1
|
Cheng ZL, Zhang S, Wang Z, Song A, Gao C, Song JB, Wang P, Zhang L, Zhou Y, Shan W, Zhang C, Zhang J, Sun Y, Xu Y, Lan F, Zhong M, Lyu LD, Huang G, Chen FX, Li G, Wang Z, Chen F, Xue J, Shi J, Liu Y, Lin Z, Wu D, Na J, Chen LL, Guan KL, Xiong Y, Ye D. Pathogen-derived glyoxylate inhibits Tet2 DNA dioxygenase to facilitate bacterial persister formation. Cell Metab 2025; 37:1137-1151.e5. [PMID: 40037360 DOI: 10.1016/j.cmet.2025.01.019] [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: 06/06/2024] [Revised: 11/15/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025]
Abstract
Pathogenic bacterial persistence enables survival during antibiotic treatment, leading to treatment failure and recurrent infections, yet its underlying mechanisms remain unclear. Here, we reveal that glyoxylate, a metabolite originally evolved for alternative carbon utilization, functions as a signaling molecule to reprogram the host transcriptome and promote persister formation. Glyoxylate inhibits the DNA dioxygenase TET2, suppressing pro-inflammatory gene expression and attenuating host immune defense. Notably, stimulating TET2 activity with vitamin C or blocking glyoxylate production by Salmonella reduces bacterial antibiotic resistance and improves infection treatment outcomes. Beyond its metabolic role, glyoxylate emerges as a regulator of host-pathogen interactions, while TET2 plays a critical role in preventing bacterial persistence. Our findings suggest that targeting glyoxylate production or enhancing TET2 activity offers promising therapeutic strategies to combat bacterial persistence and enhance the efficacy of antibiotic treatments.
Collapse
Affiliation(s)
- Zhou-Li Cheng
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Shuyuan Zhang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Zhenning Wang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Aixia Song
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Chao Gao
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Jun-Bin Song
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Pu Wang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Lei Zhang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yue Zhou
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenyan Shan
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chen Zhang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Jinye Zhang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yiping Sun
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yanhui Xu
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Fei Lan
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Ming Zhong
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liang-Dong Lyu
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Guanghua Huang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Fei Xavier Chen
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Gang Li
- Department of Laboratory Medicine, Jinshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zixin Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital affiliated to Tongji University, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Faying Chen
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital affiliated to Tongji University, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jianhuang Xue
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital affiliated to Tongji University, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jiejun Shi
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital affiliated to Tongji University, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yujun Liu
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Zihao Lin
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Duojiao Wu
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jim Na
- Cullgen Inc., 12671 High Bluff Drive, San Diego, CA 92130, USA
| | - Lei-Lei Chen
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Kun-Liang Guan
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Yue Xiong
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China.
| | - Dan Ye
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China; Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
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
|
3
|
Shi W, Li Y, Zhang W. Screening and functional characterization of isocitrate lyase AceA in the biofilm formation of Vibrio alginolyticus. Appl Environ Microbiol 2024; 90:e0069724. [PMID: 39377591 PMCID: PMC11577800 DOI: 10.1128/aem.00697-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/25/2024] [Indexed: 10/09/2024] Open
Abstract
Biofilm is a well-known sessile lifestyle for bacterial pathogens, but a little is known about the mechanism on biofilm formation in Vibrio alginolyticus. In this study, we screened V. alginolyticus strains with strong biofilm formation ability from coastal seawater. The antibiotic resistance of the biofilm cells (BFs) was higher than that of the planktonic cells (PTs). To study the genes and pathways involved in biofilm formation, we performed transcriptome analysis of the BFs and PTs of V. alginolyticus R9. A total of 685 differentially expressed genes (DEGs) were upregulated, and 517 DEGs were downregulated in the BFs. The upregulated DEGs were significantly enriched in several pathways including glyoxylate and dicarboxylate metabolism, while the downregulated genes were significantly enriched in the flagellar assembly pathways. The key gene involved in glyoxylate shunt, aceA, was cloned, and ΔaceA mutant was constructed to determine the function of AceA in carbon source utilization, biofilm formation, and virulence. Real-time reverse transcription PCR showed that the expression of aceA was higher at the mature stage but lower at the disperse stage of biofilm formation, and the expression of the flagellar related genes was upregulated in ΔaceA. This is the first study to illustrate the global gene expression profile during the biofilm formation of V. alginolyticus, and isocitrate lyase AceA, the key enzyme involved in glyoxylate shunt, was shown to maintain biofilms accompanied by downregulation of flagellation but promoted dispersal of BFs at the late stage.IMPORTANCEBiofilms pose serious public problems, not only protecting the cells in it from environmental hazard but also affecting the composition and abundance of bacteria, algae, fungi, and protozoa. The important opportunistic pathogen Vibrio alginolyticus is extremely ubiquitously present in seawater, and it also exhibited a strong ability to form biofilm; thus, investigation on the biofilm formation of V. alginolyticus at molecular level is fundamental for the deeper exploration of the environmental concerns arose by biofilm. In this study, transcriptome analysis of biofilm cells (BFs) and planktonic cells (PTs) from V. alginolyticus was performed and AceA was screened to play an important role in biofilm formation. AceA was shown to maintain biofilms accompanied by downregulation of flagellation but promoted dispersal of BFs at the disperse stage. This method was helpful to further understand the ability and mechanism of V. alginolyticus biofilm formation and provide clues for prevention of V. alginolyticus infection.
Collapse
Affiliation(s)
- Weibo Shi
- School of Marine Sciences, Ningbo University, Ningbo, P. R. China
| | - Ya Li
- School of Marine Sciences, Ningbo University, Ningbo, P. R. China
| | - Weiwei Zhang
- School of Marine Sciences, Ningbo University, Ningbo, P. R. China
| |
Collapse
|
4
|
McGettrick AF, Bourner LA, Dorsey FC, O'Neill LAJ. Metabolic Messengers: itaconate. Nat Metab 2024; 6:1661-1667. [PMID: 39060560 DOI: 10.1038/s42255-024-01092-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 07/01/2024] [Indexed: 07/28/2024]
Abstract
The metabolite itaconate has emerged as an important immunoregulator with roles in antibacterial defence, inhibition of inflammation and, more recently, as an inhibitory factor in obesity. Itaconate is one of the most upregulated metabolites in inflammatory macrophages. It is produced owing to the disturbance of the tricarboxylic acid cycle and the diversion of aconitate to itaconate via the enzyme aconitate decarboxylase 1. In immunology, initial studies concentrated on the role of itaconate in inflammatory macrophages where it was shown to be inhibitory, but this has expanded as the impact of itaconate on other cell types is starting to emerge. This review focuses on itaconate as a key immunoregulatory metabolite and describes its diverse mechanisms of action and its many impacts on the immune and inflammatory responses and in cancer. We also examine the clinical relevance of this immunometabolite and its therapeutic potential for immune and inflammatory diseases.
Collapse
Affiliation(s)
- A F McGettrick
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - L A Bourner
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - F C Dorsey
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - L A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
5
|
Xue Y, Xue B, Zhang L. Multi-Omics Integrative Analysis to Reveal the Impacts of Shewanella algae on the Development and Lifespan of Marine Nematode Litoditis marina. Int J Mol Sci 2024; 25:9111. [PMID: 39201797 PMCID: PMC11354469 DOI: 10.3390/ijms25169111] [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: 07/23/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Understanding how habitat bacteria affect animal development, reproduction, and aging is essential for deciphering animal biology. Our recent study showed that Shewanella algae impaired Litoditis marina development and lifespan, compared with Escherichia coli OP50 feeding; however, the underlying mechanisms remain unclear. Here, multi-omics approaches, including the transcriptome of both L. marina and bacteria, as well as the comparative bacterial metabolome, were utilized to investigate how bacterial food affects animal fitness and physiology. We found that genes related to iron ion binding and oxidoreductase activity pathways, such as agmo-1, cdo-1, haao-1, and tdo-2, were significantly upregulated in L. marina grown on S. algae, while extracellular structural components-related genes were significantly downregulated. Next, we observed that bacterial genes belonging to amino acid metabolism and ubiquinol-8 biosynthesis were repressed, while virulence genes were significantly elevated in S. algae. Furthermore, metabolomic analysis revealed that several toxic metabolites, such as puromycin, were enriched in S. algae, while many nucleotides were significantly enriched in OP50. Moreover, we found that the "two-component system" was enriched in S. algae, whereas "purine metabolism" and "one-carbon pool by folate" were significantly enriched in E. coli OP50. Collectively, our data provide new insights to decipher how diet modulates animal fitness and biology.
Collapse
Affiliation(s)
- Yiming Xue
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (Y.X.); (B.X.)
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Beining Xue
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (Y.X.); (B.X.)
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liusuo Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (Y.X.); (B.X.)
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| |
Collapse
|
6
|
Wu C, Wei X, Huang Z, Zheng Z, Zhang W, Chen J, Hong H, Li W. Urinary microbiome dysbiosis is associated with an inflammatory environment and perturbed fatty acids metabolism in the pathogenesis of bladder cancer. J Transl Med 2024; 22:628. [PMID: 38970045 PMCID: PMC11227203 DOI: 10.1186/s12967-024-05446-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Bladder cancer is a common malignancy with high recurrence rate. Early diagnosis and recurrence surveillance are pivotal to patients' outcomes, which require novel minimal-invasive diagnostic tools. The urinary microbiome is associated with bladder cancer and can be used as biomarkers, but the underlying mechanism is to be fully illustrated and diagnostic performance to be improved. METHODS A total of 23 treatment-naïve bladder cancer patients and 9 non-cancerous subjects were enrolled into the Before group and Control group. After surgery, 10 patients from the Before group were further assigned into After group. Void mid-stream urine samples were collected and sent for 16S rDNA sequencing, targeted metabolomic profiling, and flow cytometry. Next, correlations were analyzed between microbiota, metabolites, and cytokines. Finally, receiver operating characteristic (ROC) curves of the urinary biomarkers were plotted and compared. RESULTS Comparing to the Control group, levels of IL-6 (p < 0.01), IL-8 (p < 0.05), and IL-10 (p < 0.05) were remarkably elevated in the Before group. The α diversity of urine microbiome was also significantly higher, with the feature microbiota positively correlated to the level of IL-6 (r = 0.58, p < 0.01). Significant differences in metabolic composition were also observed between the Before and Control groups, with fatty acids and fatty acylcarnitines enriched in the Before group. After tumor resection, cytokine levels and the overall microbiome structure in the After group remained similar to that of the Before group, but fatty acylcarnitines were significantly reduced (p < 0.05). Pathway enrichment analysis revealed beta-oxidation of fatty acids was significantly involved (p < 0.001). ROC curves showed that the biomarker panel of Actinomycetaceae + arachidonic acid + IL-6 had superior diagnostic performance, with sensitivity of 0.94 and specificity of 1.00. CONCLUSIONS Microbiome dysbiosis, proinflammatory environment and altered fatty acids metabolism are involved in the pathogenesis of bladder cancer, which may throw light on novel noninvasive diagnostic tool development.
Collapse
Affiliation(s)
- Cen Wu
- Department of Urology, Fujian Medical University Affiliated Quanzhou First Hospital, Fujian, 362011, China
| | - Xiaoyu Wei
- Department of Urology, Fujian Medical University Affiliated Quanzhou First Hospital, Fujian, 362011, China
| | - Zhiyang Huang
- Department of Urology, Fujian Medical University Affiliated Quanzhou First Hospital, Fujian, 362011, China
| | - Zhixiong Zheng
- Department of Urology, Fujian Medical University Affiliated Quanzhou First Hospital, Fujian, 362011, China
| | - Wei Zhang
- Department of Urology, Fujian Medical University Affiliated Quanzhou First Hospital, Fujian, 362011, China
| | - Jiajun Chen
- Department of Urology, Fujian Medical University Affiliated Quanzhou First Hospital, Fujian, 362011, China
| | - Hongchang Hong
- Department of Urology, Fujian Medical University Affiliated Quanzhou First Hospital, Fujian, 362011, China
| | - Weili Li
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Biotechnology Co., Ltd., 180 Zhangheng Road, Pudong District, Shanghai, 201204, China.
| |
Collapse
|
7
|
Kreimendahl S, Pernas L. Metabolic immunity against microbes. Trends Cell Biol 2024; 34:496-508. [PMID: 38030541 DOI: 10.1016/j.tcb.2023.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/11/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023]
Abstract
Pathogens, including viruses, bacteria, fungi, and parasites, remodel the metabolism of their host to acquire the nutrients they need to proliferate. Thus, host cells are often perceived as mere exploitable nutrient pools during infection. Mounting reports challenge this perception and instead suggest that host cells can actively reprogram their metabolism to the detriment of the microbial invader. In this review, we present metabolic mechanisms that host cells use to defend against pathogens. We highlight the contribution of domesticated microbes to host defenses and discuss examples of host-pathogen arms races that are derived from metabolic conflict.
Collapse
Affiliation(s)
| | - Lena Pernas
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, USA; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| |
Collapse
|
8
|
Arruda MFC, da Silva Ramos RCP, de Oliveira NS, Rosa RT, Stuelp-Campelo PM, Bianchini LF, Villas-Bôas SG, Rosa EAR. Central Carbon Metabolism in Candida albicans Biofilms Is Altered by Dimethyl Sulfoxide. J Fungi (Basel) 2024; 10:337. [PMID: 38786692 PMCID: PMC11121877 DOI: 10.3390/jof10050337] [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: 02/23/2024] [Revised: 03/25/2024] [Accepted: 04/04/2024] [Indexed: 05/25/2024] Open
Abstract
The effect of dimethyl sulfoxide (DMSO) on fungal metabolism has not been well studied. This study aimed to evaluate, by metabolomics, the impact of DMSO on the central carbon metabolism of Candida albicans. Biofilms of C. albicans SC5314 were grown on paper discs, using minimum mineral (MM) medium, in a dynamic continuous flow system. The two experimental conditions were control and 0.03% DMSO (v/v). After 72 h of incubation (37 °C), the biofilms were collected and the metabolites were extracted. The extracted metabolites were subjected to gas chromatography-mass spectrometry (GC/MS). The experiment was conducted using five replicates on three independent occasions. The GC/MS analysis identified 88 compounds. Among the 88 compounds, the levels of 27 compounds were markedly different between the two groups. The DMSO group exhibited enhanced levels of putrescine and glutathione and decreased levels of methionine and lysine. Additionally, the DMSO group exhibited alterations in 13 metabolic pathways involved in primary and secondary cellular metabolism. Among the 13 altered pathways, seven were downregulated and six were upregulated in the DMSO group. These results indicated a differential intracellular metabolic profile between the untreated and DMSO-treated biofilms. Hence, DMSO was demonstrated to affect the metabolic pathways of C. albicans. These results suggest that DMSO may influence the results of laboratory tests when it is used as a solvent. Hence, the use of DMSO as a solvent must be carefully considered in drug research, as the effect of the researched drugs may not be reliably translated into clinical practice.
Collapse
Affiliation(s)
- Maria Fernanda Cordeiro Arruda
- Graduate Program on Dentistry, School of Medicine and Life Sciences, Pontifical Catholic University of Paraná, Curitiba 80215-901, Brazil; (M.F.C.A.); (R.C.P.d.S.R.)
| | - Romeu Cassiano Pucci da Silva Ramos
- Graduate Program on Dentistry, School of Medicine and Life Sciences, Pontifical Catholic University of Paraná, Curitiba 80215-901, Brazil; (M.F.C.A.); (R.C.P.d.S.R.)
| | - Nicoly Subtil de Oliveira
- Graduate Program on Animal Sciences, School of Medicine and Life Sciences, Pontifical Catholic University of Paraná, Curitiba 80215-901, Brazil;
| | - Rosimeire Takaki Rosa
- Xenobiotics Research Unit, School of Medicine and Life Sciences, Pontifical Catholic University of Paraná, Curitiba 80215-901, Brazil; (R.T.R.); (P.M.S.-C.); (L.F.B.)
| | - Patrícia Maria Stuelp-Campelo
- Xenobiotics Research Unit, School of Medicine and Life Sciences, Pontifical Catholic University of Paraná, Curitiba 80215-901, Brazil; (R.T.R.); (P.M.S.-C.); (L.F.B.)
| | - Luiz Fernando Bianchini
- Xenobiotics Research Unit, School of Medicine and Life Sciences, Pontifical Catholic University of Paraná, Curitiba 80215-901, Brazil; (R.T.R.); (P.M.S.-C.); (L.F.B.)
| | | | - Edvaldo Antonio Ribeiro Rosa
- Graduate Program on Dentistry, School of Medicine and Life Sciences, Pontifical Catholic University of Paraná, Curitiba 80215-901, Brazil; (M.F.C.A.); (R.C.P.d.S.R.)
- Graduate Program on Animal Sciences, School of Medicine and Life Sciences, Pontifical Catholic University of Paraná, Curitiba 80215-901, Brazil;
- Xenobiotics Research Unit, School of Medicine and Life Sciences, Pontifical Catholic University of Paraná, Curitiba 80215-901, Brazil; (R.T.R.); (P.M.S.-C.); (L.F.B.)
| |
Collapse
|
9
|
Sarkhel R, Priyadarsini S, Mahawar M. Nutrient limitation and oxidative stress induce the promoter of acetate operon in Salmonella Typhimurium. Arch Microbiol 2024; 206:126. [PMID: 38411730 DOI: 10.1007/s00203-024-03863-2] [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: 12/22/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/28/2024]
Abstract
Glyoxylate shunt is an important pathway for microorganisms to survive under multiple stresses. One of its enzymes, malate synthase (encoded by aceB gene), has been widely speculated for its contribution to both the pathogenesis and virulence of various microorganisms. We have previously demonstrated that malate synthase (MS) is required for the growth of Salmonella Typhimurium (S. Typhimurium) under carbon starvation and survival under oxidative stress conditions. The aceB gene is encoded by the acetate operon in S. Typhimurium. We attempted to study the activity of acetate promoter under both the starvation and oxidative stress conditions in a heterologous system. The lac promoter of the pUC19 plasmid was substituted with the putative promoter sequence of the acetate operon of S. Typhimurium upstream to the lacZ gene and transformed the vector construct into E. coli NEBα cells. The transformed cells were subjected to the stress conditions mentioned above. We observed a fourfold increase in the β-galactosidase activity in these cells resulting from the upregulation of the lacZ gene in the stationary phase of cell growth (nutrient deprived) as compared to the mid-log phase. Following exposure of stationary phase cells to hypochlorite-induced oxidative stress, we further observed a 1.6-fold increase in β galactosidase activity. These data suggest the induction of promoter activity of the acetate operon under carbon starvation and oxidative stress conditions. Thus, these observations corroborate our previous findings regarding the upregulation of aceB expression under stressful environments.
Collapse
Affiliation(s)
- Ratanti Sarkhel
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Swagatika Priyadarsini
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India.
- Indian Council of Agricultural Research- National Research Centre on Camel, Bikaner, Rajasthan, India.
| | - Manish Mahawar
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India.
| |
Collapse
|
10
|
Kumari A, Tripathi AH, Upadhyay SK, Gupta TM, Prakash PY. Enzymes conferring virulence traits among human pathogenic fungi. ENZYME BIOTECHNOLOGY FOR ENVIRONMENTAL SUSTAINABILITY 2024:339-362. [DOI: 10.1016/b978-0-443-22072-2.00001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
11
|
Cruz-Leite VRM, Moreira ALE, Silva LOS, Inácio MM, Parente-Rocha JA, Ruiz OH, Weber SS, Soares CMDA, Borges CL. Proteomics of Paracoccidioides lutzii: Overview of Changes Triggered by Nitrogen Catabolite Repression. J Fungi (Basel) 2023; 9:1102. [PMID: 37998907 PMCID: PMC10672198 DOI: 10.3390/jof9111102] [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: 10/17/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
Members of the Paracoccidioides complex are the causative agents of Paracoccidioidomycosis (PCM), a human systemic mycosis endemic in Latin America. Upon initial contact with the host, the pathogen needs to uptake micronutrients. Nitrogen is an essential source for biosynthetic pathways. Adaptation to nutritional stress is a key feature of fungi in host tissues. Fungi utilize nitrogen sources through Nitrogen Catabolite Repression (NCR). NCR ensures the scavenging, uptake and catabolism of alternative nitrogen sources, when preferential ones, such as glutamine or ammonium, are unavailable. The NanoUPLC-MSE proteomic approach was used to investigate the NCR response of Paracoccidioides lutzii after growth on proline or glutamine as a nitrogen source. A total of 338 differentially expressed proteins were identified. P. lutzii demonstrated that gluconeogenesis, β-oxidation, glyoxylate cycle, adhesin-like proteins, stress response and cell wall remodeling were triggered in NCR-proline conditions. In addition, within macrophages, yeast cells trained under NCR-proline conditions showed an increased ability to survive. In general, this study allows a comprehensive understanding of the NCR response employed by the fungus to overcome nutritional starvation, which in the human host is represented by nutritional immunity. In turn, the pathogen requires rapid adaptation to the changing microenvironment induced by macrophages to achieve successful infection.
Collapse
Affiliation(s)
- Vanessa Rafaela Milhomem Cruz-Leite
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences II, Federal University of Goiás, Goiânia 74690-900, GO, Brazil; (A.L.E.M.); (L.O.S.S.); (M.M.I.); (J.A.P.-R.); (C.M.d.A.S.)
| | - André Luís Elias Moreira
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences II, Federal University of Goiás, Goiânia 74690-900, GO, Brazil; (A.L.E.M.); (L.O.S.S.); (M.M.I.); (J.A.P.-R.); (C.M.d.A.S.)
| | - Lana O’Hara Souza Silva
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences II, Federal University of Goiás, Goiânia 74690-900, GO, Brazil; (A.L.E.M.); (L.O.S.S.); (M.M.I.); (J.A.P.-R.); (C.M.d.A.S.)
| | - Moises Morais Inácio
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences II, Federal University of Goiás, Goiânia 74690-900, GO, Brazil; (A.L.E.M.); (L.O.S.S.); (M.M.I.); (J.A.P.-R.); (C.M.d.A.S.)
- Estácio de Goiás University Center—FESGO, Goiânia 74063-010, GO, Brazil
| | - Juliana Alves Parente-Rocha
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences II, Federal University of Goiás, Goiânia 74690-900, GO, Brazil; (A.L.E.M.); (L.O.S.S.); (M.M.I.); (J.A.P.-R.); (C.M.d.A.S.)
| | - Orville Hernandez Ruiz
- MICROBA Research Group, Cellular and Molecular Biology Unit, Department of Microbiology, School of Microbiology, University of Antioquia, Medellín 050010, Colombia;
| | - Simone Schneider Weber
- Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso do Sul, Campo Grande 79304-902, MS, Brazil;
| | - Célia Maria de Almeida Soares
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences II, Federal University of Goiás, Goiânia 74690-900, GO, Brazil; (A.L.E.M.); (L.O.S.S.); (M.M.I.); (J.A.P.-R.); (C.M.d.A.S.)
| | - Clayton Luiz Borges
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences II, Federal University of Goiás, Goiânia 74690-900, GO, Brazil; (A.L.E.M.); (L.O.S.S.); (M.M.I.); (J.A.P.-R.); (C.M.d.A.S.)
| |
Collapse
|
12
|
Wang Y, Ferrinho S, Connaris H, Goss RJM. The Impact of Viral Infection on the Chemistries of the Earth's Most Abundant Photosynthesizes: Metabolically Talented Aquatic Cyanobacteria. Biomolecules 2023; 13:1218. [PMID: 37627283 PMCID: PMC10452541 DOI: 10.3390/biom13081218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Cyanobacteria are the most abundant photosynthesizers on earth, and as such, they play a central role in marine metabolite generation, ocean nutrient cycling, and the control of planetary oxygen generation. Cyanobacteriophage infection exerts control on all of these critical processes of the planet, with the phage-ported homologs of genes linked to photosynthesis, catabolism, and secondary metabolism (marine metabolite generation). Here, we analyze the 153 fully sequenced cyanophages from the National Center for Biotechnology Information (NCBI) database and the 45 auxiliary metabolic genes (AMGs) that they deliver into their hosts. Most of these AMGs are homologs of those found within cyanobacteria and play a key role in cyanobacterial metabolism-encoding proteins involved in photosynthesis, central carbon metabolism, phosphate metabolism, methylation, and cellular regulation. A greater understanding of cyanobacteriophage infection will pave the way to a better understanding of carbon fixation and nutrient cycling, as well as provide new tools for synthetic biology and alternative approaches for the use of cyanobacteria in biotechnology and sustainable manufacturing.
Collapse
Affiliation(s)
- Yunpeng Wang
- School of Chemistry, University of St Andrews, North Haugh, St Andrews KY16 9AJ, UK; (S.F.); (H.C.)
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews KY16 9SX, UK
| | - Scarlet Ferrinho
- School of Chemistry, University of St Andrews, North Haugh, St Andrews KY16 9AJ, UK; (S.F.); (H.C.)
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews KY16 9SX, UK
| | - Helen Connaris
- School of Chemistry, University of St Andrews, North Haugh, St Andrews KY16 9AJ, UK; (S.F.); (H.C.)
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews KY16 9SX, UK
| | - Rebecca J. M. Goss
- School of Chemistry, University of St Andrews, North Haugh, St Andrews KY16 9AJ, UK; (S.F.); (H.C.)
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews KY16 9SX, UK
| |
Collapse
|
13
|
Waldrop MP, Chabot CL, Liebner S, Holm S, Snyder MW, Dillon M, Dudgeon SR, Douglas TA, Leewis MC, Walter Anthony KM, McFarland JW, Arp CD, Bondurant AC, Taş N, Mackelprang R. Permafrost microbial communities and functional genes are structured by latitudinal and soil geochemical gradients. THE ISME JOURNAL 2023:10.1038/s41396-023-01429-6. [PMID: 37217592 DOI: 10.1038/s41396-023-01429-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/24/2023]
Abstract
Permafrost underlies approximately one quarter of Northern Hemisphere terrestrial surfaces and contains 25-50% of the global soil carbon (C) pool. Permafrost soils and the C stocks within are vulnerable to ongoing and future projected climate warming. The biogeography of microbial communities inhabiting permafrost has not been examined beyond a small number of sites focused on local-scale variation. Permafrost is different from other soils. Perennially frozen conditions in permafrost dictate that microbial communities do not turn over quickly, thus possibly providing strong linkages to past environments. Thus, the factors structuring the composition and function of microbial communities may differ from patterns observed in other terrestrial environments. Here, we analyzed 133 permafrost metagenomes from North America, Europe, and Asia. Permafrost biodiversity and taxonomic distribution varied in relation to pH, latitude and soil depth. The distribution of genes differed by latitude, soil depth, age, and pH. Genes that were the most highly variable across all sites were associated with energy metabolism and C-assimilation. Specifically, methanogenesis, fermentation, nitrate reduction, and replenishment of citric acid cycle intermediates. This suggests that adaptations to energy acquisition and substrate availability are among some of the strongest selective pressures shaping permafrost microbial communities. The spatial variation in metabolic potential has primed communities for specific biogeochemical processes as soils thaw due to climate change, which could cause regional- to global- scale variation in C and nitrogen processing and greenhouse gas emissions.
Collapse
Affiliation(s)
- Mark P Waldrop
- Geology, Minerals, Energy, and Geophysics Science Center, United States Geological Survey, Menlo Park, CA, 94025, USA.
| | - Christopher L Chabot
- California State University Northridge, 18111 Nordhoff St., Northridge, CA, 91330, USA
| | - Susanne Liebner
- GFZ German Research Centre for Geosciences, Section Geomicrobiology, 14473, Potsdam, Germany
- University of Potsdam, Institute of Biochemistry and Biology, 14476, Potsdam, Germany
| | - Stine Holm
- GFZ German Research Centre for Geosciences, Section Geomicrobiology, 14473, Potsdam, Germany
| | - Michael W Snyder
- California State University Northridge, 18111 Nordhoff St., Northridge, CA, 91330, USA
| | - Megan Dillon
- Earth and Environmental Sciences Area, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Steven R Dudgeon
- California State University Northridge, 18111 Nordhoff St., Northridge, CA, 91330, USA
| | - Thomas A Douglas
- U.S. Army Cold Regions Research and Engineering Laboratory 9th Avenue, Building 4070 Fort, Wainwright, AK, 99703, USA
| | - Mary-Cathrine Leewis
- Agriculture and Agri-Food Canada, 2560 Boulevard Hochelaga, Québec, QC, G1V 2J3, Canada
| | - Katey M Walter Anthony
- Water and Environmental Research Center, University Alaska Fairbanks, Fairbanks, AK, 99775, USA
| | - Jack W McFarland
- Geology, Minerals, Energy, and Geophysics Science Center, United States Geological Survey, Menlo Park, CA, 94025, USA
| | - Christopher D Arp
- Water and Environmental Research Center, University Alaska Fairbanks, Fairbanks, AK, 99775, USA
| | - Allen C Bondurant
- Water and Environmental Research Center, University Alaska Fairbanks, Fairbanks, AK, 99775, USA
| | - Neslihan Taş
- Earth and Environmental Sciences Area, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Rachel Mackelprang
- California State University Northridge, 18111 Nordhoff St., Northridge, CA, 91330, USA.
| |
Collapse
|
14
|
Chiriac MC, Haber M, Salcher MM. Adaptive genetic traits in pelagic freshwater microbes. Environ Microbiol 2023; 25:606-641. [PMID: 36513610 DOI: 10.1111/1462-2920.16313] [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: 09/24/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Pelagic microbes have adopted distinct strategies to inhabit the pelagial of lakes and oceans and can be broadly categorized in two groups: free-living, specialized oligotrophs and patch-associated generalists or copiotrophs. In this review, we aim to identify genomic traits that enable pelagic freshwater microbes to thrive in their habitat. To do so, we discuss the main genetic differences of pelagic marine and freshwater microbes that are both dominated by specialized oligotrophs and the difference to freshwater sediment microbes, where copiotrophs are more prevalent. We phylogenomically analysed a collection of >7700 metagenome-assembled genomes, classified habitat preferences on different taxonomic levels, and compared the metabolic traits of pelagic freshwater, marine, and freshwater sediment microbes. Metabolic differences are mainly associated with transport functions, environmental information processing, components of the electron transport chain, osmoregulation and the isoelectric point of proteins. Several lineages with known habitat transitions (Nitrososphaeria, SAR11, Methylophilaceae, Synechococcales, Flavobacteriaceae, Planctomycetota) and the underlying mechanisms in this process are discussed in this review. Additionally, the distribution, ecology and genomic make-up of the most abundant freshwater prokaryotes are described in details in separate chapters for Actinobacteriota, Bacteroidota, Burkholderiales, Verrucomicrobiota, Chloroflexota, and 'Ca. Patescibacteria'.
Collapse
Affiliation(s)
| | - Markus Haber
- Institute of Hydrobiology, Biology Centre CAS, Ceske Budejovice, Czechia
| | - Michaela M Salcher
- Institute of Hydrobiology, Biology Centre CAS, Ceske Budejovice, Czechia
| |
Collapse
|
15
|
The adaptive response to alternative carbon sources in the pathogen Candida albicans involves a remodeling of thiol- and glutathione-dependent redox status. Biochem J 2023; 480:197-217. [PMID: 36625375 DOI: 10.1042/bcj20220505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 01/11/2023]
Abstract
Candida albicans is an opportunist pathogen responsible for a large spectrum of infections, from superficial mycosis to systemic diseases known as candidiasis. During infection in vivo, Candida albicans must adapt to host microenvironments and this adaptive response is crucial for the survival of this organism, as it facilitates the effective assimilation of alternative carbon sources others than glucose. We performed a global proteomic analysis on the global changes in protein abundance in response to changes in micronutrient levels, and, in parallel, explored changes in the intracellular redox and metabolic status of the cells. We show here that each of the carbon sources considered - glucose, acetate and lactate - induces a unique pattern of response in C. albicans cells, and that some conditions trigger an original and specific adaptive response involving the adaptation of metabolic pathways, but also a complete remodeling of thiol-dependent antioxidant defenses. Protein S-thiolation and the overproduction of reduced glutathione are two components of the response to high glucose concentration. In the presence of acetate, glutathione-dependent oxidative stress occurs, reduced thiol groups bind to proteins, and glutathione is exported out of the cells, these changes probably being triggered by an increase in glutathione-S-transferases. Overall, our results suggest that the role of cellular redox status regulation and defenses against oxidative stress, including the thiol- and glutathione-dependent response, in the adaptive response of C. albicans to alternative carbon sources should be reconsidered.
Collapse
|
16
|
McGettrick AF, O'Neill LA. Two for the price of one: itaconate and its derivatives as an anti-infective and anti-inflammatory immunometabolite. Curr Opin Immunol 2023; 80:102268. [PMID: 36446152 DOI: 10.1016/j.coi.2022.102268] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022]
Abstract
The metabolite itaconate (ITA) and its derivatives, both chemically synthesized and endogenous, have emerged as immunoregulators, with roles in limiting inflammation but also having effects on bacterial and viral infection. Some members of the ITA family have been shown to target and inhibit multiple processes in macrophages with recently identified targets, including NLRP3, JAK1, ten-eleven translocation-2 dioxygenases, and TFEB, a key transcription factor for lysosomal biogenesis. They have also been shown to target multiple bacteria, inhibiting their replication, as well as having antiviral effects against viruses such as SARS-CoV2, Zika virus, and Influenza virus. The importance of ITA is highlighted by the fact that several pathogens have developed mechanisms to evade ITA and can manipulate ITA for their own gain. Two newly discovered isomers of ITA, mesaconate and citraconate, are also discussed, which also have immunomodulatory effects. ITA continues to be a fascination, both in terms of inflammation but also as an antibacterial and antiviral agent, with therapeutic potential in immune and inflammatory diseases.
Collapse
Affiliation(s)
- Anne F McGettrick
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland.
| | - Luke Aj O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| |
Collapse
|
17
|
De Bels D, Maillart E, Van Bambeke F, Redant S, Honoré PM. Existing and emerging therapies for the treatment of invasive candidiasis and candidemia. Expert Opin Emerg Drugs 2022; 27:405-416. [PMID: 36317695 DOI: 10.1080/14728214.2022.2142207] [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: 08/03/2021] [Accepted: 10/27/2022] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Invasive candidiasis or candidemia is a severe infection affecting more than 250,000 people worldwide every year. It is present in up to 16% of ICU patients. The prognosis of these infections is unfavorable, with global death estimated around 50,000 per year, which corresponds to up to 40% depending on patient severity and comorbidities. Therapeutic failure is not rare due to the emergence of multiresistant strains and of new species poorly responsive to current therapies like Candida auris. AREAS COVERED We first review the positioning of antifungal drugs used to treat candidiasis, namely polyenes, azoles, echinocandins and pyrimidine analogues. We then discuss the progresses brought by new formulations, new derivatives within these classes, compounds acting on new targets or repurposed drugs in terms of pharmacokinetic profile, spectrum of activity, potency, safety or risk of drug-drug interactions. EXPERT OPINION While new formulations (amphotericin B cochleate) improve oral bioavailability of the corresponding drugs, new azoles or echinocandins offer higher potency including against strains resistant to former generations of drugs. Repurposed drugs show synergism with current therapies in vitro. Results from ongoing and future clinical trials will be decisive to establish the interest for these drugs in our arsenal.
Collapse
Affiliation(s)
- David De Bels
- Intensive Care Department, Brugmann University Hospital, Brussels, Belgium
| | - Evelyne Maillart
- Department of Infectious Disease, Brugmann University Hospital, Brussels, Belgium
| | - Françoise Van Bambeke
- Louvain Drug Research Institute, Department of Pharmacology, Université Catholique de Louvain, Brussels, Belgium
| | - Sebastien Redant
- Intensive Care Department, Brugmann University Hospital, Brussels, Belgium
| | - Patrick M Honoré
- Intensive Care Department, Brugmann University Hospital, Brussels, Belgium
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- Department of ICU, CHU UCL Godinne-Namur, UCL Louvain Medical School
| |
Collapse
|
18
|
Decroës A, Mahillon M, Genard M, Lienard C, Lima-Mendez G, Gilmer D, Bragard C, Legrève A. Rhizomania: Hide and Seek of Polymyxa betae and the Beet Necrotic Yellow Vein Virus with Beta vulgaris. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2022; 35:989-1005. [PMID: 35816413 DOI: 10.1094/mpmi-03-22-0063-r] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The molecular interactions between Polymyxa betae, the protist vector of sugar beet viruses, beet necrotic yellow vein virus (BNYVV), the causal agent of rhizomania, and Beta vulgaris have not been extensively studied. Here, the transmission of BNYVV to sugar beet by P. betae zoospores was optimized using genetically characterized organisms. Molecular interactions of aviruliferous and viruliferous protist infection on sugar beet were highlighted by transcriptomic analysis. P. betae alone induced limited gene expression changes in sugar beet, as a biotrophic asymptomatic parasite. Most differentially expressed plant genes were down-regulated and included resistance gene analogs and cell wall peroxidases. Several enzymes involved in stress regulation, such as the glutathione-S-transferases, were significantly induced. With BNYVV, the first stages of the P. betae life cycle on sugar beet were accelerated with a faster increase of relative protist DNA level and an earlier appearance of sporangia and sporosori in plants roots. A clear activation of plant defenses and the modulation of genes involved in plant cell wall metabolism were observed. The P. betae transcriptome in the presence of BNYVV revealed induction of genes possibly involved in the switch to the survival stage. The interactions were different depending on the presence or absence of the virus. P. betae alone alleviates plant defense response, playing hide-and-seek with sugar beet and allowing for their mutual development. Conversely, BNYVV manipulates plant defense and promotes the rapid invasion of plant roots by P. betae. This accelerated colonization is accompanied by the development of thick-walled resting spores, supporting the virus survival. [Formula: see text] Copyright © 2022 The Author(s). This is an open access article distributed under the CC BY-NC-ND 4.0 International license.
Collapse
Affiliation(s)
- Alain Decroës
- Phytopathology-Applied Microbiology, Earth and Life Institute, UCLouvain, Louvain-la-Neuve, 1348, Belgium
| | | | - Margaux Genard
- Phytopathology-Applied Microbiology, Earth and Life Institute, UCLouvain, Louvain-la-Neuve, 1348, Belgium
| | - Charlotte Lienard
- Phytopathology-Applied Microbiology, Earth and Life Institute, UCLouvain, Louvain-la-Neuve, 1348, Belgium
| | - Gipsi Lima-Mendez
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Louvain-la-Neuve, 1348, Belgium
| | - David Gilmer
- Institut de biologie moléculaire des plantes, CNRS UPR2357, Université de Strasbourg, Strasbourg, 67084, France
| | - Claude Bragard
- Phytopathology-Applied Microbiology, Earth and Life Institute, UCLouvain, Louvain-la-Neuve, 1348, Belgium
| | - Anne Legrève
- Phytopathology-Applied Microbiology, Earth and Life Institute, UCLouvain, Louvain-la-Neuve, 1348, Belgium
| |
Collapse
|
19
|
Elicitor-Induced Metabolomics Analysis of Halodule pinifolia Suspension Culture for an Alternative Antifungal Screening Approach against Candida albicans. J Fungi (Basel) 2022; 8:jof8060609. [PMID: 35736092 PMCID: PMC9224785 DOI: 10.3390/jof8060609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/27/2022] [Accepted: 06/01/2022] [Indexed: 02/04/2023] Open
Abstract
Elicitors are the agents that stimulate the defense responses of plants, and accumulate specialized metabolites in plant tissue culture. This study investigated the elicitor-feeding response of H. pinifolia suspension cell cultures (SCC) for metabolomics analysis and screening of specialized compounds against Candida albicans. Methyl jasmonate (MeJA) was used as an elicitor, and treatment of SCC at a concentration of 20 µM MeJA resulted in the maximum rosmarinic acid (RA) accumulation (117 mg/g dry weight), with transcript levels of RA biosynthetic genes HpPAL, HpC4H, and Hp4CL being 4.2, 2.5, and 3.7-fold higher, respectively, than the controls. GC-MS-based metabolomics analysis revealed a total of 47 metabolites, including 30 organic acids, six amino acids, six flavonoids, two sugars, two plant growth regulators, and one vitamin, which were significantly different between control and MeJA-treated cells. Furthermore, five phenolic acids were discovered at higher concentrations, including p-anisic acid, p-coumaric acid, caffeic acid, vanillic acid, and rosmarinic acid, and were purified and structurally elucidated for alternative antifungal screening against C. albicans and the evaluation of ADMET properties. The results from antifungal screening revealed that RA at MIC of 31.25 mg/L exhibited the lowest growth percentage of C. albicans (1.99%), with higher inhibition of isocitrate lyase 1 (ICL 1) enzyme (93.1%), followed by p-anisic acid (86.2%) and caffeic acid (85.1%), respectively. The drug likeliness and ADMET properties of RA exhibited promising results, with a bioactivity score of 0.57, 0.15, and 0.24 for nuclear receptor ligand, protease inhibitor, and enzyme inhibitor, respectively. Therefore, MeJA appears to have a significant effect on enhanced RA accumulation in H. pinifoia cells with phenylpropanoid transcript expression, and acts as an ICL1 inhibitor of C. albicans.
Collapse
|
20
|
Xu X, Li T, Jin K. Bioinspired and Biomimetic Nanomedicines for Targeted Cancer Therapy. Pharmaceutics 2022; 14:1109. [PMID: 35631695 PMCID: PMC9147382 DOI: 10.3390/pharmaceutics14051109] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 02/05/2023] Open
Abstract
Undesirable side effects and multidrug resistance are the major obstacles in conventional chemotherapy towards cancers. Nanomedicines provide alternative strategies for tumor-targeted therapy due to their inherent properties, such as nanoscale size and tunable surface features. However, the applications of nanomedicines are hampered in vivo due to intrinsic disadvantages, such as poor abilities to cross biological barriers and unexpected off-target effects. Fortunately, biomimetic nanomedicines are emerging as promising therapeutics to maximize anti-tumor efficacy with minimal adverse effects due to their good biocompatibility and high accumulation abilities. These bioengineered agents incorporate both the physicochemical properties of diverse functional materials and the advantages of biological materials to achieve desired purposes, such as prolonged circulation time, specific targeting of tumor cells, and immune modulation. Among biological materials, mammalian cells (such as red blood cells, macrophages, monocytes, and neutrophils) and pathogens (such as viruses, bacteria, and fungi) are the functional components most often used to confer synthetic nanoparticles with the complex functionalities necessary for effective nano-biointeractions. In this review, we focus on recent advances in the development of bioinspired and biomimetic nanomedicines (such as mammalian cell-based drug delivery systems and pathogen-based nanoparticles) for targeted cancer therapy. We also discuss the biological influences and limitations of synthetic materials on the therapeutic effects and targeted efficacies of various nanomedicines.
Collapse
Affiliation(s)
- Xiaoqiu Xu
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tong Li
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Ke Jin
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
21
|
Abstract
Macrophages exposed to inflammatory stimuli including LPS undergo metabolic reprogramming to facilitate macrophage effector function. This metabolic reprogramming supports phagocytic function, cytokine release, and ROS production that are critical to protective inflammatory responses. The Krebs cycle is a central metabolic pathway within all mammalian cell types. In activated macrophages, distinct breaks in the Krebs cycle regulate macrophage effector function through the accumulation of several metabolites that were recently shown to have signaling roles in immunity. One metabolite that accumulates in macrophages because of the disturbance in the Krebs cycle is itaconate, which is derived from cis-aconitate by the enzyme cis-aconitate decarboxylase (ACOD1), encoded by immunoresponsive gene 1 (Irg1). This Review focuses on itaconate’s emergence as a key immunometabolite with diverse roles in immunity and inflammation. These roles include inhibition of succinate dehydrogenase (which controls levels of succinate, a metabolite with multiple roles in inflammation), inhibition of glycolysis at multiple levels (which will limit inflammation), activation of the antiinflammatory transcription factors Nrf2 and ATF3, and inhibition of the NLRP3 inflammasome. Itaconate and its derivatives have antiinflammatory effects in preclinical models of sepsis, viral infections, psoriasis, gout, ischemia/reperfusion injury, and pulmonary fibrosis, pointing to possible itaconate-based therapeutics for a range of inflammatory diseases. This intriguing metabolite continues to yield fascinating insights into the role of metabolic reprogramming in host defense and inflammation.
Collapse
|
22
|
Wijnants S, Vreys J, Van Dijck P. Interesting antifungal drug targets in the central metabolism of Candida albicans. Trends Pharmacol Sci 2021; 43:69-79. [PMID: 34756759 DOI: 10.1016/j.tips.2021.10.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 01/04/2023]
Abstract
To treat infections caused by Candida albicans, azoles, polyenes, and echinocandins are used. However, resistance occurs against all three, so there is an urgent need for new antifungal drugs with a novel mode of action. Recently, it became clear that central metabolism plays an important role in the virulence of C. albicans. Glycolysis is, for example, upregulated during virulence conditions, whereas the glyoxylate cycle is important upon phagocytosis by host immune cells. These findings indicate that C. albicans adapts its metabolism to the environment for maximal virulence. In this review, we provide an overview of the potency of different central metabolic pathways and their key enzymes as potential antifungal drug targets.
Collapse
Affiliation(s)
- Stefanie Wijnants
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, Leuven, Belgium; VIB-KU Leuven Center for Microbiology, Leuven, Belgium
| | - Jolien Vreys
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, Leuven, Belgium; VIB-KU Leuven Center for Microbiology, Leuven, Belgium
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, Leuven, Belgium; VIB-KU Leuven Center for Microbiology, Leuven, Belgium.
| |
Collapse
|
23
|
D'Arpa P, Karna SLR, Chen T, Leung KP. Pseudomonas aeruginosa transcriptome adaptations from colonization to biofilm infection of skin wounds. Sci Rep 2021; 11:20632. [PMID: 34667187 PMCID: PMC8526614 DOI: 10.1038/s41598-021-00073-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 09/07/2021] [Indexed: 11/09/2022] Open
Abstract
In burn patients Pseudomonas aeruginosa infection is a major cause of morbidity. Analysis of the pathogen's gene expression as it transitions from colonization to acute and then biofilm wound infection may provide strategies for infection control. Toward this goal, we seeded log-phase P. aeruginosa (PAO1) into 3-day-old, full-thickness excision wounds (rabbit ear) and harvested the bacteria during colonization (Hrs 2 and 6), acute infection (Hr 24), and biofilm infection (Days 5 and 9) for transcriptome analysis (RNA-Seq). After 2-6 h in the wound, genes for metabolism and cell replication were down-regulated while wound-adaptation genes were up-regulated (vs. expression in log-phase culture). As the infection progressed from acute to biofilm infection, more genes became up-regulated than down-regulated, but the down-regulated genes enriched in more pathways, likely because the genes and pathways that bacteria already colonizing wounds up-regulate to establish biofilm infection are less known. Across the stages of infection, carbon-utilization pathways shifted. During acute infection, itaconate produced by myeloid cells appears to have been a carbon source because myeloid cell infiltration and the expression of the host gene, ACOD1, for itaconate production peaked coincidently with the expression of the PAO1 genes for itaconate transport and catabolism. Additionally, branched-chain amino acids are suggested to be a carbon source in acute infection and in biofilm infection. In biofilm infection, fatty acid degradation was also up-regulated. These carbon sources feed into the glyoxylate cycle that was coincidently up-regulated, suggesting it provided the precursors for P. aeruginosa to synthesize macromolecules in establishing wound infection.
Collapse
Affiliation(s)
- Peter D'Arpa
- Combat Wound Repair Group and Tissue Regeneration Department, US Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX, USA.,The Geneva Foundation, Tacoma, USA
| | - S L Rajasekhar Karna
- Combat Wound Repair Group and Tissue Regeneration Department, US Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX, USA
| | - Tsute Chen
- The Forsyth Institute, Cambridge, MA, USA
| | - Kai P Leung
- Combat Wound Repair Group and Tissue Regeneration Department, US Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX, USA.
| |
Collapse
|
24
|
Medeiros Filho F, do Nascimento APB, Costa MDOCE, Merigueti TC, de Menezes MA, Nicolás MF, Dos Santos MT, Carvalho-Assef APD, da Silva FAB. A Systematic Strategy to Find Potential Therapeutic Targets for Pseudomonas aeruginosa Using Integrated Computational Models. Front Mol Biosci 2021; 8:728129. [PMID: 34616771 PMCID: PMC8488468 DOI: 10.3389/fmolb.2021.728129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/31/2021] [Indexed: 12/26/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogen that has been a constant global health problem due to its ability to cause infection at different body sites and its resistance to a broad spectrum of clinically available antibiotics. The World Health Organization classified multidrug-resistant Pseudomonas aeruginosa among the top-ranked organisms that require urgent research and development of effective therapeutic options. Several approaches have been taken to achieve these goals, but they all depend on discovering potential drug targets. The large amount of data obtained from sequencing technologies has been used to create computational models of organisms, which provide a powerful tool for better understanding their biological behavior. In the present work, we applied a method to integrate transcriptome data with genome-scale metabolic networks of Pseudomonas aeruginosa. We submitted both metabolic and integrated models to dynamic simulations and compared their performance with published in vitro growth curves. In addition, we used these models to identify potential therapeutic targets and compared the results to analyze the assumption that computational models enriched with biological measurements can provide more selective and (or) specific predictions. Our results demonstrate that dynamic simulations from integrated models result in more accurate growth curves and flux distribution more coherent with biological observations. Moreover, identifying drug targets from integrated models is more selective as the predicted genes were a subset of those found in the metabolic models. Our analysis resulted in the identification of 26 non-host homologous targets. Among them, we highlighted five top-ranked genes based on lesser conservation with the human microbiome. Overall, some of the genes identified in this work have already been proposed by different approaches and (or) are already investigated as targets to antimicrobial compounds, reinforcing the benefit of using integrated models as a starting point to selecting biologically relevant therapeutic targets.
Collapse
|
25
|
Demars A, Vitali A, Comein A, Carlier E, Azouz A, Goriely S, Smout J, Flamand V, Van Gysel M, Wouters J, Abendroth J, Edwards TE, Machelart A, Hoffmann E, Brodin P, De Bolle X, Muraille E. Aconitate decarboxylase 1 participates in the control of pulmonary Brucella infection in mice. PLoS Pathog 2021; 17:e1009887. [PMID: 34525130 PMCID: PMC8443048 DOI: 10.1371/journal.ppat.1009887] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022] Open
Abstract
Brucellosis is one of the most widespread bacterial zoonoses worldwide. Here, our aim was to identify the effector mechanisms controlling the early stages of intranasal infection with Brucella in C57BL/6 mice. During the first 48 hours of infection, alveolar macrophages (AMs) are the main cells infected in the lungs. Using RNA sequencing, we identified the aconitate decarboxylase 1 gene (Acod1; also known as Immune responsive gene 1), as one of the genes most upregulated in murine AMs in response to B. melitensis infection at 24 hours post-infection. Upregulation of Acod1 was confirmed by RT-qPCR in lungs infected with B. melitensis and B. abortus. We observed that Acod1-/- C57BL/6 mice display a higher bacterial load in their lungs than wild-type (wt) mice following B. melitensis or B. abortus infection, demonstrating that Acod1 participates in the control of pulmonary Brucella infection. The ACOD1 enzyme is mostly produced in mitochondria of macrophages, and converts cis-aconitate, a metabolite in the Krebs cycle, into itaconate. Dimethyl itaconate (DMI), a chemically-modified membrane permeable form of itaconate, has a dose-dependent inhibitory effect on Brucella growth in vitro. Interestingly, structural analysis suggests the binding of itaconate into the binding site of B. abortus isocitrate lyase. DMI does not inhibit multiplication of the isocitrate lyase deletion mutant ΔaceA B. abortus in vitro. Finally, we observed that, unlike the wt strain, the ΔaceA B. abortus strain multiplies similarly in wt and Acod1-/- C57BL/6 mice. These data suggest that bacterial isocitrate lyase might be a target of itaconate in AMs.
Collapse
Affiliation(s)
- Aurore Demars
- Unité de Recherche en Biologie des Microorganismes (URBM), NARILIS, University of Namur, Namur, Belgium
| | - Armelle Vitali
- Unité de Recherche en Biologie des Microorganismes (URBM), NARILIS, University of Namur, Namur, Belgium
| | - Audrey Comein
- Unité de Recherche en Biologie des Microorganismes (URBM), NARILIS, University of Namur, Namur, Belgium
| | - Elodie Carlier
- Unité de Recherche en Biologie des Microorganismes (URBM), NARILIS, University of Namur, Namur, Belgium
| | - Abdulkader Azouz
- Université Libre de Bruxelles, Institute for Medical Immunology, and ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| | - Stanislas Goriely
- Université Libre de Bruxelles, Institute for Medical Immunology, and ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| | - Justine Smout
- Université Libre de Bruxelles, Institute for Medical Immunology, and ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| | - Véronique Flamand
- Université Libre de Bruxelles, Institute for Medical Immunology, and ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| | - Mégane Van Gysel
- Namur Medicine and Drug Innovation Center (NAMEDIC), Namur Research Institute for Life Sciences (Narilis), Department of Chemistry, Laboratoire de Chimie Biologique Structurale (CBS), Namur, Belgium
| | - Johan Wouters
- Namur Medicine and Drug Innovation Center (NAMEDIC), Namur Research Institute for Life Sciences (Narilis), Department of Chemistry, Laboratoire de Chimie Biologique Structurale (CBS), Namur, Belgium
| | - Jan Abendroth
- UCB BioSciences, 7869 NE Day Road West Bainbridge Island, WA 98110 USA and Seattle Structural Genomics Center for Infectious Disease, Seattle, Washington, United States of America
| | - Thomas E. Edwards
- UCB BioSciences, 7869 NE Day Road West Bainbridge Island, WA 98110 USA and Seattle Structural Genomics Center for Infectious Disease, Seattle, Washington, United States of America
| | - Arnaud Machelart
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Eik Hoffmann
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Priscille Brodin
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019—UMR 9017—CIIL—Center for Infection and Immunity of Lille, Lille, France
| | - Xavier De Bolle
- Unité de Recherche en Biologie des Microorganismes (URBM), NARILIS, University of Namur, Namur, Belgium
| | - Eric Muraille
- Unité de Recherche en Biologie des Microorganismes (URBM), NARILIS, University of Namur, Namur, Belgium
- Université Libre de Bruxelles, Laboratoire de Parasitologie, and ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| |
Collapse
|
26
|
Postharvest Drying Techniques Regulate Secondary Metabolites and Anti-Neuroinflammatory Activities of Ganoderma lucidum. Molecules 2021; 26:molecules26154484. [PMID: 34361637 PMCID: PMC8347575 DOI: 10.3390/molecules26154484] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 01/11/2023] Open
Abstract
Ganoderma lucidum extract is a potent traditional remedy for curing various ailments. Drying is the most important postharvest step during the processing of Ganoderma lucidum. The drying process mainly involves heat (36 h at 60 °C) and freeze-drying (36 h at −80 °C). We investigated the effects of different postharvest drying protocols on the metabolites profiling of Ganoderma lucidum using GC-MS, followed by an investigation of the anti-neuroinflammatory potential in LPS-treated BV2 microglial cells. A total of 109 primary metabolites were detected from heat and freeze-dried samples. Primary metabolite profiling showed higher levels of amino acids (17.4%) and monosaccharides (8.8%) in the heat-dried extracts, whereas high levels of organic acids (64.1%) were present in the freeze-dried samples. The enzymatic activity, such as ATP-citrate synthase, pyruvate kinase, glyceraldehyde-3-phosphatase dehydrogenase, glutamine synthase, fructose-bisphosphate aldolase, and D-3-phosphoglycerate dehydrogenase, related to the reverse tricarboxylic acid cycle were significantly high in the heat-dried samples. We also observed a decreased phosphorylation level of the MAP kinase (Erk1/2, p38, and JNK) and NF-κB subunit p65 in the heat-dried samples of the BV2 microglia cells. The current study suggests that heat drying improves the production of ganoderic acids by the upregulation of TCA-related pathways, which, in turn, gives a significant reduction in the inflammatory response of LPS-induced BV2 cells. This may be attributed to the inhibition of NF-κB and MAP kinase signaling pathways in cells treated with heat-dried extracts.
Collapse
|
27
|
Lara AC, Corretto E, Kotrbová L, Lorenc F, Petříčková K, Grabic R, Chroňáková A. The Genome Analysis of the Human Lung-Associated Streptomyces sp. TR1341 Revealed the Presence of Beneficial Genes for Opportunistic Colonization of Human Tissues. Microorganisms 2021; 9:1547. [PMID: 34442631 PMCID: PMC8401907 DOI: 10.3390/microorganisms9081547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/12/2021] [Accepted: 07/17/2021] [Indexed: 12/16/2022] Open
Abstract
Streptomyces sp. TR1341 was isolated from the sputum of a man with a history of lung and kidney tuberculosis, recurrent respiratory infections, and COPD. It produces secondary metabolites associated with cytotoxicity and immune response modulation. In this study, we complement our previous results by identifying the genetic features associated with the production of these secondary metabolites and other characteristics that could benefit the strain during its colonization of human tissues (virulence factors, modification of the host immune response, or the production of siderophores). We performed a comparative phylogenetic analysis to identify the genetic features that are shared by environmental isolates and human respiratory pathogens. The results showed a high genomic similarity of Streptomyces sp. TR1341 to the plant-associated Streptomyces sp. endophyte_N2, inferring a soil origin of the strain. Putative virulence genes, such as mammalian cell entry (mce) genes were not detected in the TR1341's genome. The presence of a type VII secretion system, distinct from the ones found in Mycobacterium species, suggests a different colonization strategy than the one used by other actinomycete lung pathogens. We identified a higher diversity of genes related to iron acquisition and demonstrated that the strain produces ferrioxamine B in vitro. These results indicate that TR1341 may have an advantage in colonizing environments that are low in iron, such as human tissue.
Collapse
Affiliation(s)
- Ana Catalina Lara
- Institute of Soil Biology, Biology Centre Academy of Sciences of The Czech Republic, Na Sádkách 702/7, 37005 České Budějovice, Czech Republic; (A.C.L.); (E.C.); (L.K.); (F.L.)
| | - Erika Corretto
- Institute of Soil Biology, Biology Centre Academy of Sciences of The Czech Republic, Na Sádkách 702/7, 37005 České Budějovice, Czech Republic; (A.C.L.); (E.C.); (L.K.); (F.L.)
| | - Lucie Kotrbová
- Institute of Soil Biology, Biology Centre Academy of Sciences of The Czech Republic, Na Sádkách 702/7, 37005 České Budějovice, Czech Republic; (A.C.L.); (E.C.); (L.K.); (F.L.)
| | - František Lorenc
- Institute of Soil Biology, Biology Centre Academy of Sciences of The Czech Republic, Na Sádkách 702/7, 37005 České Budějovice, Czech Republic; (A.C.L.); (E.C.); (L.K.); (F.L.)
| | - Kateřina Petříčková
- Institute of Immunology and Microbiology, 1st Faculty of Medicine, Charles University, Studničkova 7, 12800 Prague 2, Czech Republic;
- Faculty of Science, University of South Bohemia, Branišovská 1645/31a, 37005 České Budějovice, Czech Republic
| | - Roman Grabic
- Faculty of Fisheries and Protection of Waters, University of South Bohemia, Zátiší 728/II, 38925 Vodňany, Czech Republic;
| | - Alica Chroňáková
- Institute of Soil Biology, Biology Centre Academy of Sciences of The Czech Republic, Na Sádkách 702/7, 37005 České Budějovice, Czech Republic; (A.C.L.); (E.C.); (L.K.); (F.L.)
| |
Collapse
|
28
|
Hwang W, Yong JH, Min KB, Lee KM, Pascoe B, Sheppard SK, Yoon SS. Genome-wide association study of signature genetic alterations among pseudomonas aeruginosa cystic fibrosis isolates. PLoS Pathog 2021; 17:e1009681. [PMID: 34161396 PMCID: PMC8274868 DOI: 10.1371/journal.ppat.1009681] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/12/2021] [Accepted: 05/31/2021] [Indexed: 12/18/2022] Open
Abstract
Pseudomonas aeruginosa (PA) is an opportunistic pathogen that causes diverse human infections including chronic airway infection in patients with cystic fibrosis (CF). Comparing the genomes of CF and non-CF PA isolates has great potential to identify the genetic basis of pathogenicity. To gain a deeper understanding of PA adaptation in CF airways, we performed a genome-wide association study (GWAS) on 1,001 PA genomes. Genetic variations identified among CF isolates were categorized into (i) alterations in protein-coding regions, either large- or small-scale, and (ii) polymorphic variation in intergenic regions. We introduced each CF-associated genetic alteration into the genome of PAO1, a prototype PA strain, and validated the outcomes experimentally. Loci readily mutated among CF isolates included genes encoding a probable sulfatase, a probable TonB-dependent receptor (PA2332~PA2336), L-cystine transporter (YecS, PA0313), and a probable transcriptional regulator (PA5438). A promoter region of a heme/hemoglobin uptake outer membrane receptor (PhuR, PA4710) was also different between the CF and non-CF isolate groups. Our analysis highlights ways in which the PA genome evolves to survive and persist within the context of chronic CF infection.
Collapse
Affiliation(s)
- Wontae Hwang
- Department of Microbiology and Immunology, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Seoul, Republic of Korea
| | - Ji Hyun Yong
- Department of Microbiology and Immunology, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Seoul, Republic of Korea
| | - Kyung Bae Min
- Department of Microbiology and Immunology, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Seoul, Republic of Korea
| | - Kang-Mu Lee
- Department of Microbiology and Immunology, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Seoul, Republic of Korea
| | - Ben Pascoe
- The Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, United Kingdom
| | - Samuel K Sheppard
- The Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, United Kingdom
| | - Sang Sun Yoon
- Department of Microbiology and Immunology, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Seoul, Republic of Korea
- Institute for Immunology and Immunological Diseases, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
29
|
Qiu X, Bajinka O, Wang L, Wu G, Tan Y. High-fat diet promotes epithelial-mesenchymal transition through enlarged growth of opportunistic pathogens and the intervention of saturated hydrogen. Am J Transl Res 2021; 13:6016-6030. [PMID: 34306341 PMCID: PMC8290812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/25/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVES This study investigated the effects and mechanism of high-fat diet on the epithelial-mesenchymal transition (EMT) of respiratory tract and the intervention of saturated hydrogen on it. METHODS 80 five-week-old C57BL6/J male mice were randomly divided into normal control group, H2 group, high-fat (HF) group and HF+H2 group, making 20 mice in each group. The weights of the mice were measured on weekly basis. Six mice from each group were executed at every second week. Blood samples were collected for lipid testing. Lung tissues were collected for 16S rRNA gene sequencing, HE staining, immunofluorescence and quantitative real-time PCR (qPCR). RESULTS Compared with the control group, the mice in the HF group showed increased inflammatory cell infiltration, decreased expression of e-cadherin (E-cad) and increased expression of Twist. There were significant differences in the composition of bacteria in the lung, and the expression of isocitrate lyase (ICL) genes in Pseudomonas aeruginosa, Staphylococcus aureus and Acinetobacter baumannii, which were significantly associated with asthma were seen with a significant increasing trend. After the treatment of saturated hydrogen, the changes in lung microbial population, lung tissue infiltration of inflammatory cells and the transformation of epithelial stroma caused by high-fat diet were moderately alleviated. CONCLUSION High-fat diet can promote inflammation and EMT in the lung by enlarging the growth of glyoxylic acid cycle-dependent bacteria, and the pathological process are partly alleviated by saturated hydrogen.
Collapse
Affiliation(s)
- Xiangjie Qiu
- Department of Medical Microbiology, Xiangya School of Medicine, Central South UniversityChangsha, China
| | - Ousman Bajinka
- Department of Medical Microbiology, Xiangya School of Medicine, Central South UniversityChangsha, China
| | - Lili Wang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South UniversityChangsha, China
| | - Guojun Wu
- Department of Medical Microbiology, Xiangya School of Medicine, Central South UniversityChangsha, China
| | - Yurong Tan
- Department of Medical Microbiology, Xiangya School of Medicine, Central South UniversityChangsha, China
- China-Africa Research Center of Infectious Diseases, School of Basic Medical Sciences, Central South UniversityChangsha 410078, Hunan, China
| |
Collapse
|
30
|
Laurian R, Ravent J, Dementhon K, Lemaire M, Soulard A, Cotton P. Candida albicans Hexokinase 2 Challenges the Saccharomyces cerevisiae Moonlight Protein Model. Microorganisms 2021; 9:microorganisms9040848. [PMID: 33920979 PMCID: PMC8071269 DOI: 10.3390/microorganisms9040848] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 12/20/2022] Open
Abstract
Survival of the pathogenic yeast Candida albicans depends upon assimilation of fermentable and non-fermentable carbon sources detected in host microenvironments. Among the various carbon sources encountered in a human body, glucose is the primary source of energy. Its effective detection, metabolism and prioritization via glucose repression are primordial for the metabolic adaptation of the pathogen. In C. albicans, glucose phosphorylation is mainly performed by the hexokinase 2 (CaHxk2). In addition, in the presence of glucose, CaHxK2 migrates in the nucleus and contributes to the glucose repression signaling pathway. Based on the known dual function of the Saccharomyces cerevisiae hexokinase 2 (ScHxk2), we intended to explore the impact of both enzymatic and regulatory functions of CaHxk2 on virulence, using a site-directed mutagenesis approach. We show that the conserved aspartate residue at position 210, implicated in the interaction with glucose, is essential for enzymatic and glucose repression functions but also for filamentation and virulence in macrophages. Point mutations and deletion into the N-terminal region known to specifically affect glucose repression in ScHxk2 proved to be ineffective in CaHxk2. These results clearly show that enzymatic and regulatory functions of the hexokinase 2 cannot be unlinked in C. albicans.
Collapse
Affiliation(s)
- Romain Laurian
- INSA Lyon, CNRS, Université de Lyon, Université Claude Bernard Lyon1, UMR5240 MAP, 69622 Villeurbanne, France; (R.L.); (J.R.); (M.L.); (A.S.)
| | - Jade Ravent
- INSA Lyon, CNRS, Université de Lyon, Université Claude Bernard Lyon1, UMR5240 MAP, 69622 Villeurbanne, France; (R.L.); (J.R.); (M.L.); (A.S.)
| | - Karine Dementhon
- UMR-CNRS 5234, Laboratoire de Microbiologie Fondamentale et Pathogénicité, Université de Bordeaux, 33076 Bordeaux, France;
| | - Marc Lemaire
- INSA Lyon, CNRS, Université de Lyon, Université Claude Bernard Lyon1, UMR5240 MAP, 69622 Villeurbanne, France; (R.L.); (J.R.); (M.L.); (A.S.)
| | - Alexandre Soulard
- INSA Lyon, CNRS, Université de Lyon, Université Claude Bernard Lyon1, UMR5240 MAP, 69622 Villeurbanne, France; (R.L.); (J.R.); (M.L.); (A.S.)
| | - Pascale Cotton
- INSA Lyon, CNRS, Université de Lyon, Université Claude Bernard Lyon1, UMR5240 MAP, 69622 Villeurbanne, France; (R.L.); (J.R.); (M.L.); (A.S.)
- Correspondence:
| |
Collapse
|
31
|
Hans S, Fatima Z, Hameed S. Metabolic fitness of Candida albicans is indispensable for functional drug efflux, ergosterol, and chitin biosynthesis. Curr Med Mycol 2021; 6:9-14. [PMID: 33834137 PMCID: PMC8018820 DOI: 10.18502/cmm.6.3.3980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background and Purpose: The increment in fungal infections, particularly due to Candida species, is alarming due to
the emergence of multidrug resistance (MDR). Hence, the identification of novel drug targets to circumvent the problem
of MDR requires immediate attention. The metabolic pathway, such as glyoxylate cycle (GC), which utilizes
key enzymes (isocitrate lyase [ICL] and malate synthase [MLS]), enables C. albicans
to adapt under glucose-deficient conditions. This study uncovers the effect of GC disruption on the major MDR mechanisms of C. albicans as a human pathogenic fungus. Materials and Methods: For the purpose of the study, efflux pump activity was assessed by phenotypic susceptibilities in the presence of substrates rhodamine 6G (R6G) and Nile red, along with R6G extracellular concentration (527 nm). In addition, ergosterol content was estimated by the alcoholic potassium hydroxide hydrolysis method. The estimation of chitin was also accomplished by the absorbance (520 nm) of glucosamine released by acid hydrolysis. Results: The results revealed that the disruption of ICL enzyme gene (Δicl1) led to the impairment of the efflux activity of multidrug transporters belonging to the ATP - binding cassette superfamily. It was further shown that Δicl1 mutant exhibited diminished ergosterol and chitin contents. In addition, all abrogated phenotypes could be rescued in the reverting strain of Δicl1 mutant. Conclusion: Based on the findings, the disruption of GC affected efflux activity and the synthesis of ergosterol and chitin. The present study for the first time revealed that metabolic fitness was associated with functional drug efflux, ergosterol and chitin biosynthesis and validated GC as an antifungal target. However, further studies are needed to comprehend and exploit this therapeutic opportunity.
Collapse
Affiliation(s)
- Sandeep Hans
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram (Manesar)-122413, India
| | - Zeeshan Fatima
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram (Manesar)-122413, India
| | - Saif Hameed
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram (Manesar)-122413, India
| |
Collapse
|
32
|
Amorim-Vaz S, Coste AT, Tran VDT, Pagni M, Sanglard D. Function Analysis of MBF1, a Factor Involved in the Response to Amino Acid Starvation and Virulence in Candida albicans. FRONTIERS IN FUNGAL BIOLOGY 2021; 2:658899. [PMID: 37744106 PMCID: PMC10512259 DOI: 10.3389/ffunb.2021.658899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 02/15/2021] [Indexed: 09/26/2023]
Abstract
Candida albicans is a commensal of human mucosae, but also one of the most common fungal pathogens of humans. Systemic infections caused by this fungus, mostly affecting immunocompromised patients, are associated to fatality rates as high as 50% despite the available treatments. In order to improve this situation, it is necessary to fully understand how C. albicans is able to cause disease and how it copes with the host defenses. Our previous studies have revealed the importance of the C. albicans gene MBF1 in virulence and ability to colonize internal organs of mammalian and insect hosts. MBF1 encodes a putative transcriptional regulator, and as such it likely has an impact in the regulation of C. albicans gene expression during host infection. Here, recent advances in RNA-seq technologies were used to obtain a detailed analysis of the impact of MBF1 on C. albicans gene expression both in vitro and during infection. MBF1 was involved in the regulation of several genes with a role in glycolysis and response to stress, particularly to nutritional stress. We also investigated whether an interaction existed between MBF1 and GCN4, a master regulator of response to starvation, and found that both genes were needed for resistance to amino acid starvation, suggesting some level of interaction between the two. Reinforcing this idea, we showed that the proteins encoded by both genes could interact. Consistent with the role of MBF1 in virulence, we also established that GCN4 was necessary for virulence in the mouse model of systemic infection as well as in the Galleria mellonella infection model.
Collapse
Affiliation(s)
- Sara Amorim-Vaz
- Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Alix T. Coste
- Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Van Du T. Tran
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Marco Pagni
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Dominique Sanglard
- Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
33
|
Chew SY, Brown AJP, Lau BYC, Cheah YK, Ho KL, Sandai D, Yahaya H, Than LTL. Transcriptomic and proteomic profiling revealed reprogramming of carbon metabolism in acetate-grown human pathogen Candida glabrata. J Biomed Sci 2021; 28:1. [PMID: 33388061 PMCID: PMC7778802 DOI: 10.1186/s12929-020-00700-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 12/21/2020] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Emergence of Candida glabrata, which causes potential life-threatening invasive candidiasis, has been widely associated with high morbidity and mortality. In order to cause disease in vivo, a robust and highly efficient metabolic adaptation is crucial for the survival of this fungal pathogen in human host. In fact, reprogramming of the carbon metabolism is believed to be indispensable for phagocytosed C. glabrata within glucose deprivation condition during infection. METHODS In this study, the metabolic responses of C. glabrata under acetate growth condition was explored using high-throughput transcriptomic and proteomic approaches. RESULTS Collectively, a total of 1482 transcripts (26.96%) and 242 proteins (24.69%) were significantly up- or down-regulated. Both transcriptome and proteome data revealed that the regulation of alternative carbon metabolism in C. glabrata resembled other fungal pathogens such as Candida albicans and Cryptococcus neoformans, with up-regulation of many proteins and transcripts from the glyoxylate cycle and gluconeogenesis, namely isocitrate lyase (ICL1), malate synthase (MLS1), phosphoenolpyruvate carboxykinase (PCK1) and fructose 1,6-biphosphatase (FBP1). In the absence of glucose, C. glabrata shifted its metabolism from glucose catabolism to anabolism of glucose intermediates from the available carbon source. This observation essentially suggests that the glyoxylate cycle and gluconeogenesis are potentially critical for the survival of phagocytosed C. glabrata within the glucose-deficient macrophages. CONCLUSION Here, we presented the first global metabolic responses of C. glabrata to alternative carbon source using transcriptomic and proteomic approaches. These findings implicated that reprogramming of the alternative carbon metabolism during glucose deprivation could enhance the survival and persistence of C. glabrata within the host.
Collapse
Affiliation(s)
- Shu Yih Chew
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Alistair J. P. Brown
- MRC Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD UK
| | - Benjamin Yii Chung Lau
- Proteomics and Metabolomics (PROMET) Group, Malaysian Palm Oil Board, Bandar Baru Bangi, 43000 Kajang, Selangor Malaysia
| | - Yoke Kqueen Cheah
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Kok Lian Ho
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Doblin Sandai
- Infectomics Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Kepala Batas, Pulau Pinang Malaysia
| | - Hassan Yahaya
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
- Department of Medical Laboratory Science, Faculty of Allied Health Sciences, Bayero University, Kano, Nigeria
| | - Leslie Thian Lung Than
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| |
Collapse
|
34
|
Kwai BXC, Collins AJ, Middleditch MJ, Sperry J, Bashiri G, Leung IKH. Itaconate is a covalent inhibitor of the Mycobacterium tuberculosis isocitrate lyase. RSC Med Chem 2021; 12:57-61. [PMID: 34046597 PMCID: PMC8130629 DOI: 10.1039/d0md00301h] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/07/2020] [Indexed: 12/20/2022] Open
Abstract
Itaconate is a mammalian antimicrobial metabolite that inhibits the isocitrate lyases (ICLs) of Mycobacterium tuberculosis. Herein, we report that ICLs form a covalent adduct with itaconate through their catalytic cysteine residue. These results reveal atomic details of itaconate inhibition and provide insights into the catalytic mechanism of ICLs.
Collapse
Affiliation(s)
- Brooke X C Kwai
- School of Chemical Sciences, The University of Auckland Private Bag 92019, Victoria Street West Auckland 1142 New Zealand
| | - Annabelle J Collins
- School of Chemical Sciences, The University of Auckland Private Bag 92019, Victoria Street West Auckland 1142 New Zealand
| | - Martin J Middleditch
- School of Biological Sciences, The University of Auckland Private Bag 92019, Victoria Street West Auckland 1142 New Zealand
- Auckland Science Analytical Services, The University of Auckland Private Bag 92019, Victoria Street West Auckland 1142 New Zealand
| | - Jonathan Sperry
- School of Chemical Sciences, The University of Auckland Private Bag 92019, Victoria Street West Auckland 1142 New Zealand
| | - Ghader Bashiri
- School of Biological Sciences, The University of Auckland Private Bag 92019, Victoria Street West Auckland 1142 New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland Private Bag 92019, Victoria Street West Auckland 1142 New Zealand
| | - Ivanhoe K H Leung
- School of Chemical Sciences, The University of Auckland Private Bag 92019, Victoria Street West Auckland 1142 New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland Private Bag 92019, Victoria Street West Auckland 1142 New Zealand
| |
Collapse
|
35
|
Interaction of Isocitrate Lyase with Proteins Involved in the Energetic Metabolism in Paracoccidioides lutzii. J Fungi (Basel) 2020; 6:jof6040309. [PMID: 33238437 PMCID: PMC7712234 DOI: 10.3390/jof6040309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/06/2020] [Accepted: 11/20/2020] [Indexed: 11/18/2022] Open
Abstract
Background: Systemic mycosis is a cause of death of immunocompromised subjects. The treatment directed to evade fungal pathogens shows severe limitations, such as time of drug exposure and side effects. The paracoccidioidomycosis (PCM) treatment depends on the severity of the infection and may last from months to years. Methods: To analyze the main interactions of Paracoccidioides lutzii isocitrate lyase (ICL) regarding the energetic metabolism through affinity chromatography, we performed blue native PAGE and co-immunoprecipitation to identify ICL interactions. We also performed in silico analysis by homology, docking, hot-spot prediction and contact preference analysis to identify the conformation of ICL complexes. Results: ICL interacted with 18 proteins in mycelium, 19 in mycelium-to-yeast transition, and 70 in yeast cells. Thirty complexes were predicted through docking and contact preference analysis. ICL has seven main regions of interaction with protein partners. Conclusions: ICL seems to interfere with energetic metabolism of P. lutzii, regulating aerobic and anaerobic metabolism as it interacts with proteins from glycolysis, gluconeogenesis, TCA and methylcitrate cycles, mainly through seven hot-spot residues.
Collapse
|
36
|
Feng J, He L, Xiao X, Chen Z, Chen C, Chu J, Lu S, Li X, Mylonakis E, Xi L. Methylcitrate cycle gene MCD is essential for the virulence of Talaromyces marneffei. Med Mycol 2020; 58:351-361. [PMID: 31290549 DOI: 10.1093/mmy/myz063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/23/2019] [Accepted: 07/03/2019] [Indexed: 01/11/2023] Open
Abstract
Talaromyces marneffei (T. marneffei), which used to be known as Penicillium marneffei, is the causative agent of the fatal systemic mycosis known as talaromycosis. For the purpose of understanding the role of methylcitrate cycle in the virulence of T. marneffei, we generated MCD deletion (ΔMCD) and complementation (ΔMCD+) mutants of T. marneffei. Growth in different carbon sources showed that ΔMCD cannot grow on propionate media and grew slowly on the valerate, valine, methionine, isoleucine, cholesterol, and YNB (carbon free) media. The macrophage killing assay showed that ΔMCD was attenuated in macrophages of mice in vitro, especially at the presence of propionate. Finally, virulence studies in a murine infection experiment revealed attenuated virulence of the ΔMCD, which indicates MCD is essential for T. marneffei virulence in the host. This experiment laid the foundation for the further study of the specific mechanisms underlying the methylcitrate cycle of T. marneffei and may provide suitable targets for new antifungals.
Collapse
Affiliation(s)
- Jiao Feng
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Liya He
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xing Xiao
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiwen Chen
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chunmei Chen
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jieming Chu
- Johns Hopkins University Bloomberg School of Public Health, Wolfe Street, Baltimore, MD, USA
| | - Sha Lu
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiqing Li
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Liyan Xi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Dermatology Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
37
|
Survival Strategies of Pathogenic Candida Species in Human Blood Show Independent and Specific Adaptations. mBio 2020; 11:mBio.02435-20. [PMID: 33024045 PMCID: PMC7542370 DOI: 10.1128/mbio.02435-20] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To ensure their survival, pathogens have to adapt immediately to new environments in their hosts, for example, during the transition from the gut to the bloodstream. Here, we investigated the basis of this adaptation in a group of fungal species which are among the most common causes of hospital-acquired infections, the Candida species. On the basis of a human whole-blood infection model, we studied which genes and processes are active over the course of an infection in both the host and four different Candida pathogens. Remarkably, we found that, while the human host response during the early phase of infection is predominantly uniform, the pathogens pursue largely individual strategies and each one regulates genes involved in largely disparate processes in the blood. Our results reveal that C. albicans, C. glabrata, C. parapsilosis, and C. tropicalis all have developed individual strategies for survival in the host. This indicates that their pathogenicity in humans has evolved several times independently and that genes which are central for survival in the host for one species may be irrelevant in another. Only four species, Candida albicans, C. glabrata, C. parapsilosis, and C. tropicalis, together account for about 90% of all Candida bloodstream infections and are among the most common causes of invasive fungal infections of humans. However, virulence potential varies among these species, and the phylogenetic tree reveals that their pathogenicity may have emerged several times independently during evolution. We therefore tested these four species in a human whole-blood infection model to determine, via comprehensive dual-species RNA-sequencing analyses, which fungal infection strategies are conserved and which are recent evolutionary developments. The ex vivo infection progressed from initial immune cell interactions to nearly complete killing of all fungal cells. During the course of infection, we characterized important parameters of pathogen-host interactions, such as fungal survival, types of interacting immune cells, and cytokine release. On the transcriptional level, we obtained a predominantly uniform and species-independent human response governed by a strong upregulation of proinflammatory processes, which was downregulated at later time points after most of the fungal cells were killed. In stark contrast, we observed that the different fungal species pursued predominantly individual strategies and showed significantly different global transcriptome patterns. Among other findings, our functional analyses revealed that the fungal species relied on different metabolic pathways and virulence factors to survive the host-imposed stress. These data show that adaptation of Candida species as a response to the host is not a phylogenetic trait, but rather has likely evolved independently as a prerequisite to cause human infections.
Collapse
|
38
|
Laurian R, Jacot-des-Combes C, Bastian F, Dementhon K, Cotton P. Carbon metabolism snapshot by ddPCR during the early step of Candida albicans phagocytosis by macrophages. Pathog Dis 2020; 78:5780227. [PMID: 32129841 DOI: 10.1093/femspd/ftaa014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/03/2020] [Indexed: 01/08/2023] Open
Abstract
During Candida macrophage interactions, phagocytosed yeast cells feed in order to grow, develop hyphae and escape. Through numerous proteomic and transcriptomic studies, two metabolic phases have been described. A shift to a starvation mode is generally identified as early as one-hour post phagocytosis, followed by a glycolytic growth mode after C. albicans escaped from the macrophage. Healthy macrophages contain low amounts of glucose. To determine if this carbon source was sensed and metabolized by the pathogen, we explored the transcription level of a delimited set of key genes expressed in C. albicans cells during phagocytosis by macrophages, at an early stage of the interaction. This analysis was performed using a technical digital droplet PCR approach to quantify reliably the expression of carbon metabolic genes after 30 min of phagocytosis. Our data confirm the technique of digital droplet PCR for the detection of C. albicans transcripts using cells recovered after a short period of phagocytosis. At this stage, carbon metabolism is clearly oriented towards the use of alternative sources. However, the activation of high-affinity glucose transport system suggests that the low amount of glucose initially present in the macrophages is detected by the pathogen.
Collapse
Affiliation(s)
- Romain Laurian
- Génétique Moléculaire des Levures, UMR-CNRS 5240 Microbiologie Adaptation et Pathogénie, Université de Lyon-Université Lyon1, Lyon, France
| | - Cécile Jacot-des-Combes
- DTAMB, FR 3728 Bio-Environnement et Santé, Université de Lyon-Université Lyon1, Lyon, France
| | - Fabiola Bastian
- DTAMB, FR 3728 Bio-Environnement et Santé, Université de Lyon-Université Lyon1, Lyon, France
| | - Karine Dementhon
- Laboratoire de Microbiologie Fondamentale et Pathogénicité, UMR-CNRS 5234, Université de Bordeaux, Bordeaux, France
| | - Pascale Cotton
- Génétique Moléculaire des Levures, UMR-CNRS 5240 Microbiologie Adaptation et Pathogénie, Université de Lyon-Université Lyon1, Lyon, France
| |
Collapse
|
39
|
Hackett EE, Sheedy FJ. An Army Marches on Its Stomach: Metabolic Intermediates as Antimicrobial Mediators in Mycobacterium tuberculosis Infection. Front Cell Infect Microbiol 2020; 10:446. [PMID: 32984072 PMCID: PMC7477320 DOI: 10.3389/fcimb.2020.00446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/21/2020] [Indexed: 12/13/2022] Open
Abstract
The cells of the immune system are reliant on their metabolic state to launch effective responses to combat mycobacterial infections. The bioenergetic profile of the cell determines the molecular fuels and metabolites available to the host, as well as to the bacterial invader. How cells utilize the nutrients in their microenvironment—including glucose, lipids and amino acids—to sustain their functions and produce antimicrobial metabolites, and how mycobacteria exploit this to evade the immune system is of great interest. Changes in flux through metabolic pathways alters the intermediate metabolites present. These intermediates are beginning to be recognized as key modulators of immune signaling as well as direct antimicrobial effectors, and their impact on tuberculosis infection is becoming apparent. A better understanding of how metabolism impacts immunity to Mycobacterium tuberculosis and how it is regulated and thus can be manipulated will open the potential for novel therapeutic interventions and vaccination strategies.
Collapse
Affiliation(s)
- Emer E Hackett
- Macrophage Homeostasis, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Frederick J Sheedy
- Macrophage Homeostasis, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
40
|
Zhao X, Duan X, Dai Y, Zhen J, Guo J, Zhang K, Wang X, Kuang Z, Wang H, Niu J, Fan L, Xie J. Mycobacterium Von Willebrand factor protein MSMEG_3641 is involved in biofilm formation and intracellular survival. Future Microbiol 2020; 15:1033-1044. [PMID: 32811177 DOI: 10.2217/fmb-2020-0064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: Mycobacterium tuberculosis in vitro biofilm is associated with the virulence and persistence capability. Our aim is to delineate factors involved in biofilms development. Materials & methods: We performed transposon mutants screen and found that mutation of MSMEG_3641, a homolog of M. tuberculosis Rv1836c, can change M. smegmatis colony morphology and biofilm. Results: MSMEG_3641 contains a vWA domain that is highly conserved among Mycobacteria. The phenotypes of MSMEG_3641 mutants include disrupted biofilm, weakened migration ability and changed colony morphology. All phenotypes might be contributed to the enhanced cell wall permeability and declined cell aggregation ability. Conclusion: To our knowledge, this is the first report concerning the mycobacteria Von Willebrand factor domain function, especially in colony morphology and biofilm development.
Collapse
Affiliation(s)
- Xiaokang Zhao
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment & Bio-Resource of The Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xiangke Duan
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment & Bio-Resource of The Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Yongdong Dai
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment & Bio-Resource of The Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Junfeng Zhen
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment & Bio-Resource of The Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Jiaohan Guo
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment & Bio-Resource of The Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Ke Zhang
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment & Bio-Resource of The Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xiaoyu Wang
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment & Bio-Resource of The Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Zhongmei Kuang
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment & Bio-Resource of The Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Hao Wang
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment & Bio-Resource of The Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Jingjing Niu
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment & Bio-Resource of The Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Lin Fan
- Shanghai Clinic & Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai Key Laboratory of Tuberculosis, Shanghai 200433, China
| | - Jianping Xie
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment & Bio-Resource of The Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
41
|
Joshi T, Joshi T, Sharma P, Pundir H, Chandra S. In silico identification of natural fungicide from Melia azedarach against isocitrate lyase of Fusarium graminearum. J Biomol Struct Dyn 2020; 39:4816-4834. [PMID: 32568603 DOI: 10.1080/07391102.2020.1780941] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Isocitrate Lyase (ICL) is a crucial enzyme involved in the Glyoxylate pathway, essential for the virulence of several fungal pathogens including Fusarium graminearum. ICL is a novel target for the discovery of antifungal compounds and F. graminearum ICL inhibitors can be used to control the growth of this fungus. Although, several inhibitors of ICL have been identified, however, most of these inhibitors are not environment-friendly. Hence there is still a need to discover natural inhibitors of ICL that can be more effective. To identify a potential antifungal compound, we performed a structure-based screening of phytochemicals of Melia azedarach against the FgICL structure by molecular docking and 104 ligands were found to have a better docking score as compared to the reference molecule. These compounds were assessed for drug-likeness and ADMET prediction. After molecular docking, drug-likeness and toxicity analysis, six potential compounds (Melianoninol (-6.6 kcal/mol), Nimbinene (-7.7 kcal/mol), Vilasinin (-8.1 kcal/mol), Fraxinellone (-6.7 kcal/mol), Gedunin (-7.8 kcal/mol), and Meldenin (-7.8 kcal/mol)) were subjected for rescoring by X-Score. The structural stability and dynamics of screened compounds at the active site of FgICL were examined using MD simulation and MM-PBSA analysis. The result of MM-PBSA revealed that four phytochemicals viz. Melianoninol, Nimbinene, Vilasinin, and Fraxinellone had binding free energy of -17.25 kcal/mol, -59.35 kcal/mol, -64.79 kcal/mol, and -29.86 kcal/mol, respectively. Molecular dynamics simulation and MM-PBSA demonstrated that these four phytochemicals displayed considerable significant structural and pharmacological properties and could be probable antifungal drug candidates against F. graminearum. These phyotchemicals of M. azedarach may be suitable candidates for further experimental analysis. [Formula: see text]Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Tanuja Joshi
- Department of Botany, Kumaun University, Almora, Uttarakhand, India
| | - Tushar Joshi
- Department of Botany, Kumaun University, Almora, Uttarakhand, India.,Department of Biotechnology, Bhimtal Campus, Kumaun University, Nainital, Uttarakhand, India
| | - Priyanka Sharma
- Department of Botany, Kumaun University, Nainital, Uttarakhand, India
| | - Hemlata Pundir
- Department of Botany, Kumaun University, Nainital, Uttarakhand, India
| | - Subhash Chandra
- Department of Botany, Kumaun University, Almora, Uttarakhand, India
| |
Collapse
|
42
|
Wijnants S, Riedelberger M, Penninger P, Kuchler K, Van Dijck P. Sugar Phosphorylation Controls Carbon Source Utilization and Virulence of Candida albicans. Front Microbiol 2020; 11:1274. [PMID: 32612591 PMCID: PMC7308821 DOI: 10.3389/fmicb.2020.01274] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/19/2020] [Indexed: 01/03/2023] Open
Abstract
Candida albicans is an opportunistic human fungal pathogen that relies upon different virulence traits, including morphogenesis, invasion, biofilm formation, and nutrient acquisition from host sources as well as metabolic adaptations during host invasion. In this study, we show how sugar kinases at the start of glycolysis modulate virulence of C. albicans. Sequence comparison with Saccharomyces cerevisiae identified four enzymes (Hxk1, Hxk2, Glk1, and Glk4) in C. albicans with putative roles in sugar phosphorylation. Hxk2, Glk1, and Glk4 demonstrate a critical role in glucose metabolism, while Hxk2 is the only kinase important for fructose metabolism. Additionally, we show that Hxk1 controls HXK2, GLK1, and GLK4 expression in the presence of fermentable as well as non-fermentable carbon sources, thereby indirectly controlling glycolysis. Moreover, these sugar kinases are important during virulence. Disabling the glycolytic pathway reduces adhesion capacity, while deletion of HXK1 decreases biofilm formation. Finally, we demonstrate that hxk2Δ/Δ glk1Δ/Δ glk4Δ/Δ and hxk1Δ/Δ hxk2Δ/Δ glk1Δ/Δ glk4Δ/Δ have attenuated virulence upon systemic infections in mice. These results indicate a regulatory role for Hxk1 during sugar phosphorylation. Furthermore, these kinases are essential during growth on glucose or fructose, and C. albicans relies on a functional glycolytic pathway for maximal virulence.
Collapse
Affiliation(s)
- Stefanie Wijnants
- Laboratory of Molecular Cell Biology, Department of Biology, Institute of Botany and Microbiology, KU Leuven, Leuven, Belgium
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
| | - Michael Riedelberger
- Max Perutz Labs Vienna, Center for Medical Biochemistry, Medical University of Vienna, Vienna, Austria
| | - Philipp Penninger
- Max Perutz Labs Vienna, Center for Medical Biochemistry, Medical University of Vienna, Vienna, Austria
| | - Karl Kuchler
- Max Perutz Labs Vienna, Center for Medical Biochemistry, Medical University of Vienna, Vienna, Austria
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, Institute of Botany and Microbiology, KU Leuven, Leuven, Belgium
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
| |
Collapse
|
43
|
Tripathi A, Liverani E, Tsygankov AY, Puri S. Iron alters the cell wall composition and intracellular lactate to affect Candida albicans susceptibility to antifungals and host immune response. J Biol Chem 2020; 295:10032-10044. [PMID: 32503842 DOI: 10.1074/jbc.ra120.013413] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/27/2020] [Indexed: 12/19/2022] Open
Abstract
Fungal pathogen Candida albicans has a complex cell wall consisting of an outer layer of mannans and an inner layer of β-glucans and chitin. The fungal cell wall is the primary target for antifungals and is recognized by host immune cells. Environmental conditions such as carbon sources, pH, temperature, and oxygen tension can modulate the fungal cell wall architecture. Cellular signaling pathways, including the mitogen-activated protein kinase (MAPK) pathways, are responsible for sensing environmental cues and mediating cell wall alterations. Although iron has recently been shown to affect β-1,3-glucan exposure on the cell wall, we report here that iron changes the composition of all major C. albicans cell wall components. Specifically, high iron decreased the levels of mannans (including phosphomannans) and chitin; and increased β-1,3-glucan levels. These changes increased the resistance of C. albicans to cell wall-perturbing antifungals. Moreover, high iron cells exhibited adequate mitochondrial functioning; leading to a reduction in accumulation of lactate that signals through the transcription factor Crz1 to induce β-1,3-glucan masking in C. albicans We show here that iron-induced changes in β-1,3-glucan exposure are lactate-dependent; and high iron causes β-1,3-glucan exposure by preventing lactate-induced, Crz1-mediated inhibition of activation of the fungal MAPK Cek1. Furthermore, despite exhibiting enhanced antifungal resistance, high iron C. albicans cells had reduced survival upon phagocytosis by macrophages. Our results underscore the role of iron as an environmental signal in multiple signaling pathways that alter cell wall architecture in C. albicans, thereby affecting its survival upon exposure to antifungals and host immune response.
Collapse
Affiliation(s)
- Aparna Tripathi
- Oral Microbiome Research Laboratory, Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania, USA
| | - Elisabetta Liverani
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Temple University Hospital, Philadelphia, Pennsylvania, USA
| | - Alexander Y Tsygankov
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Temple University Hospital, Philadelphia, Pennsylvania, USA.,Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Sumant Puri
- Oral Microbiome Research Laboratory, Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
44
|
Exposure of Mycobacterium abscessus to Environmental Stress and Clinically Used Antibiotics Reveals Common Proteome Response among Pathogenic Mycobacteria. Microorganisms 2020; 8:microorganisms8050698. [PMID: 32397563 PMCID: PMC7285101 DOI: 10.3390/microorganisms8050698] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/19/2022] Open
Abstract
Mycobacterium abscessus subsp. abscessus (MAB) is a clinically important nontuberculous mycobacterium (NTM) causing pulmonary infection in patients such as cystic fibrosis and bronchiectasis. MAB is naturally resistant to the majority of available antibiotics. In attempts to identify the fundamental response of MAB to aerobic, anaerobic, and biofilm conditions (as it is encountered in patients) and during exposure to antibiotics, we studied bacterial proteome using tandem mass tag mass spectrometry sequencing. Numerous de novo synthesized proteins belonging to diverse metabolic pathways were found in anaerobic and biofilm conditions, including glycolysis/gluconeogenesis, tricarboxylic acid (TCA) cycle, oxidative phosphorylation, nitrogen metabolism, and glyoxylate and dicarboxylate metabolism. Upon exposure to amikacin and linezolid under stress environments, MAB displayed metabolic enrichment for glycerophospholipid metabolism and oxidative phosphorylation. By comparing proteomes of two significant NTMs, MAB and M. avium subsp. hominissuis, we found highly synthesized shared enzymes of oxidative phosphorylation, TCA cycle, glycolysis/gluconeogenesis, glyoxylate/dicarboxylate, nitrogen metabolism, peptidoglycan biosynthesis, and glycerophospholipid/glycerolipid metabolism. The activation of peptidoglycan and fatty acid biosynthesis pathways indicates the attempt of bacteria to modify the cell wall, influencing the susceptibility to antibiotics. This study establishes global changes in the synthesis of enzymes promoting the metabolic shift and enhancing the pathogen resistance to antibiotics within different environments.
Collapse
|
45
|
2-Aminopyridine Analogs Inhibit Both Enzymes of the Glyoxylate Shunt in Pseudomonas aeruginosa. Int J Mol Sci 2020; 21:ijms21072490. [PMID: 32260167 PMCID: PMC7177833 DOI: 10.3390/ijms21072490] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 12/16/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen responsible for many hospital-acquired infections. P. aeruginosa can thrive in diverse infection scenarios by rewiring its central metabolism. An example of this is the production of biomass from C2 nutrient sources such as acetate via the glyoxylate shunt when glucose is not available. The glyoxylate shunt is comprised of two enzymes, isocitrate lyase (ICL) and malate synthase G (MS), and flux through the shunt is essential for the survival of the organism in mammalian systems. In this study, we characterized the mode of action and cytotoxicity of structural analogs of 2-aminopyridines, which have been identified by earlier work as being inhibitory to both shunt enzymes. Two of these analogs were able to inhibit ICL and MS in vitro and prevented growth of P. aeruginosa on acetate (indicating cell permeability). Moreover, the compounds exerted negligible cytotoxicity against three human cell lines and showed promising in vitro drug metabolism and safety profiles. Isothermal titration calorimetry was used to confirm binding of one of the analogs to ICL and MS, and the mode of enzyme inhibition was determined. Our data suggest that these 2-aminopyridine analogs have potential as anti-pseudomonal agents.
Collapse
|
46
|
Venkata S, Zeeshan F, Kamal A, Luqman AK, Saif H. Efficiency of vanillin in impeding metabolic adaptability and virulence of Candida albicans by inhibiting glyoxylate cycle, morphogenesis, and biofilm formation. Curr Med Mycol 2020; 6:1-8. [PMID: 32420501 PMCID: PMC7217250 DOI: 10.18502/cmm.6.1.2501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Background and Purpose Candida albicans is the fourth most common cause of nosocomial fungal infections across the world. The current drug regimens are suffering from such drawbacks as drug resistance, toxicity, and costliness; accordingly, they highlight the need for the discovery of novel drug agents. The metabolic adaptability under low-carbon conditions and expression of functional virulence traits mark the success of pathogens to cause infection. The metabolic pathways, such as glyoxylate cycle (GC), enable C. albicans to survive under glucose-deficient conditions prevalent in the hostile niche. Therefore, the key enzymes, namely isocitrate lyase (ICL) and malate synthase (MLS), represent attractive agents against C. albicans. Similarly, virulence traits, such as morphogenesis and biofilm formation, are the crucial determinants of C. albicans pathogenicity. Regarding this, the present study was conducted to uncover the role of vanillin (Van), a natural food flavoring agent, in inhibiting GC, yeast-to-hyphal transition, and biofilm formation in human fungal pathogen C. albicans. Materials and Methods For the determination of hypersensitivity under low-glucose conditions, phenotypic susceptibility assay was utilized. In addition, enzyme activities were estimated based on crude extracts while in-silico binding was confirmed by molecular docking. The assessment of morphogenesis was accomplished using hyphal-inducing media, and biofilm formation was estimated using calcofluor staining, MTT assay, and biomass measurement. Additionally, the in vivo efficacy of Van was demonstrated using Caenorhabditis elegans nematode model. Results Based on the results, Van was found to be a potent GC inhibitor that phenocopied ICL1 deletion mutant and displayed hypersensitivity under low-carbon conditions. Accordingly, Van facilitated the inhibition of ICL and MLS activities in vitro. Molecular docking analyses revealed the in-silico binding affinity of Van with Icl1p and Mls1p. Those analyses were also confirmative of the binding of Van to the active sites of both proteins with better binding energy in comparison to their known inhibitors. Furthermore, Van led to the attenuation of such virulence traits as morphogenesis, biofilm formation, and cell adherence. Finally, the antifungal efficacy of Van was demonstrated by the enhanced survival of C. elegans with Candida infection. The results also confirmed negligible hemolytic activity on erythrocytes. Conclusion As the findings of the present study indicated, Van is a persuasive natural compound that warrants further attention to exploit its anticandidal potential.
Collapse
Affiliation(s)
- Saibabu Venkata
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram, India.,Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Fatima Zeeshan
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram, India
| | - Ahmad Kamal
- Department of Pharmaceutical Chemistry, Jamia Hamdard, New Delhi, India
| | | | - Hameed Saif
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram, India
| |
Collapse
|
47
|
Abstract
Candida albicans has remained the main etiological agent of candidiasis, challenges clinicians with high mortality and morbidity. The emergence of resistance to antifungal drugs, toxicity and lower efficacy have all contributed to an urgent need to develop alternative drugs aiming at novel targets in C. albicans. Targeting the production of virulence factors, which are essential processes for infectious agents, represents an attractive substitute for the development of newer anti-infectives. The present review highlights the recent developments made in the understanding of the pathogenicity of C. albicans. Production of hydrolytic enzymes, morphogenesis and biofilm formation, along with their molecular and metabolic regulation in Candida are discussed with regard to the development of novel antipathogenic drugs against candidiasis. Over the last decade, candidiasis has remained a major problematic disease worldwide. In spite of the existence of many antifungal drugs, the treatment of such diseases has still remained unsuccessful due to drug inefficacy. Therefore, there is a need to discover antifungals with different modes of action, such as antipathogenic drugs against Candida albicans. Here, we describe how various types of virulence factors such as proteinase, phospholipase, hemolysin, adhesion, morphogenesis and biofilm formation, could be targeted to develop novel therapeutics. We can inhibit production of these virulence factors by controlling their molecular/metabolic regulation.
Collapse
|
48
|
Duval C, Macabiou C, Garcia C, Lesuisse E, Camadro J, Auchère F. The adaptive response to iron involves changes in energetic strategies in the pathogen Candida albicans. Microbiologyopen 2020; 9:e970. [PMID: 31788966 PMCID: PMC7002100 DOI: 10.1002/mbo3.970] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/22/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
Candida albicans is an opportunist pathogen responsible for a large spectrum of infections, from superficial mycosis to systemic diseases known as candidiasis. Its ability to grow in different morphological forms, such as yeasts or filamentous hyphae, contributes to its survival in diverse microenvironments. Iron uptake has been associated with virulence, and C. albicans has developed elaborate strategies for acquiring iron from its host. In this work, we analyze the metabolic changes in response to changes in iron content in the growth medium and compare C. albicans adaptation to the presence or absence of iron. Functional and morphological studies, correlated to a quantitative proteomic analysis, were performed to assess the specific pathways underlying the response to iron, both in the yeast and filamentous forms. Overall, the results show that the adaptive response to iron is associated with a metabolic remodeling affecting the energetic pathways of the pathogen. This includes changes in the thiol-dependent redox status, the activity of key mitochondrial enzymes and the respiratory chain. Iron deficiency stimulates bioenergetic pathways, whereas iron-rich condition is associated with greater biosynthetic needs, particularly in filamentous forms. Moreover, we found that C. albicans yeast cells have an extraordinary capability to adapt to changes in environmental conditions.
Collapse
Affiliation(s)
- Celia Duval
- Laboratoire MitochondriesMétaux et Stress OxydantInstitut Jacques MonodUMR 7592Université Paris‐Diderot/CNRS (USPC)ParisFrance
| | - Carole Macabiou
- Laboratoire MitochondriesMétaux et Stress OxydantInstitut Jacques MonodUMR 7592Université Paris‐Diderot/CNRS (USPC)ParisFrance
| | - Camille Garcia
- Plateforme Protéomique structurale et fonctionnelle/Spectrométrie de masseInstitut Jacques MonodUMR 7592Université Paris‐Diderot/CNRS (USPC)ParisFrance
| | - Emmanuel Lesuisse
- Laboratoire MitochondriesMétaux et Stress OxydantInstitut Jacques MonodUMR 7592Université Paris‐Diderot/CNRS (USPC)ParisFrance
| | - Jean‐Michel Camadro
- Laboratoire MitochondriesMétaux et Stress OxydantInstitut Jacques MonodUMR 7592Université Paris‐Diderot/CNRS (USPC)ParisFrance
| | - Françoise Auchère
- Laboratoire MitochondriesMétaux et Stress OxydantInstitut Jacques MonodUMR 7592Université Paris‐Diderot/CNRS (USPC)ParisFrance
| |
Collapse
|
49
|
Burgain A, Pic É, Markey L, Tebbji F, Kumamoto CA, Sellam A. A novel genetic circuitry governing hypoxic metabolic flexibility, commensalism and virulence in the fungal pathogen Candida albicans. PLoS Pathog 2019; 15:e1007823. [PMID: 31809527 PMCID: PMC6919631 DOI: 10.1371/journal.ppat.1007823] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 12/18/2019] [Accepted: 10/18/2019] [Indexed: 01/04/2023] Open
Abstract
Inside the human host, the pathogenic yeast Candida albicans colonizes predominantly oxygen-poor niches such as the gastrointestinal and vaginal tracts, but also oxygen-rich environments such as cutaneous epithelial cells and oral mucosa. This suppleness requires an effective mechanism to reversibly reprogram the primary metabolism in response to oxygen variation. Here, we have uncovered that Snf5, a subunit of SWI/SNF chromatin remodeling complex, is a major transcriptional regulator that links oxygen status to the metabolic capacity of C. albicans. Snf5 and other subunits of SWI/SNF complex were required to activate genes of carbon utilization and other carbohydrates related process specifically under hypoxia. snf5 mutant exhibited an altered metabolome reflecting that SWI/SNF plays an essential role in maintaining metabolic homeostasis and carbon flux in C. albicans under hypoxia. Snf5 was necessary to activate the transcriptional program linked to both commensal and invasive growth. Accordingly, snf5 was unable to maintain its growth in the stomach, the cecum and the colon of mice. snf5 was also avirulent as it was unable to invade Galleria larvae or to cause damage to human enterocytes and murine macrophages. Among candidates of signaling pathways in which Snf5 might operate, phenotypic analysis revealed that mutants of Ras1-cAMP-PKA pathway, as well as mutants of Yak1 and Yck2 kinases exhibited a similar carbon flexibility phenotype as did snf5 under hypoxia. Genetic interaction analysis indicated that the adenylate cyclase Cyr1, a key component of the Ras1-cAMP pathway interacted genetically with Snf5. Our study yielded new insight into the oxygen-sensitive regulatory circuit that control metabolic flexibility, stress, commensalism and virulence in C. albicans. A critical aspect of eukaryotic cell fitness is the ability to sense and adapt to variations in oxygen level in their local environment. Hypoxia leads to a substantial remodeling of cell metabolism and energy homeostasis, and thus, organisms must develop an effective regulatory mechanism to cope with oxygen depletion. Candida albicans is an opportunistic yeast that is the most prevalent human fungal pathogens. This yeast colonizes diverse niches inside the human host with contrasting carbon sources and oxygen concentrations. While hypoxia is the predominant condition that C. albicans encounters inside most of the niches, the impact of this condition on metabolic flexibility, a major determinant of fungal virulence, was completely unexplored. Here, we uncovered that the chromatin remodelling complex SWI/SNF is a master regulator of the circuit that links oxygen status to a broad spectrum of carbon utilization routes. Snf5 was essential for the maintenance of C. albicans as a commensal and also for the expression of its virulence. The oxygen-sensitive regulators identified in this work provide a framework to comprehensively understand the virulence of human fungal pathogens and represent a therapeutic value to fight fungal infections.
Collapse
Affiliation(s)
- Anaïs Burgain
- CHU de Québec Research Center (CHUQ), Université Laval, Quebec City, Quebec, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Émilie Pic
- CHU de Québec Research Center (CHUQ), Université Laval, Quebec City, Quebec, Canada
| | - Laura Markey
- Program in Molecular Microbiology, Tufts University, Boston, Massachusetts, United States of America
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, United States of America
| | - Faiza Tebbji
- CHU de Québec Research Center (CHUQ), Université Laval, Quebec City, Quebec, Canada
| | - Carol A. Kumamoto
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, United States of America
| | - Adnane Sellam
- CHU de Québec Research Center (CHUQ), Université Laval, Quebec City, Quebec, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada
- Big Data Research Centre (BDRC-UL), Université Laval, Faculty of Sciences and Engineering, Quebec City, Quebec, Canada
- * E-mail:
| |
Collapse
|
50
|
Loureiro A, Pais C, Sampaio P. Relevance of Macrophage Extracellular Traps in C. albicans Killing. Front Immunol 2019; 10:2767. [PMID: 31866996 PMCID: PMC6904331 DOI: 10.3389/fimmu.2019.02767] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/12/2019] [Indexed: 12/23/2022] Open
Abstract
Candida albicans causes systemic life-threatening infections, particularly in immunocompromised individuals, such as patients in intensive care units, patients undergoing chemotherapy, and post-surgical and neutropenic patients. The proliferation of invading Candida cells is mainly limited by the action of the human innate immune system, in which phagocytic cells play a fundamental role. This function is, however, limited in neutropenic patients, who rely mainly on the protective immunity mediated by macrophages. Macrophages have been shown to release extracellular DNA fibers, known as macrophage extracellular traps (METs), which can entrap and kill various microbes by a process called ETosis. In this study, we observed that, upon contact with C. albicans, macrophages became active in phagocyting and engulfing yeast cells. ETosis was induced in 6% of macrophages within the first 30 min of contact, and this percentage increased with the multiplicity of infection until a plateau was reached. After 2.5 h incubation, the presence of extracellular macrophage DNA was observed in approximately half of the cells. This study suggests that the formation of METs occurs before pyroptosis (first 6–8 h) and macrophage cell death (up to 24 h), and thus, METs could be included in models describing C. albicans–macrophage interactions. We also observed that macrophage ETosis and phagocytosis can occur simultaneously and that, in the first hours of infection, both processes are similarly important in controlling the proliferation of yeast cells, this being critical in neutropenic patients. Finally, it can also be concluded that, since C. albicans can degrade DNA, the structural component of METs, yeast extracellular DNase activity can be considered as an important virulence factor.
Collapse
Affiliation(s)
- Ana Loureiro
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Braga, Portugal
| | - Célia Pais
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Braga, Portugal
| | - Paula Sampaio
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Braga, Portugal
| |
Collapse
|