(s)
- Vu Van Loi
- Freie Universität Berlin, Institute of Biology-Microbiology, D-14195, Berlin, Germany
| | - Tobias Busche
- Microbial Genomics and Biotechnology, Center for Biotechnology, Bielefeld University, D-33615, Bielefeld, Germany
| | - Benno Kuropka
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, D-14195, Berlin, Germany
| | - Susanne Müller
- Freie Universität Berlin, Institute of Biology-Microbiology, D-14195, Berlin, Germany
| | - Karen Methling
- Department of Cellular Biochemistry and Metabolomics, University of Greifswald, 17487, Greifswald, Germany
| | - Michael Lalk
- Department of Cellular Biochemistry and Metabolomics, University of Greifswald, 17487, Greifswald, Germany
| | - Jörn Kalinowski
- Microbial Genomics and Biotechnology, Center for Biotechnology, Bielefeld University, D-33615, Bielefeld, Germany
| | - Haike Antelmann
- Freie Universität Berlin, Institute of Biology-Microbiology, D-14195, Berlin, Germany.
| |
Collapse
|
40
|
Li Y, Xu Y, Li W, Li J, Wu W, Kang J, Jiang H, Liu P, Liu J, Gong W, Li X, Ni C, Liu M, Chen L, Li S, Wu X, Zhao Y, Ren J. Itaconate inhibits SYK through alkylation and suppresses inflammation against hvKP induced intestinal dysbiosis. Cell Mol Life Sci 2023; 80:337. [PMID: 37897551 PMCID: PMC11073195 DOI: 10.1007/s00018-023-04971-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/23/2023] [Accepted: 09/18/2023] [Indexed: 10/30/2023]
Abstract
Hypervirulent Klebsiella pneumoniae (hvKP) is a highly lethal opportunistic pathogen that elicits more severe inflammatory responses compared to classical Klebsiella pneumoniae (cKP). In this study, we investigated the interaction between hvKP infection and the anti-inflammatory immune response gene 1 (IRG1)-itaconate axis. Firstly, we demonstrated the activation of the IRG1-itaconate axis induced by hvKP, with a dependency on SYK signaling rather than STING. Importantly, we discovered that exogenous supplementation of itaconate effectively inhibited excessive inflammation by directly inhibiting SYK kinase at the 593 site through alkylation. Furthermore, our study revealed that itaconate effectively suppressed the classical activation phenotype (M1 phenotype) and macrophage cell death induced by hvKP. In vivo experiments demonstrated that itaconate administration mitigated hvKP-induced disturbances in intestinal immunopathology and homeostasis, including the restoration of intestinal barrier integrity and alleviation of dysbiosis in the gut microbiota, ultimately preventing fatal injury. Overall, our study expands the current understanding of the IRG1-itaconate axis in hvKP infection, providing a promising foundation for the development of innovative therapeutic strategies utilizing itaconate for the treatment of hvKP infections.
Collapse
Affiliation(s)
- Yangguang Li
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Yu Xu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Weizhen Li
- School of Medicine, Anhui University of Science and Technology, Huainan, 232000, China
| | - Jiayang Li
- School of Medicine, Southeast University, Nanjing, 210000, China
| | - Wenqi Wu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jiaqi Kang
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Haiyang Jiang
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Peizhao Liu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Juanhan Liu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenbin Gong
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shanxi Province, China
| | - Xuanheng Li
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chujun Ni
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Mingda Liu
- The Core Laboratory, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Lijuan Chen
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Sicheng Li
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiuwen Wu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Yun Zhao
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210009, China.
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210009, China.
| |
Collapse
|
41
|
Tomlinson KL, Chen YT, Junker A, Urso A, Wong Fok Lung T, Ahn D, Hofstaedter CE, Baskota SU, Ernst RK, Prince A, Riquelme SA. Ketogenesis promotes tolerance to Pseudomonas aeruginosa pulmonary infection. Cell Metab 2023; 35:1767-1781.e6. [PMID: 37793346 PMCID: PMC10558090 DOI: 10.1016/j.cmet.2023.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/14/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023]
Abstract
Pseudomonas aeruginosa is a common cause of pulmonary infection. As a Gram-negative pathogen, it can initiate a brisk and highly destructive inflammatory response; however, most hosts become tolerant to the bacterial burden, developing chronic infection. Using a murine model of pneumonia, we demonstrate that this shift from inflammation to disease tolerance is promoted by ketogenesis. In response to pulmonary infection, ketone bodies are generated in the liver and circulate to the lungs where they impose selection for P. aeruginosa strains unable to display surface lipopolysaccharide (LPS). Such keto-adapted LPS strains fail to activate glycolysis and tissue-damaging cytokines and, instead, facilitate mitochondrial catabolism of fats and oxidative phosphorylation (OXPHOS), which maintains airway homeostasis. Within the lung, P. aeruginosa exploits the host immunometabolite itaconate to further stimulate ketogenesis. This environment enables host-P. aeruginosa coexistence, supporting both pathoadaptive changes in the bacteria and the maintenance of respiratory integrity via OXPHOS.
Collapse
Affiliation(s)
- Kira L Tomlinson
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Ying-Tsun Chen
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Alex Junker
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - AndreaCarola Urso
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | | | - Danielle Ahn
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Casey E Hofstaedter
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD 21201, USA
| | - Swikrity U Baskota
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Robert K Ernst
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD 21201, USA
| | - Alice Prince
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | | |
Collapse
|
42
|
Ben-Arosh H, Avraham R. Tissue-specific macrophage immunometabolism. Curr Opin Immunol 2023; 84:102369. [PMID: 37473458 DOI: 10.1016/j.coi.2023.102369] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/19/2023] [Accepted: 06/19/2023] [Indexed: 07/22/2023]
Abstract
Macrophages are phagocytic cells distributed across tissues that sustain homeostasis by constantly probing their local environment. Upon perturbations, macrophages rewire their energy metabolism to execute their immune programs. Intensive research in the field of immunometabolism highlights cell-intrinsic immunometabolites such as succinate and itaconate as immunomodulatory signals. A role for cell-extrinsic stimuli now emerges with evidence for signals that shape macrophages' metabolism in a tissue-specific manner. In this review, we will cover macrophage immunometabolism in the gut, a complex metabolic and immunologically active tissue. During homeostasis, gut macrophages are constantly exposed to pro-inflammatory ligands from the microbiota, and in contrast, are balanced by microbiota-derived anti-inflammatory metabolites. Given their extensive metabolic changes during activation, spatial analyses of the tissue will allow the characterization of metabolic niches of macrophage in the gut. Identifying metabolic perturbations of macrophage subsets during chronic inflammation and infection can direct future tissue-specific metabolotherapies.
Collapse
Affiliation(s)
- Hadar Ben-Arosh
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Roi Avraham
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
43
|
Kahl LJ, Stremmel N, Esparza-Mora MA, Wheatley RM, MacLean RC, Ralser M. Interkingdom interactions between Pseudomonas aeruginosa and Candida albicans affect clinical outcomes and antimicrobial responses. Curr Opin Microbiol 2023; 75:102368. [PMID: 37677865 DOI: 10.1016/j.mib.2023.102368] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 09/09/2023]
Abstract
Infections that involve interkingdom microbial communities, such as those between bacteria and yeast pathogens, are difficult to treat, associated with worse patient outcomes, and may be a source of antimicrobial resistance. In this review, we address co-occurrence and co-infections of Candida albicans and Pseudomonas aeruginosa, two pathogens that occupy multiple infection niches in the human body, especially in immunocompromised patients. The interaction between the pathogen species influences microbe-host interactions, the effectiveness of antimicrobials and even infection outcomes, and may thus require adapted treatment strategies. However, the molecular details of bacteria-fungal interactions both inside and outside the infection sites, are insufficiently characterised. We argue that comprehensively understanding the P. aeruginosa-C. albicans interaction network through integrated systems biology approaches will capture the highly dynamic and complex nature of these polymicrobial infections and lead to a more comprehensive understanding of clinical observations such as reshaped immune defences and low antimicrobial treatment efficacy.
Collapse
Affiliation(s)
- Lisa J Kahl
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | - Nina Stremmel
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany
| | | | - Rachel M Wheatley
- University of Oxford, Department of Biology, Oxford OX1 3SZ, United Kingdom
| | - R Craig MacLean
- University of Oxford, Department of Biology, Oxford OX1 3SZ, United Kingdom
| | - Markus Ralser
- Charité Universitätsmedizin Berlin, Department of Biochemistry, 10117 Berlin, Germany; University of Oxford, The Wellcome Centre for Human Genetics, Nuffield Department of Medicine, Oxford OX3 7BN, United Kingdom; Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany.
| |
Collapse
|
44
|
Kragh KN, Tolker-Nielsen T, Lichtenberg M. The non-attached biofilm aggregate. Commun Biol 2023; 6:898. [PMID: 37658117 PMCID: PMC10474055 DOI: 10.1038/s42003-023-05281-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 08/24/2023] [Indexed: 09/03/2023] Open
Abstract
Biofilms have conventionally been perceived as dense bacterial masses on surfaces, following the five-step model of development. Initial biofilm research focused on surface-attached formations, but detached aggregates have received increasing attention in the past decade due to their pivotal role in chronic infections. Understanding their nature sparked fervent discussions in biofilm conferences and scientific literature. This review consolidates current insights on non-attached aggregates, offering examples of their occurrence in nature and diseases. We discuss their formation and dispersion mechanisms, resilience to antibiotics and immune-responses, drawing parallels to surface-attached biofilms. Moreover, we outline available in vitro models for studying non-attached aggregates.
Collapse
Affiliation(s)
- Kasper N Kragh
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Tim Tolker-Nielsen
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Mads Lichtenberg
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
45
|
Hastings CJ, Syed SS, Marques CNH. Subversion of the Complement System by Pseudomonas aeruginosa. J Bacteriol 2023; 205:e0001823. [PMID: 37436150 PMCID: PMC10464199 DOI: 10.1128/jb.00018-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen heavily implicated in chronic diseases. Immunocompromised patients that become infected with P. aeruginosa usually are afflicted with a lifelong chronic infection, leading to worsened patient outcomes. The complement system is an integral piece of the first line of defense against invading microorganisms. Gram-negative bacteria are thought to be generally susceptible to attack from complement; however, P. aeruginosa can be an exception, with certain strains being serum resistant. Various molecular mechanisms have been described that confer P. aeruginosa unique resistance to numerous aspects of the complement response. In this review, we summarize the current published literature regarding the interactions of P. aeruginosa and complement, as well as the mechanisms used by P. aeruginosa to exploit various complement deficiencies and the strategies used to disrupt or hijack normal complement activities.
Collapse
Affiliation(s)
- Cody James Hastings
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | - Shazrah Salim Syed
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | - Cláudia Nogueira Hora Marques
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
46
|
Chimi LY, Bisso BN, Njateng GSS, Dzoyem JP. Antibiotic-Potentiating Effect of Some Bioactive Natural Products against Planktonic Cells, Biofilms, and Virulence Factors of Pseudomonas aeruginosa. BIOMED RESEARCH INTERNATIONAL 2023; 2023:9410609. [PMID: 37663785 PMCID: PMC10470073 DOI: 10.1155/2023/9410609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/25/2023] [Accepted: 08/12/2023] [Indexed: 09/05/2023]
Abstract
Background Pseudomonas aeruginosa is an opportunistic human pathogen that causes infections that are mediated by both virulence factor production and biofilm formation. In addition, many antibiotics are increasingly losing their efficacy due to the development of resistance. The screening of potentially bioactive natural compounds that have both antivirulence and antibiofilm activities to enhance antibiotic efficacy and reverse antibiotic resistance is a good strategy to overcome these issues. In this study, the antibacterial, antibiofilm, and antivirulence factor activities of some bioactive natural products in combination with conventional antibiotics were evaluated against clinical isolates of P. aeruginosa. Methods The broth microdilution method was used to determine the antibacterial and antibiofilm activities. The checkerboard method was used to evaluate the combination interactions. Spectrophotometric and agar plate techniques were used to assess the effect of the combination on the pyocyanin production and the motility in P. aeruginosa ATCC 27853 strain. Results Out of the eighteen combinations tested, ten exhibited synergistic effects against planktonic cells, seven against biofilm inhibition, and five against the eradication of mature biofilm of P. aeruginosa biofilm. The best synergistic effect was the association of amikacin and sinapic acid against planktonic cells (FICI = 0.08) with a 70-fold reduction in the MIC value of amikacin. The same combination showed significant synergistic inhibition of biofilm formation (FICI = 0.1) and biofilm eradication (FICI = 0.15) reducing the MBIC and MBEC of amikacin by 32-fold. Some selected synergistic combinations showed statistically significant differences (p < 0.01 or p < 0.001) in the inhibition of virulence factors compared to the antimicrobials alone. Conclusion In summary, this study revealed sinapic acid as an antibiotic adjuvant and antivirulence compound to overcome P. aeruginosa infections. This finding indicates that the combinations of amikacin plus sinapic acid, ceftazidime plus thymol, and norfloxacin plus curcumin could be considered promising candidates for the development of combination therapies targeting virulence factors against P. aeruginosa infections.
Collapse
Affiliation(s)
- Larissa Yetendje Chimi
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Borel Ndezo Bisso
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | | | - Jean Paul Dzoyem
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| |
Collapse
|
47
|
Forde AJ, Kolter J, Zwicky P, Baasch S, Lohrmann F, Eckert M, Gres V, Lagies S, Gorka O, Rambold AS, Buescher JM, Kammerer B, Lachmann N, Prinz M, Groß O, Pearce EJ, Becher B, Henneke P. Metabolic rewiring tunes dermal macrophages in staphylococcal skin infection. Sci Immunol 2023; 8:eadg3517. [PMID: 37566679 DOI: 10.1126/sciimmunol.adg3517] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 07/19/2023] [Indexed: 08/13/2023]
Abstract
The skin needs to balance tolerance of colonizing microflora with rapid detection of potential pathogens. Flexible response mechanisms would seem most suitable to accommodate the dynamic challenges of effective antimicrobial defense and restoration of tissue homeostasis. Here, we dissected macrophage-intrinsic mechanisms and microenvironmental cues that tune macrophage signaling in localized skin infection with the colonizing and opportunistic pathogen Staphylococcus aureus. Early in skin infection, the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) produced by γδ T cells and hypoxic conditions within the dermal microenvironment diverted macrophages away from a homeostatic M-CSF- and hypoxia-inducible factor 1α (HIF-1α)-dependent program. This allowed macrophages to be metabolically rewired for maximal inflammatory activity, which requires expression of Irg1 and generation of itaconate, but not HIF-1α. This multifactorial macrophage rewiring program was required for both the timely clearance of bacteria and for the provision of local immune memory. These findings indicate that immunometabolic conditioning allows dermal macrophages to cycle between antimicrobial activity and protection against secondary infections.
Collapse
Affiliation(s)
- Aaron James Forde
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Julia Kolter
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Pascale Zwicky
- Institute of Experimental Immunology, University of Zurich, CH-8057 Zurich, Switzerland
| | - Sebastian Baasch
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Florens Lohrmann
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
- Center for Pediatrics and Adolescent Medicine, University Medical Center, 79106 Freiburg, Germany
| | - Marleen Eckert
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Vitka Gres
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Simon Lagies
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
- 1 Core Competence Metabolomics, Institute of Organic Chemistry, University of Freiburg, 79104 Freiburg, Germany
| | - Oliver Gorka
- Institute of Neuropathology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Angelika S Rambold
- Department of Developmental Immunology, Max-Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Joerg M Buescher
- Department of Immunometabolism, Max-Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Bernd Kammerer
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
- 1 Core Competence Metabolomics, Institute of Organic Chemistry, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centre's BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Nico Lachmann
- Department of Pediatric Pneumology, Allergology and Neonatology and Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centre's BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- CIBSS-Center for Integrative Biological Signaling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Olaf Groß
- Institute of Neuropathology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centre's BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- CIBSS-Center for Integrative Biological Signaling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Edward J Pearce
- Department of Immunometabolism, Max-Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, CH-8057 Zurich, Switzerland
| | - Philipp Henneke
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Center for Pediatrics and Adolescent Medicine, University Medical Center, 79106 Freiburg, Germany
- CIBSS-Center for Integrative Biological Signaling Studies, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
48
|
Wu R, Liu J, Tang D, Kang R. The Dual Role of ACOD1 in Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:518-526. [PMID: 37549395 DOI: 10.4049/jimmunol.2300101] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 03/27/2023] [Indexed: 08/09/2023]
Abstract
Immunometabolism is an interdisciplinary field that focuses on the relationship between metabolic pathways and immune responses. Dysregulated immunometabolism contributes to many pathological settings, such as cytokine storm or immune tolerance. Aconitate decarboxylase 1 (ACOD1, also known as immunoresponsive gene 1), the mitochondrial enzyme responsible for catalyzing itaconate production, was originally identified as a bacterial LPS-inducible gene involved in innate immunity in mouse macrophages. We now know that the upregulation of ACOD1 expression in immune or nonimmune cells plays a context-dependent role in metabolic reprogramming, signal transduction, inflammasome regulation, and protein modification. The emerging function of ACOD1 in inflammation and infection is a double-edged sword. In this review, we discuss how ACOD1 regulates anti-inflammatory or proinflammatory responses in an itaconate-dependent or -independent manner. Further understanding of ACOD1 expression and function may pave the way for the development of precision therapies for inflammatory diseases.
Collapse
Affiliation(s)
- Runliu Wu
- Department of Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiao Liu
- DAMP Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
49
|
Dos Santos PAS, Rodrigues YC, Marcon DJ, Lobato ARF, Cazuza TB, Gouveia MIM, Silva MJA, Souza AB, Lima LNGC, Quaresma AJPG, Brasiliense DM, Lima KVB. Endemic High-Risk Clone ST277 Is Related to the Spread of SPM-1-Producing Pseudomonas aeruginosa during the COVID-19 Pandemic Period in Northern Brazil. Microorganisms 2023; 11:2069. [PMID: 37630629 PMCID: PMC10457858 DOI: 10.3390/microorganisms11082069] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/27/2023] Open
Abstract
Pseudomonas aeruginosa is a high-priority bacterial agent that causes healthcare-acquired infections (HAIs), which often leads to serious infections and poor prognosis in vulnerable patients. Its increasing resistance to antimicrobials, associated with SPM production, is a case of public health concern. Therefore, this study aims to determine the antimicrobial resistance, virulence, and genotyping features of P. aeruginosa strains producing SPM-1 in the Northern region of Brazil. To determine the presence of virulence and resistance genes, the PCR technique was used. For the susceptibility profile of antimicrobials, the Kirby-Bauer disk diffusion method was performed on Mueller-Hinton agar. The MLST technique was used to define the ST of the isolates. The exoS+/exoU- virulotype was standard for all strains, with the aprA, lasA, toxA, exoS, exoT, and exoY genes as the most prevalent. All the isolates showed an MDR or XDR profile against the six classes of antimicrobials tested. HRC ST277 played a major role in spreading the SPM-1-producing P. aeruginosa strains.
Collapse
Affiliation(s)
- Pabllo Antonny Silva Dos Santos
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil; (P.A.S.D.S.); (D.J.M.); (L.N.G.C.L.); (D.M.B.)
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (A.R.F.L.); (T.B.C.); (M.I.M.G.); (M.J.A.S.); (A.B.S.); (A.J.P.G.Q.)
| | - Yan Corrêa Rodrigues
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (A.R.F.L.); (T.B.C.); (M.I.M.G.); (M.J.A.S.); (A.B.S.); (A.J.P.G.Q.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
- Department of Natural Science, State University of Pará (DCNA/UEPA), Belém 66050-540, PA, Brazil
| | - Davi Josué Marcon
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil; (P.A.S.D.S.); (D.J.M.); (L.N.G.C.L.); (D.M.B.)
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (A.R.F.L.); (T.B.C.); (M.I.M.G.); (M.J.A.S.); (A.B.S.); (A.J.P.G.Q.)
| | - Amália Raiana Fonseca Lobato
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (A.R.F.L.); (T.B.C.); (M.I.M.G.); (M.J.A.S.); (A.B.S.); (A.J.P.G.Q.)
| | - Thalyta Braga Cazuza
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (A.R.F.L.); (T.B.C.); (M.I.M.G.); (M.J.A.S.); (A.B.S.); (A.J.P.G.Q.)
| | - Maria Isabel Montoril Gouveia
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (A.R.F.L.); (T.B.C.); (M.I.M.G.); (M.J.A.S.); (A.B.S.); (A.J.P.G.Q.)
| | - Marcos Jessé Abrahão Silva
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (A.R.F.L.); (T.B.C.); (M.I.M.G.); (M.J.A.S.); (A.B.S.); (A.J.P.G.Q.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | - Alex Brito Souza
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (A.R.F.L.); (T.B.C.); (M.I.M.G.); (M.J.A.S.); (A.B.S.); (A.J.P.G.Q.)
| | - Luana Nepomuceno Gondim Costa Lima
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil; (P.A.S.D.S.); (D.J.M.); (L.N.G.C.L.); (D.M.B.)
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (A.R.F.L.); (T.B.C.); (M.I.M.G.); (M.J.A.S.); (A.B.S.); (A.J.P.G.Q.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | - Ana Judith Pires Garcia Quaresma
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (A.R.F.L.); (T.B.C.); (M.I.M.G.); (M.J.A.S.); (A.B.S.); (A.J.P.G.Q.)
| | - Danielle Murici Brasiliense
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil; (P.A.S.D.S.); (D.J.M.); (L.N.G.C.L.); (D.M.B.)
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (A.R.F.L.); (T.B.C.); (M.I.M.G.); (M.J.A.S.); (A.B.S.); (A.J.P.G.Q.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | - Karla Valéria Batista Lima
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil; (P.A.S.D.S.); (D.J.M.); (L.N.G.C.L.); (D.M.B.)
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (A.R.F.L.); (T.B.C.); (M.I.M.G.); (M.J.A.S.); (A.B.S.); (A.J.P.G.Q.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
| |
Collapse
|
50
|
Maurice NM, Sadikot RT. Mitochondrial Dysfunction in Bacterial Infections. Pathogens 2023; 12:1005. [PMID: 37623965 PMCID: PMC10458073 DOI: 10.3390/pathogens12081005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
Mitochondria are critical in numerous cellular processes, including energy generation. Bacterial pathogens target host cell mitochondria through various mechanisms to disturb the host response and improve bacterial survival. We review recent advances in the understanding of how bacteria cause mitochondrial dysfunction through perturbations in mitochondrial cell-death pathways, energy production, mitochondrial dynamics, mitochondrial quality control, DNA repair, and the mitochondrial unfolded protein response. We also briefly highlight possible therapeutic approaches aimed at restoring the host mitochondrial function as a novel strategy to enhance the host response to bacterial infection.
Collapse
Affiliation(s)
- Nicholas M. Maurice
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
- Atlanta Veterans Affairs Health Care System, Decatur, GA 30033, USA
| | - Ruxana T. Sadikot
- VA Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|