1
|
Lima RJV, Zanchi FB, Guimarães RMDS, E Silva ADA. Modeling, virtual screening, and enzymatic docking of trehalose 6-phosphate phosphatase and evaluation of the insecticidal effect of phthalimide, N-(p-tolylsulfonyl) on Aedes aegypti (Diptera: Culicidae). PEST MANAGEMENT SCIENCE 2025. [PMID: 40265736 DOI: 10.1002/ps.8841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Aedes aegypti Linnaeus is a medically important vector because of its role in transmitting several arboviruses. Trehalose-6-phosphate phosphatase (TPP), an enzyme from the trehalose pathway, was the focus of this study, which aimed to model it, perform molecular docking and select potential ligands to evaluate their larvicidal and adulticidal activity on the mosquito. RESULTS Because no TPP structure for A. aegypti was described, the modeling was done by homology, using the TPP from Mycobacterium tuberculosis Zopf as a template, with 31% similarity. Following virtual screening, a search for TPP-like molecules on PubChem resulted in 227 molecules, and phthalimide, N-(p-tolylsulfonyl) (PNT) was selected and tested in vivo. Larvicidal tests were conducted in 24-well plates, and adulticidal tests used sugar baits with several concentrations (5 to 100 ppm) of PNT. In larval tests, mortality ranged from 38% to 52% at 24 h and reached 92% at 100 ppm PNT after 72 h. Larval mortality progressively increased over 96 h, with estimated lethal concentration (LC50 and LC90) values after 48 h of 18 and 198 ppm respectively. In adulticidal tests, despite high bait uptake, acute ingestion of PNT did not cause mortality in adult mosquitoes. CONCLUSION PNT demonstrated larvicidal activity against A. aegypti, suggesting that mosquito TPP could be a target in the search for new-generation insecticides. © 2025 The Author(s). Pest Management Science published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Raquel Jemima Viana Lima
- Departamento de Biologia, Programa de Pós-Graduação em Conservação e uso de Recursos Naturais, Universidade Federal de Rondônia, Porto Velho, Brazil
| | - Fernando Berton Zanchi
- Laboratório de Bioinformática e Química Medicinal, Fundação Oswaldo Cruz, Porto Velho, Brazil
- Programa de Pós-Graduação em Biologia Experimental, Porto Velho, Brazil
| | | | - Alexandre de Almeida E Silva
- Programa de Pós-Graduação em Biologia Experimental, Porto Velho, Brazil
- Laboratório de Bioecologia de Insetos, Universidade Federal de Rondônia, Porto Velho, Brazil
| |
Collapse
|
2
|
Cha S, Jo JH, Lee JK, Park W, Moon M, Park GW, Kim MS, Hahn JS. Enhancing D-lactic acid production from methane through metabolic engineering of Methylomonas sp. DH-1. Microb Cell Fact 2025; 24:70. [PMID: 40128822 PMCID: PMC11934524 DOI: 10.1186/s12934-025-02695-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/13/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Methane is an abundant and low-cost carbon source with great potential for conversion into value-added chemicals. Methanotrophs, microorganisms that utilize methane as their sole carbon and energy source, present a promising platform for biotechnological applications. This study aimed to engineer Methylomonas sp. DH-1 to enhance D-LA production through metabolic pathway optimization during large-scale cultivation. RESULTS In this study, we regulated the expression of D-lactate dehydrogenase (D-LDH) using a Ptac promoter with IPTG induction to mitigate the toxic effects of lactate accumulation. To further optimize carbon flow away from glycogen, the glgA gene was deleted. However, this modification led to growth inhibition, especially during scale-up, likely due to the accumulation of ADP-glucose caused by the rewired carbon flux under carbon-excess conditions. Deleting the glgC gene, which encodes glucose 1-phosphate adenylyltransferase, alleviated this issue. The final optimized strain, JHM805, achieved a D-LA production of 6.17 g/L in a 5-L bioreactor, with a productivity of 0.057 g/L/h, marking a significant improvement in D-LA production from methane. CONCLUSIONS The metabolic engineering strategies employed in this study, including the use of an inducible promoter and alleviation of ADP-glucose accumulation toxicity, successfully enhanced the ability of the strain to produce D-LA from methane. Furthermore, optimizing the bioreactor fermentation process through methane and nitrate supplementation resulted in a significant increase in both the titer and productivity, exceeding previously reported values.
Collapse
Affiliation(s)
- Seungwoo Cha
- Department of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Jae-Hwan Jo
- Bioenergy and Resources Upcycling Research Laboratory, Korea Institute of Energy Research, 152 Gajeong-Ro, Yuseong-Gu, Daejeon, 34129, Republic of Korea
- Interdisciplinary Program for Agriculture and Life Sciences, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Jong Kwan Lee
- Department of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Wooyoung Park
- Department of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Myounghoon Moon
- Gwangju Clean Energy Research Center, Korea Institute of Energy Research, 270 Samso-Ro, Buk-Gu, Gwangju, 61003, Republic of Korea
| | - Gwon Woo Park
- Gwangju Clean Energy Research Center, Korea Institute of Energy Research, 270 Samso-Ro, Buk-Gu, Gwangju, 61003, Republic of Korea
| | - Min-Sik Kim
- Bioenergy and Resources Upcycling Research Laboratory, Korea Institute of Energy Research, 152 Gajeong-Ro, Yuseong-Gu, Daejeon, 34129, Republic of Korea.
| | - Ji-Sook Hahn
- Department of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
3
|
Schubert C, Nguyen BD, Sichert A, Näpflin N, Sintsova A, Feer L, Näf J, Daniel BBJ, Steiger Y, von Mering C, Sauer U, Hardt WD. Monosaccharides drive Salmonella gut colonization in a context-dependent or -independent manner. Nat Commun 2025; 16:1735. [PMID: 39966379 PMCID: PMC11836396 DOI: 10.1038/s41467-025-56890-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
The carbohydrates that fuel gut colonization by S. Typhimurium are not fully known. To investigate this, we designed a quality-controlled mutant pool to probe the metabolic capabilities of this enteric pathogen. Using neutral genetic barcodes, we tested 35 metabolic mutants across five different mouse models with varying microbiome complexities, allowing us to differentiate between context-dependent and context-independent nutrient sources. Results showed that S. Typhimurium uses D-mannose, D-fructose and likely D-glucose as context-independent carbohydrates across all five mouse models. The utilization of D-galactose, N-acetylglucosamine and hexuronates, on the other hand, was context-dependent. Furthermore, we showed that D-fructose is important in strain-to-strain competition between Salmonella serovars. Complementary experiments confirmed that D-glucose, D-fructose, and D-galactose are excellent niches for S. Typhimurium to exploit during colonization. Quantitative measurements revealed sufficient amounts of carbohydrates, such as D-glucose or D-galactose, in the murine cecum to drive S. Typhimurium colonization. Understanding these key substrates and their context-dependent or -independent use by enteric pathogens will inform the future design of probiotics and therapeutics to prevent diarrheal infections such as non-typhoidal salmonellosis.
Collapse
Affiliation(s)
- Christopher Schubert
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| | - Bidong D Nguyen
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Andreas Sichert
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Nicolas Näpflin
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Anna Sintsova
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Lilith Feer
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Jana Näf
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Benjamin B J Daniel
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Yves Steiger
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Christian von Mering
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Uwe Sauer
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
4
|
Schwieters A, Cole AL, Rego E, Gao C, Kebriaei R, Wysocki VH, Gunn JS, Ahmer BMM. MtlD as a therapeutic target for intestinal and systemic bacterial infections. J Bacteriol 2025; 207:e0048024. [PMID: 39727397 PMCID: PMC11784389 DOI: 10.1128/jb.00480-24] [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: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
The ability to treat infections is threatened by the rapid emergence of antibiotic resistance among pathogenic microbes. Therefore, new antimicrobials are needed. Here we evaluate mannitol-1-phosphate 5-dehydrogenase (MtlD) as a potential new drug target. In many bacteria, mannitol is transported into the cell and phosphorylated by MtlA, the EIICBA component of a phosphoenolpyruvate-dependent sugar phosphotransferase system. MtlD catalyzes the conversion of mannitol-1-phosphate (Mtl-1P) to fructose-6-phosphate, which enters the glycolytic pathway. Mutants lacking mtlD are sensitive to mannitol due to accumulation of Mtl-1P. Here, we constructed mtlD mutants in four different bacterial species (Cronobacter sakazakii, Pseudomonas aeruginosa, five serovars of Salmonella enterica, and three strains of Escherichia coli), confirming and quantifying their mannitol sensitivity. The quantification of mannitol sensitivity in vitro was complicated by an inoculum effect and a resumption of growth following mannitol intoxication. The rate of resumption at different mannitol concentrations and cell population densities is fairly constant and reveals what is likely an intoxication processing rate. Provision of mannitol in drinking water, or by intraperitoneal injection, dramatically attenuates infection of a Salmonella enterica serovar Typhimurium mtlD mutant in mouse models of both gastroenteritis and systemic infection. Using CC003/Unc mice, we find that a mtlD mutant of Salmonella enterica serovar Typhi is also attenuated by provision of mannitol in drinking water. Therefore, we postulate that MtlD could be a valuable new therapeutic target. IMPORTANCE The ability to treat infections is threatened by the rapid emergence of antibiotic resistance. Mannitol is a polyol used in human medicine and the food industry. During catabolism of mannitol, many bacteria transport mannitol across the inner membrane forming the toxic intermediate mannitol-1-phosphate (Mtl-1P). Mtl-1P must be processed by mannitol dehydrogenase (MtlD) or it accumulates intracellularly, causing growth attenuation. We test and confirm here that mtlD mutants of Escherichia coli (including UPEC, and EHEC), Salmonella (including serovars Typhi, and Paratyphi A, B, and C), Cronobacter, and Pseudomonas experience mannitol sensitivity in vitro. Furthermore, providing mannitol in drinking water can alleviate both gastrointestinal and systemic Salmonella infections in mice. This suggests that inhibition of MtlD could be a viable antimicrobial strategy.
Collapse
Affiliation(s)
- Andrew Schwieters
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
| | - Allysa L. Cole
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
- Center for Microbial Pathogenesis, Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Emily Rego
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
| | - Chengyu Gao
- Campus Chemical Instrument Center, The Ohio State University, Columbus, Ohio, USA
| | - Razieh Kebriaei
- Department of Outcomes and Translational Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Vicki H. Wysocki
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
- National Resource for Native MS-Guided Structural Biology, Columbus, Ohio, USA
| | - John S. Gunn
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
- Center for Microbial Pathogenesis, Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Brian M. M. Ahmer
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
5
|
Bao L, Zhu Z, Ismail A, Zhu B, Anandan V, Whiteley M, Kitten T, Xu P. Experimental evolution of gene essentiality in bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.600122. [PMID: 39071448 PMCID: PMC11275930 DOI: 10.1101/2024.07.16.600122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Essential gene products carry out fundamental cellular activities in interaction with other components. However, the lack of essential gene mutants and appropriate methodologies to link essential gene functions with their partners poses significant challenges. Here, we have generated deletion mutants in 32 genes previously identified as essential, with 23 mutants showing extremely slow growth in the SK36 strain of Streptococcus sanguinis. The 23 genes corresponding to these mutants encode components of diverse pathways, are widely conserved among bacteria, and are essential in many other bacterial species. Whole-genome sequencing of 243 independently evolved populations of these mutants has identified >1000 spontaneous suppressor mutations in experimental evolution. Many of these mutations define new gene and pathway relationships, such as F1Fo-ATPase/V1Vo-ATPase/TrkA1-H1 that were demonstrated across multiple Streptococcus species. Patterns of spontaneous mutations occurring in essential gene mutants differed from those found in wildtype. While gene duplications occurred rarely and appeared most often at later stages of evolution, substitutions, deletions, and insertions were prevalent in evolved populations. These essential gene deletion mutants and spontaneous mutations fixed in the mutant populations during evolution establish a foundation for understanding gene essentiality and the interaction of essential genes in networks.
Collapse
Affiliation(s)
- Liang Bao
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Zan Zhu
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Ahmed Ismail
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Bin Zhu
- Massey Cancer Center, Virginia Commonwealth University, Virginia, USA
| | - Vysakh Anandan
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Marvin Whiteley
- School of Biological Sciences, Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Georgia, USA
| | - Todd Kitten
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Ping Xu
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| |
Collapse
|
6
|
Nguyen BD, Sintsova A, Schubert C, Sichert A, Scheidegger C, Näf J, Huttman J, Lentsch V, Keys T, Rutschmann C, Christen P, Kiefer P, Keller P, Barthel M, Cuenca M, Christen B, Sauer U, Slack E, Vorholt JA, Sunagawa S, Hardt WD. Salmonella Typhimurium screen identifies shifts in mixed-acid fermentation during gut colonization. Cell Host Microbe 2024; 32:1758-1773.e4. [PMID: 39293436 DOI: 10.1016/j.chom.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 07/10/2024] [Accepted: 08/21/2024] [Indexed: 09/20/2024]
Abstract
How enteric pathogens adapt their metabolism to a dynamic gut environment is not yet fully understood. To investigate how Salmonella enterica Typhimurium (S.Tm) colonizes the gut, we conducted an in vivo transposon mutagenesis screen in a gnotobiotic mouse model. Our data implicate mixed-acid fermentation in efficient gut-luminal growth and energy conservation throughout infection. During initial growth, the pathogen utilizes acetate fermentation and fumarate respiration. After the onset of gut inflammation, hexoses appear to become limiting, as indicated by carbohydrate analytics and the increased need for gluconeogenesis. In response, S.Tm adapts by ramping up ethanol fermentation for redox balancing and supplying the TCA cycle with α-ketoglutarate for additional energy. Our findings illustrate how S.Tm flexibly adapts mixed fermentation and its use of the TCA cycle to thrive in the changing gut environment. Similar metabolic wiring in other pathogenic Enterobacteriaceae may suggest a broadly conserved mechanism for gut colonization.
Collapse
Affiliation(s)
- Bidong D Nguyen
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| | - Anna Sintsova
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Christopher Schubert
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Andreas Sichert
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Clio Scheidegger
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Jana Näf
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland; Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Julien Huttman
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Verena Lentsch
- Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Tim Keys
- Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | | | - Philipp Christen
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Patrick Kiefer
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Philipp Keller
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Manja Barthel
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Miguelangel Cuenca
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Beat Christen
- Institute of Microbiology, University of Stuttgart, Stuttgart, Germany
| | - Uwe Sauer
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Emma Slack
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland; Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Julia A Vorholt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Shinichi Sunagawa
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
7
|
Rogers AWL, Radlinski LC, Nguyen H, Tiffany CR, Carvalho TP, Masson HLP, Goodson ML, Bechtold L, Yamazaki K, Liou MJ, Miller BM, Mahan SP, Young BM, Demars AM, Gretler SR, Larabi AB, Lee JY, Bays DJ, Tsolis RM, Bäumler AJ. Salmonella re-engineers the intestinal environment to break colonization resistance in the presence of a compositionally intact microbiota. Cell Host Microbe 2024; 32:1774-1786.e9. [PMID: 39181125 PMCID: PMC11466686 DOI: 10.1016/j.chom.2024.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/29/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024]
Abstract
The gut microbiota prevents harmful microbes from entering the body, a function known as colonization resistance. The enteric pathogen Salmonella enterica serovar (S.) Typhimurium uses its virulence factors to break colonization resistance through unknown mechanisms. Using metabolite profiling and genetic analysis, we show that the initial rise in luminal pathogen abundance was powered by a combination of aerobic respiration and mixed acid fermentation of simple sugars, such as glucose, which resulted in their depletion from the metabolome. The initial rise in the abundance of the pathogen in the feces coincided with a reduction in the cecal concentrations of acetate and butyrate and an increase in epithelial oxygenation. Notably, these changes in the host environment preceded changes in the microbiota composition. We conclude that changes in the host environment can weaken colonization resistance even in the absence of overt compositional changes in the gut microbiota.
Collapse
Affiliation(s)
- Andrew W L Rogers
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Lauren C Radlinski
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Henry Nguyen
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Connor R Tiffany
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Thaynara Parente Carvalho
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Hugo L P Masson
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Michael L Goodson
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Lalita Bechtold
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Kohei Yamazaki
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA; Laboratory of Veterinary Public Health, School of Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Megan J Liou
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Brittany M Miller
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Scott P Mahan
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Briana M Young
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Aurore M Demars
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Sophie R Gretler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Anaïs B Larabi
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Jee-Yon Lee
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Derek J Bays
- Department of Internal Medicine, Division of Infectious Diseases, School of Medicine, University of California at Davis, One Shields Avenue, Sacramento, CA 95817, USA
| | - Renee M Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA.
| |
Collapse
|
8
|
Machado CM, de-Souza-Ferreira E, Silva GFS, Pimentel FSA, De-Souza EA, Silva-Rodrigues T, Gandara ACP, Zeidler JD, Fernandes-Siqueira LO, De-Queiroz ALFV, Andrade-Silva L, Victória-Martins K, Fernandes-Carvalho C, Chini EN, Passos JF, Da Poian AT, Montero-Lomelí M, Galina A, Masuda CA. Galactose-1-phosphate inhibits cytochrome c oxidase and causes mitochondrial dysfunction in classic galactosemia. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167340. [PMID: 38986816 DOI: 10.1016/j.bbadis.2024.167340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024]
Abstract
Classic galactosemia is an inborn error of metabolism caused by mutations in the GALT gene resulting in the diminished activity of the galactose-1-phosphate uridyltransferase enzyme. This reduced GALT activity leads to the buildup of the toxic intermediate galactose-1-phosphate and a decrease in ATP levels upon exposure to galactose. In this work, we focused our attention on mitochondrial oxidative phosphorylation in the context of this metabolic disorder. We observed that galactose-1-phosphate accumulation reduced respiratory rates in vivo and changed mitochondrial function and morphology in yeast models of galactosemia. These alterations are harmful to yeast cells since the mitochondrial retrograde response is activated as part of the cellular adaptation to galactose toxicity. In addition, we found that galactose-1-phosphate directly impairs cytochrome c oxidase activity of mitochondrial preparations derived from yeast, rat liver, and human cell lines. These results highlight the evolutionary conservation of this biochemical effect. Finally, we discovered that two compounds - oleic acid and dihydrolipoic acid - that can improve the growth of cell models of mitochondrial diseases, were also able to improve galactose tolerance in this model of galactosemia. These results reveal a new molecular mechanism relevant to the pathophysiology of classic galactosemia - galactose-1-phosphate-dependent mitochondrial dysfunction - and suggest that therapies designed to treat mitochondrial diseases may be repurposed to treat galactosemia.
Collapse
Affiliation(s)
- Caio M Machado
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Eduardo de-Souza-Ferreira
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Bioquímica e Biofísica Celular, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Guilherme F S Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Felipe S A Pimentel
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Evandro A De-Souza
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Thaia Silva-Rodrigues
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Bioquímica e Biofísica Celular, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Ana C P Gandara
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Julianna D Zeidler
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Bioquímica e Biofísica Celular, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Lorena O Fernandes-Siqueira
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Bioquímica e Biofísica Celular, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Ana Luiza F V De-Queiroz
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Letícia Andrade-Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Klara Victória-Martins
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Clara Fernandes-Carvalho
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Bioquímica e Biofísica Celular, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Eduardo N Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA; Department of Anesthesiology and Perioperative Medicine Mayo Clinic, Jacksonville, FL 32224, USA
| | - João F Passos
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Andrea T Da Poian
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Bioquímica e Biofísica Celular, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Mónica Montero-Lomelí
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Antonio Galina
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Bioquímica e Biofísica Celular, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Claudio A Masuda
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| |
Collapse
|
9
|
Taylor ZA, Chen P, Noeparvar P, Pham DN, Walker AR, Kitten T, Zeng L. Glycerol metabolism contributes to competition by oral streptococci through production of hydrogen peroxide. J Bacteriol 2024; 206:e0022724. [PMID: 39171915 PMCID: PMC11411925 DOI: 10.1128/jb.00227-24] [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: 06/10/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024] Open
Abstract
As a biological byproduct from both humans and microbes, glycerol's contribution to microbial homeostasis in the oral cavity remains understudied. In this study, we examined glycerol metabolism by Streptococcus sanguinis, a commensal associated with oral health. Genetic mutants of glucose-PTS enzyme II (manL), glycerol metabolism (glp and dha pathways), and transcriptional regulators were characterized with regard to glycerol catabolism, growth, production of hydrogen peroxide (H2O2), transcription, and competition with Streptococcus mutans. Biochemical assays identified the glp pathway as a novel source for H2O2 production by S. sanguinis that is independent of pyruvate oxidase (SpxB). Genetic analysis indicated that the glp pathway requires glycerol and a transcriptional regulator, GlpR, for expression and is negatively regulated by PTS, but not the catabolite control protein, CcpA. Conversely, deletion of either manL or ccpA increased the expression of spxB and a second, H2O2-non-producing glycerol metabolic pathway (dha), indicative of a mode of regulation consistent with conventional carbon catabolite repression (CCR). In a plate-based antagonism assay and competition assays performed with planktonic and biofilm-grown cells, glycerol greatly benefited the competitive fitness of S. sanguinis against S. mutans. The glp pathway appears to be conserved in several commensal streptococci and actively expressed in caries-free plaque samples. Our study suggests that glycerol metabolism plays a more significant role in the ecology of the oral cavity than previously understood. Commensal streptococci, though not able to use glycerol as a sole carbohydrate source for growth, benefit from the catabolism of glycerol through production of both ATP and H2O2. IMPORTANCE Glycerol is an abundant carbohydrate in the oral cavity. However, little is understood regarding the metabolism of glycerol by commensal streptococci, some of the most abundant oral bacteria. This was in part because most streptococci cannot grow on glycerol as the sole carbon source. In this study, we show that Streptococcus sanguinis, a commensal associated with dental health, can degrade glycerol for persistence and competition through two pathways, one of which generates hydrogen peroxide at levels capable of inhibiting Streptococcus mutans. Preliminary studies suggest that several additional commensal streptococci are also able to catabolize glycerol, and glycerol-related genes are actively expressed in human dental plaque samples. Our findings reveal the potential of glycerol to significantly impact microbial homeostasis, which warrants further exploration.
Collapse
Affiliation(s)
- Zachary A. Taylor
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Ping Chen
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Payam Noeparvar
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Danniel N. Pham
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Alejandro R. Walker
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Todd Kitten
- Philips Institute for Oral Health Research, Virginia Commonwealth University School of Dentistry, Richmond, Virginia, USA
| | - Lin Zeng
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| |
Collapse
|
10
|
Taylor ZA, Chen P, Noeparvar P, Pham DN, Walker AR, Kitten T, Zeng L. Glycerol Metabolism Contributes to Competition by Oral Streptococci through Production of Hydrogen Peroxide. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.598274. [PMID: 38979179 PMCID: PMC11230354 DOI: 10.1101/2024.06.28.598274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
As a biological byproduct from both humans and microbes, glycerol's contribution to microbial homeostasis in the oral cavity remains understudied. Here we examined glycerol metabolism by Streptococcus sanguinis, a commensal associated with oral health. Genetic mutants of glucose-PTS enzyme II ( manL ), glycerol metabolism ( glp and dha pathways), and transcriptional regulators were characterized with regard to glycerol catabolism, growth, production of hydrogen peroxide (H 2 O 2 ), transcription, and competition with Streptococcus mutans . Biochemical assays identified the glp pathway as a novel source of H 2 O 2 production by S. sanguinis that is independent of pyruvate oxidase (SpxB). Genetic analysis indicated that the glp pathway requires glycerol and a transcriptional regulator, GlpR, for expression and is negatively regulated by PTS, but not the catabolite control protein, CcpA. Conversely, deletion of either manL or ccpA increased expression of spxB and a second, H 2 O 2 -non-producing glycerol metabolic pathway ( dha ), indicative of a mode of regulation consistent with conventional carbon catabolite repression (CCR). In a plate-based antagonism assay and competition assays performed with planktonic and biofilm-grown cells, glycerol greatly benefited the competitive fitness of S. sanguinis against S. mutans. The glp pathway appears to be conserved in several commensal streptococci and actively expressed in caries-free plaque samples. Our study suggests that glycerol metabolism plays a more significant role in the ecology of the oral cavity than previously understood. Commensal streptococci, though not able to use glycerol as a sole carbohydrate for growth, benefit from catabolism of glycerol through production of both ATP and H 2 O 2 . Importance Glycerol is an abundant carbohydrate found in oral cavity, both due to biological activities of humans and microbes, and as a common ingredient of foods and health care products. However, very little is understood regarding the metabolism of glycerol by some of the most abundant oral bacteria, commensal streptococci. This was in part because most streptococci cannot grow on glycerol as the sole carbon source. Here we show that Streptococcus sanguinis , an oral commensal associated with dental health, can degrade glycerol for persistence and competition through two independent pathways, one of which generates hydrogen peroxide at levels capable of inhibiting a dental pathobiont, Streptococcus mutans . Preliminary studies suggest that several other commensal streptococci are also able to catabolize glycerol, and glycerol-related genes are being actively expressed in human dental plaque samples. Our findings reveal the potential of glycerol to significantly impact microbial homeostasis which warrants further exploration.
Collapse
|
11
|
Hallenbeck M, Chua M, Collins J. The role of the universal sugar transport system components PtsI (EI) and PtsH (HPr) in Enterococcus faecium. FEMS MICROBES 2024; 5:xtae018. [PMID: 38988831 PMCID: PMC11234649 DOI: 10.1093/femsmc/xtae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 07/12/2024] Open
Abstract
Vancomycin-resistant enterococci (VRE) pose a serious threat to public health because of their limited treatment options. Therefore, there is an increasing need to identify novel targets to develop new drugs. Here, we examined the roles of the universal PTS components, PtsI and PtsH, in Enterococcus faecium to determine their roles in carbon metabolism, biofilm formation, stress response, and the ability to compete in the gastrointestinal tract. Clean deletion of ptsHI resulted in a significant reduction in the ability to import and metabolize simple sugars, attenuated growth rate, reduced biofilm formation, and decreased competitive fitness both in vitro and in vivo. However, no significant difference in stress survival was observed when compared with the wild type. These results suggest that targeting universal or specific PTS may provide a novel treatment strategy by reducing the fitness of E. faecium.
Collapse
Affiliation(s)
- Michelle Hallenbeck
- Department of Microbiology & Immunology, University of Louisville, Louisville, KY 40202, United States
- Center for Predictive Medicine, University of Louisville, Louisville, KY 40202, United States
| | - Michelle Chua
- Department of Microbiology & Immunology, University of Louisville, Louisville, KY 40202, United States
- Center for Predictive Medicine, University of Louisville, Louisville, KY 40202, United States
| | - James Collins
- Department of Microbiology & Immunology, University of Louisville, Louisville, KY 40202, United States
- Center for Predictive Medicine, University of Louisville, Louisville, KY 40202, United States
- Center for Microbiomics, Inflammation and Pathogenicity, University of Louisville, Louisville, KY 40202, United States
| |
Collapse
|
12
|
Pan X, Heacock ML, Abdulaziz EN, Violante S, Zuckerman AL, Shrestha N, Yao C, Goodman RP, Cross JR, Cracan V. A genetically encoded tool to increase cellular NADH/NAD + ratio in living cells. Nat Chem Biol 2024; 20:594-604. [PMID: 37884806 PMCID: PMC11045668 DOI: 10.1038/s41589-023-01460-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 09/25/2023] [Indexed: 10/28/2023]
Abstract
Impaired redox metabolism is a key contributor to the etiology of many diseases, including primary mitochondrial disorders, cancer, neurodegeneration and aging. However, mechanistic studies of redox imbalance remain challenging due to limited strategies that can perturb redox metabolism in various cellular or organismal backgrounds. Most studies involving impaired redox metabolism have focused on oxidative stress; consequently, less is known about the settings where there is an overabundance of NADH reducing equivalents, termed reductive stress. Here we introduce a soluble transhydrogenase from Escherichia coli (EcSTH) as a novel genetically encoded tool to promote reductive stress in living cells. When expressed in mammalian cells, EcSTH, and a mitochondrially targeted version (mitoEcSTH), robustly elevated the NADH/NAD+ ratio in a compartment-specific manner. Using this tool, we determined that metabolic and transcriptomic signatures of the NADH reductive stress are cellular background specific. Collectively, our novel genetically encoded tool represents an orthogonal strategy to promote reductive stress.
Collapse
Affiliation(s)
- Xingxiu Pan
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
| | - Mina L Heacock
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
- Calibr, The Scripps Research Institute, La Jolla, CA, USA
| | - Evana N Abdulaziz
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
- Process Development Associate, Amgen, Thousand Oaks, CA, USA
| | - Sara Violante
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Austin L Zuckerman
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
- Program in Mathematics and Science Education, University of California San Diego, San Diego, CA, USA
- Program in Mathematics and Science Education, San Diego State University, San Diego, USA
| | - Nirajan Shrestha
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Canglin Yao
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
| | - Russell P Goodman
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Justin R Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Valentin Cracan
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA.
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
13
|
Murphy AC, McReynolds MR. Toying with reductive stress. Nat Chem Biol 2024; 20:542-543. [PMID: 37884808 DOI: 10.1038/s41589-023-01461-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Affiliation(s)
- Alexandria C Murphy
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | - Melanie R McReynolds
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA.
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
14
|
Kane DL, Burke B, Diaz M, Wolf C, Fonzi WA. Lethal metabolism of Candida albicans respiratory mutants. PLoS One 2024; 19:e0300630. [PMID: 38578754 PMCID: PMC10997084 DOI: 10.1371/journal.pone.0300630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/01/2024] [Indexed: 04/07/2024] Open
Abstract
The destructive impact of fungi in agriculture and animal and human health, coincident with increases in antifungal resistance, underscores the need for new and alternative drug targets to counteract these trends. Cellular metabolism relies on many intermediates with intrinsic toxicity and promiscuous enzymatic activity generates others. Fuller knowledge of these toxic entities and their generation may offer opportunities of antifungal development. From this perspective our observation of media-conditional lethal metabolism in respiratory mutants of the opportunistic fungal pathogen Candida albicans was of interest. C. albicans mutants defective in NADH:ubiquinone oxidoreductase (Complex I of the electron transport chain) exhibit normal growth in synthetic complete medium. In YPD medium, however, the mutants grow normally until early stationary phase whereupon a dramatic loss of viability occurs. Upwards of 90% of cells die over the subsequent four to six hours with a loss of membrane integrity. The extent of cell death was proportional to the amount of BactoPeptone, and to a lesser extent, the amount of yeast extract. YPD medium conditioned by growth of the mutant was toxic to wild-type cells indicating mutant metabolism established a toxic milieu in the media. Conditioned media contained a volatile component that contributed to toxicity, but only in the presence of a component of BactoPeptone. Fractionation experiments revealed purine nucleosides or bases as the synergistic component. GC-mass spectrometry analysis revealed acetal (1,1-diethoxyethane) as the active volatile. This previously unreported and lethal synergistic interaction of acetal and purines suggests a hitherto unrecognized toxic metabolism potentially exploitable in the search for antifungal targets.
Collapse
Affiliation(s)
- D. Lucas Kane
- Department of Chemistry and Medicinal Chemistry Shared Resource, Georgetown University, Washington, DC, United States of America
| | - Brendan Burke
- Department of Microbiology, Georgetown University, Washington, DC, United States of America
| | - Monica Diaz
- Department of Microbiology, Georgetown University, Washington, DC, United States of America
| | - Christian Wolf
- Department of Chemistry and Medicinal Chemistry Shared Resource, Georgetown University, Washington, DC, United States of America
| | - William A. Fonzi
- Department of Microbiology, Georgetown University, Washington, DC, United States of America
| |
Collapse
|
15
|
Xu M, Hu J, Li H, Li K, Xu D. Research overview on the genetic mechanism underlying the biosynthesis of polysaccharide in tuber plants. PeerJ 2024; 12:e17052. [PMID: 38464751 PMCID: PMC10924778 DOI: 10.7717/peerj.17052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/13/2024] [Indexed: 03/12/2024] Open
Abstract
Tuber plants are of great significance in the world as human food crops. Polysaccharides, important metabolites in tuber plants, also serve as a source of innovative drugs with significant pharmacological effects. These drugs are particularly known for their immunomodulation and antitumor properties. To fully exploit the potential value of tuber plant polysaccharides and establish a synthetic system for their targeted synthesis, it is crucial to dissect their metabolic processes and genetic regulatory mechanisms. In this article, we provide a comprehensive summary of the basic pathways involved in the synthesis of various types of tuber plant polysaccharides. We also outline the key research progress that has been made in this area in recent years. We classify the main types and functions of tuber plant polysaccharides and analyze the biosynthetic processes and genetic regulation mechanisms of key enzymes involved in the metabolic pathways of starch, cellulose, pectin, and fructan in tuber plants. We have identified hexokinase and glycosyltransferase as the key enzymes involved in the polysaccharide synthesis process. By elucidating the synthesis pathway of polysaccharides in tuber plants and understanding the underlying mechanism of action of key enzymes in the metabolic pathway, we can provide a theoretical framework for enhancing the yield of polysaccharides and other metabolites in plant culture cells. This will ultimately lead to increased production efficiency.
Collapse
Affiliation(s)
- Mengwei Xu
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiao Hu
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Hongwei Li
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Kunqian Li
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
| | - Delin Xu
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, Guizhou, China
- Guizhou Provincial Demonstration Center of Basic Medical Experimental Teaching, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
16
|
Stanley S, Wang X, Liu Q, Kwon YY, Frey AM, Hicks ND, Vickers AJ, Hui S, Fortune SM. Ongoing evolution of the Mycobacterium tuberculosis lactate dehydrogenase reveals the pleiotropic effects of bacterial adaption to host pressure. PLoS Pathog 2024; 20:e1012050. [PMID: 38422159 DOI: 10.1371/journal.ppat.1012050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/12/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024] Open
Abstract
The bacterial determinants that facilitate Mycobacterium tuberculosis (Mtb) adaptation to the human host environment are poorly characterized. We have sought to decipher the pressures facing the bacterium in vivo by assessing Mtb genes that are under positive selection in clinical isolates. One of the strongest targets of selection in the Mtb genome is lldD2, which encodes a quinone-dependent L-lactate dehydrogenase (LldD2) that catalyzes the oxidation of lactate to pyruvate. Lactate accumulation is a salient feature of the intracellular environment during infection and lldD2 is essential for Mtb growth in macrophages. We determined the extent of lldD2 variation across a set of global clinical isolates and defined how prevalent mutations modulate Mtb fitness. We show the stepwise nature of lldD2 evolution that occurs as a result of ongoing lldD2 selection in the background of ancestral lineage-defining mutations and demonstrate that the genetic evolution of lldD2 additively augments Mtb growth in lactate. Using quinone-dependent antibiotic susceptibility as a functional reporter, we also find that the evolved lldD2 mutations functionally increase the quinone-dependent activity of LldD2. Using 13C-lactate metabolic flux tracing, we find that lldD2 is necessary for robust incorporation of lactate into central carbon metabolism. In the absence of lldD2, label preferentially accumulates in dihydroxyacetone phosphate (DHAP) and glyceraldehyde-3-phosphate (G3P) and is associated with a discernible growth defect, providing experimental evidence for accrued lactate toxicity via the deleterious buildup of sugar phosphates. The evolved lldD2 variants increase lactate incorporation to pyruvate while altering triose phosphate flux, suggesting both an anaplerotic and detoxification benefit to lldD2 evolution. We further show that the mycobacterial cell is transcriptionally sensitive to the changes associated with altered lldD2 activity which affect the expression of genes involved in cell wall lipid metabolism and the ESX- 1 virulence system. Together, these data illustrate a multifunctional role of LldD2 that provides context for the selective advantage of lldD2 mutations in adapting to host stress.
Collapse
Affiliation(s)
- Sydney Stanley
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Xin Wang
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Qingyun Liu
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Young Yon Kwon
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Abigail M Frey
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Nathan D Hicks
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Andrew J Vickers
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Sheng Hui
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Sarah M Fortune
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| |
Collapse
|
17
|
Frizzell JK, Taylor RL, Ryno LM. Constitutive Activation of RpoH and the Addition of L-arabinose Influence Antibiotic Sensitivity of PHL628 E. coli. Antibiotics (Basel) 2024; 13:143. [PMID: 38391529 PMCID: PMC10886279 DOI: 10.3390/antibiotics13020143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/24/2024] Open
Abstract
Antibiotics are used to combat the ever-present threat of infectious diseases, but bacteria are continually evolving an assortment of defenses that enable their survival against even the most potent treatments. While the demand for novel antibiotic agents is high, the discovery of a new agent is exceedingly rare. We chose to focus on understanding how different signal transduction pathways in the gram-negative bacterium Escherichia coli (E. coli) influence the sensitivity of the organism to antibiotics from three different classes: tetracycline, chloramphenicol, and levofloxacin. Using the PHL628 strain of E. coli, we exogenously overexpressed two transcription factors, FliA and RpoH.I54N (a constitutively active mutant), to determine their influence on the minimum inhibitory concentration (MIC) and minimum duration of killing (MDK) concentration for each of the studied antibiotics. We hypothesized that activating these pathways, which upregulate genes that respond to specific stressors, could mitigate bacterial response to antibiotic treatment. We also compared the exogenous overexpression of the constitutively active RpoH mutant to thermal heat shock that has feedback loops maintained. While FliA overexpression had no impact on MIC or antibiotic tolerance, RpoH.I54N overexpression reduced the MIC for tetracycline and chloramphenicol but had no independent impact on antibiotic tolerance. Thermal heat shock alone also did not affect MIC or antibiotic tolerance. L-arabinose, the small molecule used to induce expression in our system, unexpectedly independently increased the MICs for tetracycline (>2-fold) and levofloxacin (3-fold). Additionally, the combination of thermal heat shock and arabinose provided a synergistic, 5-fold increase in MIC for chloramphenicol. Arabinose increased the tolerance, as assessed by MDK99, for chloramphenicol (2-fold) and levofloxacin (4-fold). These experiments highlight the potential of the RpoH pathway to modulate antibiotic sensitivity and the emerging implication of arabinose in enhanced MIC and antibiotic tolerance.
Collapse
Affiliation(s)
- Jenna K Frizzell
- Department of Chemistry and Biochemistry, Oberlin College, Oberlin, OH 44074, USA
| | - Ryan L Taylor
- Department of Chemistry and Biochemistry, Oberlin College, Oberlin, OH 44074, USA
| | - Lisa M Ryno
- Department of Chemistry and Biochemistry, Oberlin College, Oberlin, OH 44074, USA
| |
Collapse
|
18
|
Liu M, Zhang J, Zhou Y, Xiong S, Zhou M, Wu L, Liu Q, Chen Z, Jiang H, Yang J, Liu Y, Wang Y, Chen C, Huang L. Gut microbiota affects the estrus return of sows by regulating the metabolism of sex steroid hormones. J Anim Sci Biotechnol 2023; 14:155. [PMID: 38115159 PMCID: PMC10731813 DOI: 10.1186/s40104-023-00959-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/06/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Sex hormones play important roles in the estrus return of post-weaning sows. Previous studies have demonstrated a complex and bi-directional regulation between sex hormones and gut microbiota. However, the extent to which the gut microbiota affects estrus return of post-weaning sows is largely unknown. RESULTS In this study, we first screened 207 fecal samples from well-phenotyped sows by 16S rRNA gene sequencing and identified significant associations between microbes and estrus return of post-weaning sows. Using metagenomic sequencing data from 85 fecal samples, we identified 37 bacterial species that were significantly associated with estrus return. Normally returning sows were characterized by increased abundances of L. reuteri and P. copri and decreased abundances of B. fragilis, S. suis, and B. pseudolongum. The changes in gut microbial composition significantly altered the functional capacity of steroid hormone biosynthesis in the gut microbiome. The results were confirmed in a validation cohort. Significant changes in sex steroid hormones and related compounds were found between normal and non-return sows via metabolome analysis. An integrated analysis of differential bacterial species, metagenome, and fecal metabolome provided evidence that normal return-associated bacterial species L. reuteri and Prevotella spp. participated in the degradation of pregnenolone, progesterone, and testosterone, thereby promoting estrogen biosynthesis. Furthermore, the microbial metabolites related to sow energy and nutrient supply or metabolic disorders also showed relationships with sow estrus return. CONCLUSIONS An integrated analysis of differentially abundant bacterial species, metagenome, and fecal metabolome revealed the involvement of L. reuteri and Prevotella spp. in sow estrus return. These findings provide deep insight into the role of gut microbiota in the estrus return of post-weaning sows and the complex cross-talk between gut microbiota and sex hormones, suggesting that the manipulation of the gut microbiota could be an effective strategy to improve sow estrus return after weaning.
Collapse
Affiliation(s)
- Min Liu
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Jia Zhang
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Yunyan Zhou
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Shuqi Xiong
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Mengqing Zhou
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Lin Wu
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Qin Liu
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Zhe Chen
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Hui Jiang
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Jiawen Yang
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Yuxin Liu
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Yaxiang Wang
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Congying Chen
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China.
| | - Lusheng Huang
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
19
|
Song G, Plumlee P, Ahn JY, Wong ST, Zhao H. Translational strategies and systems biology insights for blood-brain barrier opening and delivery in brain tumors and Alzheimer's disease. Biomed Pharmacother 2023; 167:115450. [PMID: 37703663 PMCID: PMC10591819 DOI: 10.1016/j.biopha.2023.115450] [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: 07/23/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/15/2023] Open
Abstract
The blood-brain barrier (BBB) plays a critical role in determining the effectiveness of systemic treatments for brain diseases. Over the years, several innovative approaches in BBB opening and drug delivery have been developed and progressed into clinical testing phases, including focused ultrasound (FUS) with circulating microbubbles, mannitol-facilitated delivery of anti-neoplastic drugs, receptor-mediated transcytosis (RMT) by antibody-drug conjugates (ADCs), and viral vectors for gene therapy. We provided a comprehensive review of the most recent clinical applications of these approaches in managing brain tumors and Alzheimer's disease (AD), two major devastating brain diseases. Moreover, the spatial-temporal molecular heterogeneity of the BBB under disease states emphasized the importance of utilizing emerging spatial systems biology approaches to unravel novel targets for intervention within BBB and tailor strategies for enhancing drug delivery to the brain. SEARCH STRATEGY AND SELECTION CRITERIA: Data for this Review were identified by searches of clinicaltrials.gov, MEDLINE, Current Contents, PubMed, and references from relevant articles using the search terms "blood-brain barrier", "CNS drug delivery", "BBB modulation", "clinical trials", "systems biology", "primary or metastatic brain tumors", "Alzheimer's disease". Abstracts and reports from meetings were included only when they related directly to previously published work. Only articles published in English between 1980 and 2023 were included.
Collapse
Affiliation(s)
- Gefei Song
- T. T. and W. F. Chao Center for BRAIN and Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston TX 77030, USA
| | - Pierce Plumlee
- T. T. and W. F. Chao Center for BRAIN and Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston TX 77030, USA
| | - Ju Young Ahn
- T. T. and W. F. Chao Center for BRAIN and Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston TX 77030, USA; Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Stephen Tc Wong
- T. T. and W. F. Chao Center for BRAIN and Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston TX 77030, USA; Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA.
| | - Hong Zhao
- T. T. and W. F. Chao Center for BRAIN and Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston TX 77030, USA.
| |
Collapse
|
20
|
Elston R, Mulligan C, Thomas GH. Flipping the switch: dynamic modulation of membrane transporter activity in bacteria. MICROBIOLOGY (READING, ENGLAND) 2023; 169. [PMID: 37948297 DOI: 10.1099/mic.0.001412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
The controlled entry and expulsion of small molecules across the bacterial cytoplasmic membrane is essential for efficient cell growth and cellular homeostasis. While much is known about the transcriptional regulation of genes encoding transporters, less is understood about how transporter activity is modulated once the protein is functional in the membrane, a potentially more rapid and dynamic level of control. In this review, we bring together literature from the bacterial transport community exemplifying the extensive and diverse mechanisms that have evolved to rapidly modulate transporter function, predominantly by switching activity off. This includes small molecule feedback, inhibition by interaction with small peptides, regulation through binding larger signal transduction proteins and, finally, the emerging area of controlled proteolysis. Many of these examples have been discovered in the context of metal transport, which has to finely balance active accumulation of elements that are essential for growth but can also quickly become toxic if intracellular homeostasis is not tightly controlled. Consistent with this, these transporters appear to be regulated at multiple levels. Finally, we find common regulatory themes, most often through the fusion of additional regulatory domains to transporters, which suggest the potential for even more widespread regulation of transporter activity in biology.
Collapse
Affiliation(s)
- Rory Elston
- Department of Biology, University of York, York, UK
| | | | | |
Collapse
|
21
|
Pokorzynski ND, Groisman EA. How Bacterial Pathogens Coordinate Appetite with Virulence. Microbiol Mol Biol Rev 2023; 87:e0019822. [PMID: 37358444 PMCID: PMC10521370 DOI: 10.1128/mmbr.00198-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023] Open
Abstract
Cells adjust growth and metabolism to nutrient availability. Having access to a variety of carbon sources during infection of their animal hosts, facultative intracellular pathogens must efficiently prioritize carbon utilization. Here, we discuss how carbon source controls bacterial virulence, with an emphasis on Salmonella enterica serovar Typhimurium, which causes gastroenteritis in immunocompetent humans and a typhoid-like disease in mice, and propose that virulence factors can regulate carbon source prioritization by modifying cellular physiology. On the one hand, bacterial regulators of carbon metabolism control virulence programs, indicating that pathogenic traits appear in response to carbon source availability. On the other hand, signals controlling virulence regulators may impact carbon source utilization, suggesting that stimuli that bacterial pathogens experience within the host can directly impinge on carbon source prioritization. In addition, pathogen-triggered intestinal inflammation can disrupt the gut microbiota and thus the availability of carbon sources. By coordinating virulence factors with carbon utilization determinants, pathogens adopt metabolic pathways that may not be the most energy efficient because such pathways promote resistance to antimicrobial agents and also because host-imposed deprivation of specific nutrients may hinder the operation of certain pathways. We propose that metabolic prioritization by bacteria underlies the pathogenic outcome of an infection.
Collapse
Affiliation(s)
- Nick D. Pokorzynski
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
| | - Eduardo A. Groisman
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
- Yale Microbial Sciences Institute, West Haven, Connecticut, USA
| |
Collapse
|
22
|
Han M, Schierstaedt J, Duan Y, Nietschke M, Jechalke S, Wolf J, Hensel M, Neumann-Schaal M, Schikora A. Salmonella enterica relies on carbon metabolism to adapt to agricultural environments. Front Microbiol 2023; 14:1213016. [PMID: 37744895 PMCID: PMC10513388 DOI: 10.3389/fmicb.2023.1213016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/11/2023] [Indexed: 09/26/2023] Open
Abstract
Salmonella enterica, a foodborne and human pathogen, is a constant threat to human health. Agricultural environments, for example, soil and plants, can be ecological niches and vectors for Salmonella transmission. Salmonella persistence in such environments increases the risk for consumers. Therefore, it is necessary to investigate the mechanisms used by Salmonella to adapt to agricultural environments. We assessed the adaptation strategy of S. enterica serovar Typhimurium strain 14028s to agricultural-relevant situations by analyzing the abundance of intermediates in glycolysis and the tricarboxylic acid pathway in tested environments (diluvial sand soil suspension and leaf-based media from tomato and lettuce), as well as in bacterial cells grown in such conditions. By reanalyzing the transcriptome data of Salmonella grown in those environments and using an independent RT-qPCR approach for verification, several genes were identified as important for persistence in root or leaf tissues, including the pyruvate dehydrogenase subunit E1 encoding gene aceE. In vivo persistence assay in tomato leaves confirmed the crucial role of aceE. A mutant in another tomato leaf persistence-related gene, aceB, encoding malate synthase A, displayed opposite persistence features. By comparing the metabolites and gene expression of the wild-type strain and its aceB mutant, fumarate accumulation was discovered as a potential way to replenish the effects of the aceB mutation. Our research interprets the mechanism of S. enterica adaptation to agriculture by adapting its carbon metabolism to the carbon sources available in the environment. These insights may assist in the development of strategies aimed at diminishing Salmonella persistence in food production systems.
Collapse
Affiliation(s)
- Min Han
- Federal Research Centre for Cultivated Plants, Julius Kühn Institute (JKI), Institute for Epidemiology and Pathogen Diagnostics, Braunschweig, Germany
| | - Jasper Schierstaedt
- Federal Research Centre for Cultivated Plants, Julius Kühn Institute (JKI), Institute for Epidemiology and Pathogen Diagnostics, Braunschweig, Germany
- Department Plant-Microbe Systems, Leibniz Institute of Vegetable and Ornamental Crops (IGZ), Großbeeren, Germany
| | - Yongming Duan
- Federal Research Centre for Cultivated Plants, Julius Kühn Institute (JKI), Institute for Epidemiology and Pathogen Diagnostics, Braunschweig, Germany
| | - Monika Nietschke
- Division of Microbiology, Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Sven Jechalke
- Institute of Phytopathology, Research Centre for Biosystems, Land Use and Nutrition (IFZ), Justus-Liebig-University Gießen, Gießen, Germany
| | - Jacqueline Wolf
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Michael Hensel
- Division of Microbiology, Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Meina Neumann-Schaal
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Adam Schikora
- Federal Research Centre for Cultivated Plants, Julius Kühn Institute (JKI), Institute for Epidemiology and Pathogen Diagnostics, Braunschweig, Germany
| |
Collapse
|
23
|
Keller M, Han X, Dörr T. Disrupting Central Carbon Metabolism Increases β-Lactam Antibiotic Susceptibility in Vibrio cholerae. J Bacteriol 2023; 205:e0047622. [PMID: 36840595 PMCID: PMC10029711 DOI: 10.1128/jb.00476-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/31/2023] [Indexed: 02/24/2023] Open
Abstract
Antibiotic tolerance, the ability of bacteria to sustain viability in the presence of typically bactericidal antibiotics for extended time periods, is an understudied contributor to treatment failure. The Gram-negative pathogen Vibrio cholerae, the causative agent of cholera, becomes highly tolerant to β-lactam antibiotics (penicillin and related compounds) in a process requiring the two-component system VxrAB. VxrAB is induced by exposure to cell wall damaging conditions, which results in the differential regulation of >100 genes. While the effectors of VxrAB are relatively well known, VxrAB environment-sensing and activation mechanisms remain a mystery. Here, we used transposon mutagenesis to screen for mutants that spontaneously upregulate VxrAB signaling. This screen was answered by genes known to be required for proper cell envelope homeostasis, validating the approach. Unexpectedly, we also uncovered a new connection between central carbon metabolism and antibiotic tolerance in Vibrio cholerae. Inactivation of pgi (vc0374, coding for glucose-6-phosphate isomerase) resulted in an intracellular accumulation of glucose-6-phosphate and fructose-6-phosphate, concomitant with a marked cell envelope defect, resulting in VxrAB induction. Deletion of pgi also increased sensitivity to β-lactams and conferred a growth defect on salt-free LB, phenotypes that could be suppressed by deleting sugar uptake systems and by supplementing cell wall precursors in the growth medium. Our data suggest an important connection between central metabolism and cell envelope integrity and highlight a potential new target for developing novel antimicrobial agents. IMPORTANCE Antibiotic tolerance (the ability to survive exposure to antibiotics) is a stepping stone toward antibiotic resistance (the ability to grow in the presence of antibiotics), an increasingly common cause of antibiotic treatment failure. The mechanisms promoting tolerance are poorly understood. Here, we identified central carbon metabolism as a key contributor to antibiotic tolerance and resistance. A strain with a mutation in a sugar utilization pathway accumulates metabolites that likely shut down the synthesis of cell wall precursors, which weakens the cell wall and thus increases susceptibility to cell wall-active drugs. Our results illuminate the connection between central carbon metabolism and cell wall homeostasis in V. cholerae and suggest that interfering with metabolism may be a fruitful future strategy for the development of antibiotic adjuvants.
Collapse
Affiliation(s)
- Megan Keller
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA
| | - Xiang Han
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA
| | - Tobias Dörr
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA
- Department of Microbiology, Cornell University, Ithaca, New York, USA
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, USA
| |
Collapse
|
24
|
Thirugnanasambantham P, Bashian E, Zaleski R, Gopalan V. Demonstrating the utility of sugar-phosphate phosphatases in coupled enzyme assays: galactose-1-phosphate uridylyltransferase as proof-of-concept. Glycobiology 2023; 33:95-98. [PMID: 36585843 PMCID: PMC9990984 DOI: 10.1093/glycob/cwac085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/05/2022] [Accepted: 12/17/2022] [Indexed: 01/01/2023] Open
Abstract
During our biochemical characterization of select bacterial phosphatases belonging to the haloacid dehalogenase superfamily of hydrolases, we discovered a strong bias of Salmonella YidA for glucose-1-phosphate (Glc-1-P) over galactose-1-phosphate (Gal-1-P). We sought to exploit this ability of YidA to discriminate these two sugar-phosphate epimers in a simple coupled assay that could be a substitute for current cumbersome alternatives. To this end, we focused on Gal-1-P uridylyltransferase (GalT) that is defective in individuals with classical galactosemia, an inborn disorder. GalT catalyzes the conversion of Gal-1-P and UDP-glucose to Glc-1-P and UDP-galactose. When recombinant YidA was coupled to GalT, the final orthophosphate product (generated from selective hydrolysis of Glc-1-P by YidA) could be easily measured using the inexpensive malachite green reagent. When this new YidA-based colorimetric assay was benchmarked using a recombinant Duarte GalT variant, it yielded kcat/Km values that are ~2.5-fold higher than the standard coupled assay that employs phosphoglucomutase and glucose-6-phosphate dehydrogenase. Although the simpler design of our new GalT coupled assay might find appeal in diagnostics, a testable expectation, we spotlight the GalT example to showcase the untapped potential of sugar-phosphate phosphatases with distinctive substrate-recognition properties for measuring the activity of various metabolic enzymes (e.g. trehalose-6-phosphate synthase, N-acetyl-glucosamine-6-phosphate deacetylase, phosphofructokinase).
Collapse
Affiliation(s)
| | - Eleanor Bashian
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Rosemary Zaleski
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Venkat Gopalan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
25
|
Sabag-Daigle A, Boulanger EF, Thirugnanasambantham P, Law JD, Bogard AJ, Behrman EJ, Gopalan V, Ahmer BMM. Identification of Small-Molecule Inhibitors of the Salmonella FraB Deglycase Using a Live-Cell Assay. Microbiol Spectr 2023; 11:e0460622. [PMID: 36809033 PMCID: PMC10100877 DOI: 10.1128/spectrum.04606-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/18/2023] [Indexed: 02/23/2023] Open
Abstract
Nontyphoidal salmonellosis is one of the most significant foodborne diseases in the United States and globally. There are no vaccines available for human use to prevent this disease, and only broad-spectrum antibiotics are available to treat complicated cases of the disease. However, antibiotic resistance is on the rise and new therapeutics are needed. We previously identified the Salmonella fraB gene, that mutation of causes attenuation of fitness in the murine gastrointestinal tract. The FraB gene product is encoded in an operon responsible for the uptake and utilization of fructose-asparagine (F-Asn), an Amadori product found in several human foods. Mutations in fraB cause an accumulation of the FraB substrate, 6-phosphofructose-aspartate (6-P-F-Asp), which is toxic to Salmonella. The F-Asn catabolic pathway is found only in the nontyphoidal Salmonella serovars, a few Citrobacter and Klebsiella isolates, and a few species of Clostridium; it is not found in humans. Thus, targeting FraB with novel antimicrobials is expected to be Salmonella specific, leaving the normal microbiota largely intact and having no effect on the host. We performed high-throughput screening (HTS) to identify small-molecule inhibitors of FraB using growth-based assays comparing a wild-type Salmonella and a Δfra island mutant control. We screened 224,009 compounds in duplicate. After hit triage and validation, we found three compounds that inhibit Salmonella in an fra-dependent manner, with 50% inhibitory concentration (IC50) values ranging from 89 to 150 μM. Testing these compounds with recombinant FraB and synthetic 6-P-F-Asp confirmed that they are uncompetitive inhibitors of FraB with Ki' (inhibitor constant) values ranging from 26 to 116 μM. IMPORTANCE Nontyphoidal salmonellosis is a serious threat in the United States and globally. We have recently identified an enzyme, FraB, that when mutated renders Salmonella growth defective in vitro and unfit in mouse models of gastroenteritis. FraB is quite rare in bacteria and is not found in humans or other animals. Here, we have identified small-molecule inhibitors of FraB that inhibit the growth of Salmonella. These could provide the foundation for a therapeutic to reduce the duration and severity of Salmonella infections.
Collapse
Affiliation(s)
- Anice Sabag-Daigle
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Erin F. Boulanger
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | | | - Jamison D. Law
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | - Alex J. Bogard
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | - Edward J. Behrman
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | - Venkat Gopalan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | - Brian M. M. Ahmer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
26
|
Inducing Self-Poisoning: a Feasible Antibacterial Strategy? J Bacteriol 2022; 204:e0041122. [PMID: 36383006 PMCID: PMC9765000 DOI: 10.1128/jb.00411-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Development of novel antibacterial strategies is required to tackle the alarming threat for global health due to antimicrobial resistance. In this issue of the Journal of Bacteriology, Boulanger et al. provide evidence supporting that the blocking of metabolic pathways to induce accumulation of toxic intermediates can be a possible approach to combat bacterial infections (E. F. Boulanger, A. Sabag-Daigle, M. Baniasad, K. Kokkinias, et al., J Bacteriol 204:e00344-22, 2022, https://doi.org/10.1128/jb.00344-22).
Collapse
|
27
|
Boulanger EF, Sabag-Daigle A, Baniasad M, Kokkinias K, Schwieters A, Wrighton KC, Wysocki VH, Ahmer BMM. Sugar-Phosphate Toxicities Attenuate Salmonella Fitness in the Gut. J Bacteriol 2022; 204:e0034422. [PMID: 36383008 PMCID: PMC9765134 DOI: 10.1128/jb.00344-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 10/21/2022] [Indexed: 11/17/2022] Open
Abstract
Pathogens are becoming resistant to antimicrobials at an increasing rate, and novel therapeutic strategies are needed. Using Salmonella as a model, we have investigated the induction of sugar-phosphate toxicity as a potential therapeutic modality. The approach entails providing a nutrient while blocking the catabolism of that nutrient, resulting in the accumulation of a toxic intermediate. We hypothesize that this build-up will decrease the fitness of the organism during infection given nutrient availability. We tested this hypothesis using mutants lacking one of seven genes whose mutation is expected to cause the accumulation of a toxic metabolic intermediate. The araD, galE, rhaD, glpD, mtlD, manA, and galT mutants were then provided the appropriate sugars, either in vitro or during gastrointestinal infection of mice. All but the glpD mutant had nutrient-dependent growth defects in vitro, suggestive of sugar-phosphate toxicity. During gastrointestinal infection of mice, five mutants had decreased fitness. Providing the appropriate nutrient in the animal's drinking water was required to cause fitness defects with the rhaD and manA mutants and to enhance the fitness defect of the araD mutant. The galE and mtlD mutants were severely attenuated regardless of the nutrient being provided in the drinking water. Homologs of galE are widespread among bacteria and in humans, rendering the specific targeting of bacterial pathogens difficult. However, the araD, mtlD, and rhaD genes are not present in humans, appear to be rare in most phyla of bacteria, and are common in several genera of Enterobacteriaceae, making the encoded enzymes potential narrow-spectrum therapeutic targets. IMPORTANCE Bacterial pathogens are becoming increasingly resistant to antibiotics. There is an urgent need to identify novel drug targets and therapeutic strategies. In this work we have assembled and characterized a collection of mutations in our model pathogen, Salmonella enterica, that block a variety of sugar utilization pathways in such a way as to cause the accumulation of a toxic sugar-phosphate. Mutations in three genes, rhaD, araD, and mtlD, dramatically decrease the fitness of Salmonella in a mouse model of gastroenteritis, suggesting that RhaD, AraD, and MtlD may be good narrow-spectrum drug targets. The induction of sugar-phosphate toxicities may be a therapeutic strategy that is broadly relevant to other bacterial and fungal pathogens.
Collapse
Affiliation(s)
- Erin F. Boulanger
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Anice Sabag-Daigle
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Maryam Baniasad
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | - Katherine Kokkinias
- Department of Soil and Crop Science, Colorado State University, Ft. Collins, Colorado, USA
| | - Andrew Schwieters
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Kelly C. Wrighton
- Department of Soil and Crop Science, Colorado State University, Ft. Collins, Colorado, USA
| | - Vicki H. Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, USA
| | - Brian M. M. Ahmer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
28
|
Dev C, Jilani SB, Yazdani SS. Adaptation on xylose improves glucose-xylose co-utilization and ethanol production in a carbon catabolite repression (CCR) compromised ethanologenic strain. Microb Cell Fact 2022; 21:154. [PMID: 35933385 PMCID: PMC9356451 DOI: 10.1186/s12934-022-01879-1] [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: 05/19/2022] [Accepted: 07/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Sugar hydrolysates from lignocellulosic biomass are majorly composed of glucose and xylose that can be fermented to biofuels. Bacteria, despite having the natural ability to consume xylose are unable to consume it in presence of glucose due to a carbon catabolite repression (CCR) mechanism. This leads to overall reduced productivity as well as incomplete xylose utilization due to ethanol build-up from glucose utilization. In our effort to develop a strain for simultaneous fermentation of glucose and xylose into ethanol, we deleted ptsG in ethanologenic E. coli SSK42 to make it deficient in CCR and performed adaptive laboratory evolution to achieve accelerated growth rate, sugar consumption and ethanol production. Finally, we performed proteomics study to identify changes that might have been responsible for the observed improved phenotype of the evolved strain. RESULTS The parental strain of SSK42, i.e., wild-type E. coli B, did not co-utilize glucose and xylose as expected. After deleting the ptsG gene encoding the EIIBCGlc subunit of PTS system, glucose consumption is severely affected in wild-type E. coli B. However, the ethanologenic, SSK42 strain, which was evolved in our earlier study on both glucose and xylose, didn't show such a drastic effect of EIIBCGlc deletion, instead consumed glucose first, followed by xylose without delay for switching from one sugar to another. To improve growth on xylose and co-utilization capabilities, the ptsG deleted SSK42 was evolved on xylose. The strain evolved for 78 generations, strain SCD78, displayed significant co-utilization of glucose and xylose sugars. At the bioreactor level, the strain SCD78 produced 3-times the ethanol titer of the parent strain with significant glucose-xylose co-utilization. The rate of glucose and xylose consumption also increased 3.4-fold and 3-fold, respectively. Proteome data indicates significant upregulation of TCA cycle proteins, respiration-related proteins, and some transporters, which may have a role in increasing the total sugar consumption and co-utilization of sugars. CONCLUSION Through adaptive evolution, we have obtained a strain that has a significant glucose-xylose co-utilization phenotype with 3-fold higher total sugar consumption rate and ethanol production rate compared to the unevolved strain. This study also points out that adaptation on xylose is enough to impart glucose-xylose co-utilization property in CCR compromised ethanologenic strain SSK42.
Collapse
Affiliation(s)
- Chandra Dev
- Microbial Engineering Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.,DBT-ICGEB Centre for Advanced Bioenergy Research, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Syed Bilal Jilani
- Microbial Engineering Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Syed Shams Yazdani
- Microbial Engineering Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India. .,DBT-ICGEB Centre for Advanced Bioenergy Research, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.
| |
Collapse
|
29
|
Bhayani J, Iglesias MJ, Minen RI, Cereijo AE, Ballicora MA, Iglesias AA, Asencion Diez MD. Carbohydrate Metabolism in Bacteria: Alternative Specificities in ADP-Glucose Pyrophosphorylases Open Novel Metabolic Scenarios and Biotechnological Tools. Front Microbiol 2022; 13:867384. [PMID: 35572620 PMCID: PMC9093745 DOI: 10.3389/fmicb.2022.867384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
We explored the ability of ADP-glucose pyrophosphorylase (ADP-Glc PPase) from different bacteria to use glucosamine (GlcN) metabolites as a substrate or allosteric effectors. The enzyme from the actinobacteria Kocuria rhizophila exhibited marked and distinctive sensitivity to allosteric activation by GlcN-6P when producing ADP-Glc from glucose-1-phosphate (Glc-1P) and ATP. This behavior is also seen in the enzyme from Rhodococcus spp., the only one known so far to portray this activation. GlcN-6P had a more modest effect on the enzyme from other Actinobacteria (Streptomyces coelicolor), Firmicutes (Ruminococcus albus), and Proteobacteria (Agrobacterium tumefaciens) groups. In addition, we studied the catalytic capacity of ADP-Glc PPases from the different sources using GlcN-1P as a substrate when assayed in the presence of their respective allosteric activators. In all cases, the catalytic efficiency of Glc-1P was 1-2 orders of magnitude higher than GlcN-1P, except for the unregulated heterotetrameric protein (GlgC/GgD) from Geobacillus stearothermophilus. The Glc-1P substrate preference is explained using a model of ADP-Glc PPase from A. tumefaciens based on the crystallographic structure of the enzyme from potato tuber. The substrate-binding domain localizes near the N-terminal of an α-helix, which has a partial positive charge, thus favoring the interaction with a hydroxyl rather than a charged primary amine group. Results support the scenario where the ability of ADP-Glc PPases to use GlcN-1P as an alternative occurred during evolution despite the enzyme being selected to use Glc-1P and ATP for α-glucans synthesis. As an associated consequence in such a process, certain bacteria could have improved their ability to metabolize GlcN. The work also provides insights in designing molecular tools for producing oligo and polysaccharides with amino moieties.
Collapse
Affiliation(s)
- Jaina Bhayani
- Department of Chemistry and Biochemistry, Loyola University Chicago, Chicago, IL, United States
| | - Maria Josefina Iglesias
- Facultad de Bioquímica y Ciencias Biológicas, Instituto de Agrobiotecnología del Litoral, Universidad Nacional del Litoral, Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| | - Romina I. Minen
- Facultad de Bioquímica y Ciencias Biológicas, Instituto de Agrobiotecnología del Litoral, Universidad Nacional del Litoral, Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| | - Antonela E. Cereijo
- Facultad de Bioquímica y Ciencias Biológicas, Instituto de Agrobiotecnología del Litoral, Universidad Nacional del Litoral, Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| | - Miguel A. Ballicora
- Department of Chemistry and Biochemistry, Loyola University Chicago, Chicago, IL, United States
| | - Alberto A. Iglesias
- Facultad de Bioquímica y Ciencias Biológicas, Instituto de Agrobiotecnología del Litoral, Universidad Nacional del Litoral, Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| | - Matias D. Asencion Diez
- Facultad de Bioquímica y Ciencias Biológicas, Instituto de Agrobiotecnología del Litoral, Universidad Nacional del Litoral, Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| |
Collapse
|