1
|
Wang L, Fu N, Wang M, Zhan Z, Luo Y, Wu J, Ren L. Integrative Transcriptome and Metabolome Analysis Reveals Candidate Genes Related to Terpenoid Synthesis in Amylostereum areolatum ( Russulales: Amylostereaceae). J Fungi (Basel) 2025; 11:383. [PMID: 40422717 DOI: 10.3390/jof11050383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/06/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Amylostereum areolatum (Chaillet ex Fr.) Boidin (Russulales: Amylostereaceae) is a symbiotic fungus of Sirex noctilio Fabricius that has ecological significance. Terpenoids are key mediators in fungal-insect interactions, yet the biosynthetic mechanisms of terpenoids in this species remain unclear. Under nutritional conditions that mimic natural growth, A. areolatum was sampled during the lag phase (day 7), exponential phase (day 14), and stationary phase (day 21). Metabolome (solid-phase microextraction (SPME) combined with gas chromatography-mass spectrometry (GC-MS) and liquid chromatography-mass spectrometry (LC-MS)) and transcriptome (Illumina NovaSeq) profiles were integrated to investigate terpenoid-gene correlations. This analysis identified 103 terpenoids in A. areolatum, substantially expanding the known repertoire of terpenoid compounds in this species. Total terpenoid abundance progressively increased across three developmental stages, with triterpenoids and sesquiterpenoids demonstrating the highest diversity and abundance levels. Transcriptomic profiling (61.66 Gb clean data) revealed 26 terpenoid biosynthesis-associated genes, establishing a comprehensive transcriptional framework for fungal terpenoid metabolism. Among 11 differentially expressed genes (DEGs) (|log2Fold Change| ≥ 1, adjusted p < 0.05), HMGS1, HMGR2, and AaTPS1-3 emerged as key regulators potentially governing terpenoid biosynthesis. These findings provide foundational insights into the molecular mechanisms underlying terpenoid production in A. areolatum and related basidiomycetes.
Collapse
Affiliation(s)
- Lixia Wang
- Key Laboratory of Forest Disaster Warning and Control of Yunnan Province, Southwest Forestry University, Kunming 650224, China
- Beijing Key Laboratory for Forest Pest Control, Beijing Forestry University, Beijing 100083, China
| | - Ningning Fu
- Beijing Key Laboratory for Forest Pest Control, Beijing Forestry University, Beijing 100083, China
- College of Forestry, Hebei Agricultural University, Baoding 071000, China
| | - Ming Wang
- Beijing Key Laboratory for Forest Pest Control, Beijing Forestry University, Beijing 100083, China
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
- Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Zhongyi Zhan
- Key Laboratory of Forest Disaster Warning and Control of Yunnan Province, Southwest Forestry University, Kunming 650224, China
- Beijing Key Laboratory for Forest Pest Control, Beijing Forestry University, Beijing 100083, China
| | - Youqing Luo
- Beijing Key Laboratory for Forest Pest Control, Beijing Forestry University, Beijing 100083, China
- Sino-French Joint Laboratory for Invasive Forest Pests in Eurasia, INRAE-Beijing Forestry University, Beijing 100083, China
| | - Jianrong Wu
- Key Laboratory of Forest Disaster Warning and Control of Yunnan Province, Southwest Forestry University, Kunming 650224, China
| | - Lili Ren
- Beijing Key Laboratory for Forest Pest Control, Beijing Forestry University, Beijing 100083, China
- Sino-French Joint Laboratory for Invasive Forest Pests in Eurasia, INRAE-Beijing Forestry University, Beijing 100083, China
| |
Collapse
|
2
|
Chen Y, Xie J, Yang M, Cai R, Cai C, Gan Y, Aweya JJ, Cai G, Wang H. Marine Flavobacteriaceae produce zeaxanthin via the mevalonate pathway. MARINE LIFE SCIENCE & TECHNOLOGY 2025; 7:132-143. [PMID: 40027326 PMCID: PMC11871244 DOI: 10.1007/s42995-024-00268-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 10/18/2024] [Indexed: 03/05/2025]
Abstract
Zeaxanthin, an oxygenated carotenoid derivative with potent antioxidative properties, is produced by many organism taxa. Flavobacteriaceae are widely distributed in marine environments; however, the zeaxanthin biosynthesis property in this family remains incompletely explored. Here, we characterized zeaxanthin production by marine Flavobacteriaceae strains and elucidated underlying molecular mechanisms. Eight Flavobacteriaceae strains were isolated from the phycosphere of various dinoflagellates. Analyses of the zeaxanthin production in these strains revealed yields ranging from 5 to 3289 µg/g of dry cell weight. Genomic and molecular biology analyses revealed the biosynthesized zeaxanthin through the mevalonate (MVA) pathway diverging from the 2-C-methyl-d-erythritol-4-phosphate (MEP) pathway commonly observed in most Gram-negative bacteria. Furthermore, comprehensive genome analyses of 322 culturable marine Flavobacteriale strains indicated that the majority of Flavobacteriaceae members possess the potential to synthesize zeaxanthin using precursors derived from the MVA pathway. These data provide insight into the zeaxanthin biosynthesis property in marine Flavobacteriaceae strains, highlighting their ecological and biotechnological relevance. Supplementary Information The online version contains supplementary material available at 10.1007/s42995-024-00268-4.
Collapse
Affiliation(s)
- Yuerong Chen
- Guangdong Provincial Key Laboratory of Marine Biology, College of Science, Shantou University, Shantou, 515063 China
| | - Jianmin Xie
- Guangdong Provincial Key Laboratory of Marine Biology, College of Science, Shantou University, Shantou, 515063 China
| | - Min Yang
- Guangdong Provincial Key Laboratory of Marine Biology, College of Science, Shantou University, Shantou, 515063 China
| | - Runlin Cai
- Guangdong Provincial Key Laboratory of Marine Biology, College of Science, Shantou University, Shantou, 515063 China
| | - Chao Cai
- Guangdong Provincial Key Laboratory of Marine Biology, College of Science, Shantou University, Shantou, 515063 China
| | - Yongliang Gan
- Guangdong Provincial Key Laboratory of Marine Biology, College of Science, Shantou University, Shantou, 515063 China
| | - Jude Juventus Aweya
- Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, 361021 China
| | - Guanjing Cai
- Guangdong Provincial Key Laboratory of Marine Biology, College of Science, Shantou University, Shantou, 515063 China
| | - Hui Wang
- Guangdong Provincial Key Laboratory of Marine Biology, College of Science, Shantou University, Shantou, 515063 China
| |
Collapse
|
3
|
Chen X, Mo L, Zhang L, Huang L, Gao Z, Peng J, Yu Z, Zhang X. Taxonomic Diversity, Predicted Metabolic Pathway, and Interaction Pattern of Bacterial Community in Sea Urchin Anthocidaris crassispina. Microorganisms 2024; 12:2094. [PMID: 39458402 PMCID: PMC11514596 DOI: 10.3390/microorganisms12102094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Bacterial assemblages associated with sea urchin are critical to their physiology and ecology within marine ecosystems. In this study, we characterized the bacterial communities in wild sea urchin Anthocidaris crassispina captured in Daya Bay, South China Sea. A total of 363 amplicon sequence variants belonging to nine phyla and 141 genera were classified from intestine, body surface, and surrounding seawater samples. Proteobacteria, Firmicutes, and Bacteroidetes were the dominant bacteria phyla found in this study. A network analysis of bacterial interspecies interactions revealed varying complexity, stability, connectivity, and relationship patterns across the samples, with the most intricate network observed in the surrounding seawater. Metagenomic predictions highlighted the distinct bacterial metabolic pathways, with significant differences between intestine and seawater samples. Notably, pathways associated with polysaccharide degradation, including chitin derivatives, starch, and CoM biosynthesis, were markedly abundant, underscoring the gut microbiota's key role in digesting algae. In addition, other metabolic pathways in intestine samples were linked to immune response regulation of sea urchins. Overall, this study provides a comprehensive overview of the bacterial community structure and potential functional roles in A. crassispina.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zonghe Yu
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoyong Zhang
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
4
|
Huang R, Zhang F, Wang X, Yang F, Ma C. Proteomic Profiling in Pediococcus pentosaceus SF11 Exposed to Condensed Tannins from Sainfoin. ACS OMEGA 2024; 9:41148-41156. [PMID: 39398120 PMCID: PMC11465268 DOI: 10.1021/acsomega.3c08947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 08/26/2024] [Accepted: 09/06/2024] [Indexed: 10/15/2024]
Abstract
The antibacterial mechanism of condensed tannins (CTs) obtained from tea has been elucidated, but the mechanism of legume-derived CTs remains unclear. The mechanisms of legume- and tea-derived CTs probably differ due to the diverse compositions of CTs. Previous research found that sainfoin CTs directly inhibited the growth of Pediococcus. The present study investigated the inhibition mechanism of CTs against Pediococcus pentosaceus SF11 (SF11) through proteomic analysis. The results showed that the minimum inhibitory concentration (MIC) of CTs against SF11 was 1500 mg/L and that CTs increased cell membrane permeability in a dose-dependent manner. In total, 418 differentially expressed proteins (DEPs) were identified between the CT treatment and the control, among which 341 were down-regulated and 77 were up-regulated in the CT treatment. The protein interaction network showed that the expression of only two DEPs was highly different between CT treated and control (|log2FC|> 2); the atpD protein was up-regulated in the CT-treated group, which was involved in ATP synthesis; down-regulated DEPs were most involved in lipoteichoic acid synthesis, peptidoglycan synthesis, and glycine metabolism. Twenty-seven proteins were not detected after CT treatment, which were involved in functions including fatty acid synthesis, RNA synthesis and translation, drug resistance, and cell membrane permeability in SF11. Therefore, the findings suggest that the inhibition mechanism of CTs may be related to cell membrane damage and inhibition of cell reproduction.
Collapse
Affiliation(s)
- Rongzheng Huang
- Grassland Science,
School
of Animal Technology, Shihezi University, Shihezi 832000, China
| | - Fanfan Zhang
- Grassland Science,
School
of Animal Technology, Shihezi University, Shihezi 832000, China
| | - Xuzhe Wang
- Grassland Science,
School
of Animal Technology, Shihezi University, Shihezi 832000, China
| | - Fan Yang
- Grassland Science,
School
of Animal Technology, Shihezi University, Shihezi 832000, China
| | - Chunhui Ma
- Grassland Science,
School
of Animal Technology, Shihezi University, Shihezi 832000, China
| |
Collapse
|
5
|
Zheng D, Wilén BM, Öberg O, Wik T, Modin O. "Metagenomics reveal the potential for geosmin and 2-methylisoborneol production across multiple bacterial phyla in recirculating aquaculture systems". Environ Microbiol 2024; 26:e16696. [PMID: 39379175 DOI: 10.1111/1462-2920.16696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/14/2024] [Indexed: 10/10/2024]
Abstract
Geosmin and 2-methylisoborneol (MIB) are known to cause taste-and-odour problems in recirculating aquaculture systems (RAS). Both geosmin and MIB are microbial metabolites belonging to terpenoids. Precursors for terpenoids are biosynthesized via the methylerythritol phosphate (MEP) and the mevalonate (MVA) pathways. We carried out a metagenomic analysis of 50 samples from five RAS to investigate terpenoid biosynthesis and metabolic potential for geosmin and MIB production in RAS microbiomes. A total of 1008 metagenome-assembled genomes (MAGs) representing 26 bacterial and three archaeal phyla were recovered. Although most archaea are thought to use the MVA pathway for terpenoid precursor biosynthesis, an Iainarchaeota archaeal MAG is shown to harbour a complete set of genes encoding the MEP pathway but lacking genes associated with the MVA pathway. In this study, a total of 16 MAGs affiliated with five bacterial phyla (Acidobacteriota, Actinobacteriota, Bacteroidota, Chloroflexota, and Myxococcota) were identified as possessing potential geosmin or MIB synthases. These putative taste and odour producers were diverse, many were taxonomically unidentified at the genus or species level, and their relative abundance differed between the investigated RAS farms. The metagenomic study of the RAS microbiomes revealed a previously unknown phylogenetic diversity of the potential to produce geosmin and MIB.
Collapse
Affiliation(s)
- Dan Zheng
- Division of Water Environment Technology, Department of Architecture and Civil Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Britt-Marie Wilén
- Division of Water Environment Technology, Department of Architecture and Civil Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | | | - Torsten Wik
- Department of Electrical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Oskar Modin
- Division of Water Environment Technology, Department of Architecture and Civil Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
6
|
Zia MP, Jain M, Muthukumaran J, Singh AK. Exploration of potential hit compounds targeting 1-deoxy-d-xylulose 5-phosphate reductoisomerase (IspC) from Acinetobacter baumannii: an in silico investigation. 3 Biotech 2024; 14:72. [PMID: 38362590 PMCID: PMC10864239 DOI: 10.1007/s13205-024-03923-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 01/07/2024] [Indexed: 02/17/2024] Open
Abstract
The emergence of carbapenem-resistant Acinetobacter baumannii, a highly concerning bacterial species designated as a Priority 1: Critical pathogen by the WHO, has become a formidable global threat. In this study, we utilised computational methods to explore the potent molecules capable of inhibiting the IspC enzyme, which plays a crucial role in the methylerythritol 4-phosphate (MEP) biosynthetic pathway. Employing high-throughput virtual screening of small molecules from the Enamine library, we focused on the highly conserved substrate binding site of the DXR target protein, resulting in the identification of 1000 potential compounds. Among these compounds, we selected the top two candidates (Z2615855584 and Z2206320703) based on Lipinski's rule of Five and ADMET filters, along with FR900098, a known IspC inhibitor, and DXP, the substrate of IspC, for molecular dynamics (MD) simulations. The MD simulation trajectories revealed remarkable structural and thermodynamic stability, as well as strong binding affinity, for all the IspC-ligand complexes. Furthermore, binding free energy calculations based on MM/PBSA (Molecular Mechanics/Poisson-Boltzmann Surface Area) methodology demonstrated significant interactions between the selected ligand molecules and IspC. Taking into consideration all the aforementioned criteria, we suggest Z2206320703 as the potent lead candidate against IspC. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-024-03923-w.
Collapse
Affiliation(s)
- Mahrukh Parveez Zia
- Department of Biotechnology, School of Engineering and Technology, Sharda University, P.C. 201310, Greater Noida, Uttar Pradesh India
| | - Monika Jain
- Department of Biotechnology, School of Engineering and Technology, Sharda University, P.C. 201310, Greater Noida, Uttar Pradesh India
| | - Jayaraman Muthukumaran
- Department of Biotechnology, School of Engineering and Technology, Sharda University, P.C. 201310, Greater Noida, Uttar Pradesh India
| | - Amit Kumar Singh
- Department of Biotechnology, School of Engineering and Technology, Sharda University, P.C. 201310, Greater Noida, Uttar Pradesh India
| |
Collapse
|
7
|
Qu G, Liu Y, Ma Q, Li J, Du G, Liu L, Lv X. Progress and Prospects of Natural Glycoside Sweetener Biosynthesis: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15926-15941. [PMID: 37856872 DOI: 10.1021/acs.jafc.3c05074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
To achieve an adequate sense of sweetness with a healthy low-sugar diet, it is necessary to explore and produce sugar alternatives. Recently, glycoside sweeteners and their biosynthetic approaches have attracted the attention of researchers. In this review, we first outlined the synthetic pathways of glycoside sweeteners, including the key enzymes and rate-limiting steps. Next, we reviewed the progress in engineered microorganisms producing glycoside sweeteners, including de novo synthesis, whole-cell catalysis synthesis, and in vitro synthesis. The applications of metabolic engineering strategies, such as cofactor engineering and enzyme modification, in the optimization of glycoside sweetener biosynthesis were summarized. Finally, the prospects of combining enzyme engineering and machine learning strategies to enhance the production of glycoside sweeteners were discussed. This review provides a perspective on synthesizing glycoside sweeteners in microbial cells, theoretically guiding the bioproduction of glycoside sweeteners.
Collapse
Affiliation(s)
- Guanyi Qu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, P. R. China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, P. R. China
- Shandong Jincheng Biological Pharmaceutical Company, Limited, Zibo 255000, P. R. China
| | - Yanfeng Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, P. R. China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, P. R. China
| | - Qinyuan Ma
- Shandong Jincheng Biological Pharmaceutical Company, Limited, Zibo 255000, P. R. China
| | - Jianghua Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, P. R. China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, P. R. China
| | - Guocheng Du
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, P. R. China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, P. R. China
| | - Long Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, P. R. China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, P. R. China
- Yixing Institute of Food Biotechnology Company, Limited, Yixing 214200, P. R. China
- Food Laboratory of Zhongyuan, Jiangnan University, Wuxi 214122, P. R. China
| | - Xueqin Lv
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, P. R. China
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, P. R. China
- Yixing Institute of Food Biotechnology Company, Limited, Yixing 214200, P. R. China
| |
Collapse
|
8
|
Liquid chromatography-mass spectrometry analyses of polyprenyl phosphates in Escherichia coli cells in which genes for isoprenoid synthesis are amplified. J Biosci Bioeng 2023; 135:382-388. [PMID: 36868984 DOI: 10.1016/j.jbiosc.2023.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 03/05/2023]
Abstract
Overproduction of isopentenyl diphosphate by the amplification of the genes for the methylerythritol 4-phosphate pathway, dxs and dxr, is known to be deleterious for the growth of Escherichia coli. We hypothesized that overproduction of one of the endogenous isoprenoids, in addition to isopentenyl diphosphate itself, might be the cause of the reported reduced growth rate and attempted to identify the causative agent. In order to analyze polyprenyl phosphates, they were methylated by the reaction with diazomethane. The resulting dimethyl esters of polyprenyl phosphates with carbon numbers from 40 to 60 were quantitated by high-performance liquid chromatography-mass spectrometric analysis detecting ion peaks of the sodium ion adducts. The E. coli was transformed by a multi-copy plasmid carrying both the dxs and dxr genes. Amplification of dxs and dxr significantly increased the levels of polyprenyl phosphates and 2-octaprenylphenol. The levels of Z,E-mixed polyprenyl phosphates with carbon numbers of 50-60 in the strain in which ispB was co-amplified with dxs and dxr were lower than those in the control strain where only dxs and dxr were amplified. The levels of (all-E)-octaprenyl phosphate and 2-octaprenylphenol in the strains in which ispU/rth or crtE was co-amplified with dxs and dxr were lower than those in the control strain. Although the increase in the level of each isoprenoid intermediate was blocked, the growth rates of these strains were not restored. Neither polyprenyl phosphates nor 2-octaprenylphenol can be determined to be the cause of the growth rate reduction seen with dxs and dxr amplification.
Collapse
|
9
|
Pauleta SR, Grazina R, Carepo MS, Moura JJ, Moura I. Iron-sulfur clusters – functions of an ancient metal site. COMPREHENSIVE INORGANIC CHEMISTRY III 2023:105-173. [DOI: 10.1016/b978-0-12-823144-9.00116-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
10
|
Arjmand G, Haeri MR. Antibacterial Effect of Some Eukaryotic Sterol Biosynthesis Inhibitors. Adv Biomed Res 2022; 11:90. [PMID: 36518857 PMCID: PMC9744079 DOI: 10.4103/abr.abr_291_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/24/2022] [Accepted: 03/12/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Isoprenoids and their derivatives are building blocks for the synthesis of biomolecules with important biological functions such as cholesterol in eukaryotes and lipid carrier undecaprenol, which is involved in cell wall biosynthesis in bacteria. With the global threat of multidrug-resistant bacteria, there is a need for finding new metabolic targets for killing bacteria. In the present study, we examined the impact of eukaryotic sterol biosynthesis inhibitors on the growth of four pathogenic bacteria. MATERIALS AND METHODS Antibacterial effect of HMG CoA reductase inhibitor (simvastatin), farnesyl pyrophosphate synthase inhibitor (alendronate), squalene epoxidase inhibitor (terbinafine), and lanosterol demethylase inhibitor (ketoconazole) were studied against four pathogenic bacteria: two gram-positive bacteria, Staphylococcus aureus and Enterococcus faecalis and two gram-negative bacteria, Escherichia coli and Pseudomonas aeruginosa. Broth microdilution method was used for assessing the antibacterial susceptibility of the components using 96 well plats. MIC and MBC were determined visibly. RESULTS MIC of Ketoconazole for Staphylococcus aureus and Enterococcus faecalis were 0.166 and 1 mg/mL, respectively. Terbinafine had a weak inhibitory effect on Staphylococcus aureus (MIC: 8 mg/mL). Ketoconazole and terbinafine had no inhibitory effect on gram-negative bacteria. MBC of Simvastatin for both Staphylococcus aureus and Enterococcus faecalis was 0.5 mg/mL and of Alendronate for Pseudomonas aeruginosa was 6.6 mg/mL. CONCLUSION Our results show that farnesyl pyrophosphate synthase and class II HMG-CoA reductases inhibitors (ketoconazole and simvastatin) have reasonable antibacterial activity against gram-positive bacteria. These two enzymes provide suitable targets for designing new antibiotics based on modifying the chemical structure of currently used drugs to obtain maximum activity.
Collapse
Affiliation(s)
- Ghasem Arjmand
- Department of Biochemistry, Faculty of Science, Payam Noor University, Tehran Branch, Tehran, Iran
| | - Mohammad Reza Haeri
- Department of Clinical Biochemistry, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran,Address for correspondence: Dr. Mohammad Reza Haeri, Department of Clinical Biochemistry, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran. E-mail:
| |
Collapse
|
11
|
Kwak S, Crook N, Yoneda A, Ahn N, Ning J, Cheng J, Dantas G. Functional mining of novel terpene synthases from metagenomes. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2022; 15:104. [PMID: 36209178 PMCID: PMC9548185 DOI: 10.1186/s13068-022-02189-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/29/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Terpenes are one of the most diverse and abundant classes of natural biomolecules, collectively enabling a variety of therapeutic, energy, and cosmetic applications. Recent genomics investigations have predicted a large untapped reservoir of bacterial terpene synthases residing in the genomes of uncultivated organisms living in the soil, indicating a vast array of putative terpenoids waiting to be discovered. RESULTS We aimed to develop a high-throughput functional metagenomic screening system for identifying novel terpene synthases from bacterial metagenomes by relieving the toxicity of terpene biosynthesis precursors to the Escherichia coli host. The precursor toxicity was achieved using an inducible operon encoding the prenyl pyrophosphate synthetic pathway and supplementation of the mevalonate precursor. Host strain and screening procedures were finely optimized to minimize false positives arising from spontaneous mutations, which avoid the precursor toxicity. Our functional metagenomic screening of human fecal metagenomes yielded a novel β-farnesene synthase, which does not show amino acid sequence similarity to known β-farnesene synthases. Engineered S. cerevisiae expressing the screened β-farnesene synthase produced 120 mg/L β-farnesene from glucose (2.86 mg/g glucose) with a productivity of 0.721 g/L∙h. CONCLUSIONS A unique functional metagenomic screening procedure was established for screening terpene synthases from metagenomic libraries. This research proves the potential of functional metagenomics as a sequence-independent avenue for isolating targeted enzymes from uncultivated organisms in various environmental habitats.
Collapse
Affiliation(s)
- Suryang Kwak
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine in St. Louis, 4515 McKinley Avenue, Room 5121, Campus Box 8510, Saint Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110 USA
| | - Nathan Crook
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine in St. Louis, 4515 McKinley Avenue, Room 5121, Campus Box 8510, Saint Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110 USA
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695 USA
| | - Aki Yoneda
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine in St. Louis, 4515 McKinley Avenue, Room 5121, Campus Box 8510, Saint Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110 USA
| | - Naomi Ahn
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine in St. Louis, 4515 McKinley Avenue, Room 5121, Campus Box 8510, Saint Louis, MO 63110 USA
| | - Jie Ning
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine in St. Louis, 4515 McKinley Avenue, Room 5121, Campus Box 8510, Saint Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110 USA
| | - Jiye Cheng
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine in St. Louis, 4515 McKinley Avenue, Room 5121, Campus Box 8510, Saint Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110 USA
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine in St. Louis, 4515 McKinley Avenue, Room 5121, Campus Box 8510, Saint Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110 USA
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130 USA
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, Saint Louis, MO 63110 USA
| |
Collapse
|
12
|
Dewachter L, Dénéréaz J, Liu X, de Bakker V, Costa C, Baldry M, Sirard JC, Veening JW. Amoxicillin-resistant Streptococcus pneumoniae can be resensitized by targeting the mevalonate pathway as indicated by sCRilecs-seq. eLife 2022; 11:e75607. [PMID: 35748540 PMCID: PMC9363119 DOI: 10.7554/elife.75607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 06/23/2022] [Indexed: 11/23/2022] Open
Abstract
Antibiotic resistance in the important opportunistic human pathogen Streptococcus pneumoniae is on the rise. This is particularly problematic in the case of the β-lactam antibiotic amoxicillin, which is the first-line therapy. It is therefore crucial to uncover targets that would kill or resensitize amoxicillin-resistant pneumococci. To do so, we developed a genome-wide, single-cell based, gene silencing screen using CRISPR interference called sCRilecs-seq (subsets of CRISPR interference libraries extracted by fluorescence activated cell sorting coupled to next generation sequencing). Since amoxicillin affects growth and division, sCRilecs-seq was used to identify targets that are responsible for maintaining proper cell size. Our screen revealed that downregulation of the mevalonate pathway leads to extensive cell elongation. Further investigation into this phenotype indicates that it is caused by a reduced availability of cell wall precursors at the site of cell wall synthesis due to a limitation in the production of undecaprenyl phosphate (Und-P), the lipid carrier that is responsible for transporting these precursors across the cell membrane. The data suggest that, whereas peptidoglycan synthesis continues even with reduced Und-P levels, cell constriction is specifically halted. We successfully exploited this knowledge to create a combination treatment strategy where the FDA-approved drug clomiphene, an inhibitor of Und-P synthesis, is paired up with amoxicillin. Our results show that clomiphene potentiates the antimicrobial activity of amoxicillin and that combination therapy resensitizes amoxicillin-resistant S. pneumoniae. These findings could provide a starting point to develop a solution for the increasing amount of hard-to-treat amoxicillin-resistant pneumococcal infections.
Collapse
Affiliation(s)
- Liselot Dewachter
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore BuildingLausanneSwitzerland
| | - Julien Dénéréaz
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore BuildingLausanneSwitzerland
| | - Xue Liu
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore BuildingLausanneSwitzerland
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, International Cancer Center, Shenzhen University Health Science CenterShenzhenChina
| | - Vincent de Bakker
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore BuildingLausanneSwitzerland
| | - Charlotte Costa
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of LilleLilleFrance
| | - Mara Baldry
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of LilleLilleFrance
| | - Jean-Claude Sirard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of LilleLilleFrance
| | - Jan-Willem Veening
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore BuildingLausanneSwitzerland
| |
Collapse
|
13
|
Pierrel F, Burgardt A, Lee JH, Pelosi L, Wendisch VF. Recent advances in the metabolic pathways and microbial production of coenzyme Q. World J Microbiol Biotechnol 2022; 38:58. [PMID: 35178585 PMCID: PMC8854274 DOI: 10.1007/s11274-022-03242-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/30/2022] [Indexed: 12/13/2022]
Abstract
Coenzyme Q (CoQ) serves as an electron carrier in aerobic respiration and has become an interesting target for biotechnological production due to its antioxidative effect and benefits in supplementation to patients with various diseases. Here, we review discovery of the pathway with a particular focus on its superstructuration and regulation, and we summarize the metabolic engineering strategies for overproduction of CoQ by microorganisms. Studies in model microorganisms elucidated the details of CoQ biosynthesis and revealed the existence of multiprotein complexes composed of several enzymes that catalyze consecutive reactions in the CoQ pathways of Saccharomyces cerevisiae and Escherichia coli. Recent findings indicate that the identity and the total number of proteins involved in CoQ biosynthesis vary between species, which raises interesting questions about the evolution of the pathway and could provide opportunities for easier engineering of CoQ production. For the biotechnological production, so far only microorganisms have been used that naturally synthesize CoQ10 or a related CoQ species. CoQ biosynthesis requires the aromatic precursor 4-hydroxybenzoic acid and the prenyl side chain that defines the CoQ species. Up to now, metabolic engineering strategies concentrated on the overproduction of the prenyl side chain as well as fine-tuning the expression of ubi genes from the ubiquinone modification pathway, resulting in high CoQ yields. With expanding knowledge about CoQ biosynthesis and exploration of new strategies for strain engineering, microbial CoQ production is expected to improve.
Collapse
Affiliation(s)
- Fabien Pierrel
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, 38000, Grenoble, France.
| | - Arthur Burgardt
- Genetics of Prokaryotes, Faculty of Biology and Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Jin-Ho Lee
- Department of Food Science & Biotechnology, Kyungsung University, Busan, South Korea
| | - Ludovic Pelosi
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, 38000, Grenoble, France
| | - Volker F Wendisch
- Genetics of Prokaryotes, Faculty of Biology and Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany.
| |
Collapse
|
14
|
Biosynthesis and regulation of terpenoids from basidiomycetes: exploration of new research. AMB Express 2021; 11:150. [PMID: 34779947 PMCID: PMC8594250 DOI: 10.1186/s13568-021-01304-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/16/2021] [Indexed: 12/15/2022] Open
Abstract
Basidiomycetes, also known as club fungi, consist of a specific group of fungi. Basidiomycetes produce a large number of secondary metabolites, of which sesquiterpenoids, diterpenoids and triterpenoids are the primary components. However, these terpenoids tend to be present in low amounts, which makes it difficult to meet application requirements. Terpenoid biosynthesis improves the quantity of these secondary metabolites. However, current understanding of the biosynthetic mechanism of terpenoids in basidiomycetes is insufficient. Therefore, this article reviews the latest research on the biosynthesis of terpenoids in basidiomycetes and summarizes the CYP450 involved in the biosynthesis of terpenoids in basidiomycetes. We also propose opportunities and challenges for chassis microbial heterologous production of terpenoids in basidiomycetes and provide a reference basis for the better development of basidiomycete engineering.
Collapse
|
15
|
Applying a ‘Metabolic Funnel’ for Phenol Production in Escherichia coli. FERMENTATION 2021. [DOI: 10.3390/fermentation7040216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Phenol is an important petrochemical that is conventionally used as a precursor for synthesizing an array of plastics and fine chemicals. As an emerging alternative to its traditional petrochemical production, multiple enzyme pathways have been engineered to date to enable its renewable biosynthesis from biomass feedstocks, each incorporating unique enzyme chemistries and intermediate molecules. Leveraging all three of the unique phenol biosynthesis pathways reported to date, a series of synthetic ‘metabolic funnels’ was engineered, each with the goal of maximizing net precursor assimilation and flux towards phenol via the parallel co-expression of multiple distinct pathways within the same Escherichia coli host. By constructing and evaluating all possible binary and tertiary pathway combinations, one ‘funnel’ was ultimately identified, which supported enhanced phenol production relative to all three individual pathways by 16 to 69%. Further host engineering to increase endogenous precursor availability then allowed for 26% greater phenol production, reaching a final titer of 554 ± 19 mg/L and 28.8 ± 0.34 mg/g yield on glucose. Lastly, using a diphasic culture including dibutyl phthalate for in situ phenol extraction, final titers were further increased to a maximum of 812 ± 145 mg/L at a yield of 40.6 ± 7.2 mg/g. The demonstrated ‘funneling’ pathway holds similar promise in support of phenol production by other, non-E. coli hosts, while this general approach can be readily extended towards a diversity of other value-added bioproducts of interest.
Collapse
|
16
|
Amiri Moghaddam J, Jautzus T, Alanjary M, Beemelmanns C. Recent highlights of biosynthetic studies on marine natural products. Org Biomol Chem 2021; 19:123-140. [PMID: 33216100 DOI: 10.1039/d0ob01677b] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Marine bacteria are excellent yet often underexplored sources of structurally unique bioactive natural products. In this review we cover the diversity of marine bacterial biomolecules and highlight recent studies on structurally novel natural products. We include different compound classes and discuss the latest progress related to their biosynthetic pathway analysis and engineering: examples range from fatty acids over terpenes to PKS, NRPS and hybrid PKS-NRPS biomolecules.
Collapse
Affiliation(s)
- Jamshid Amiri Moghaddam
- Junior Research Group Chemical Biology of Microbe-Host Interactions, Leibniz Institute for Natural Product Research and Infection Biology (HKI), Beutenbergstr. 11a, 07745 Jena, Germany.
| | | | | | | |
Collapse
|
17
|
Zhan Z, Seager S, Petkowski JJ, Sousa-Silva C, Ranjan S, Huang J, Bains W. Assessment of Isoprene as a Possible Biosignature Gas in Exoplanets with Anoxic Atmospheres. ASTROBIOLOGY 2021; 21:765-792. [PMID: 33798392 DOI: 10.1089/ast.2019.2146] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The search for possible biosignature gases in habitable exoplanet atmospheres is accelerating, although actual observations are likely years away. This work adds isoprene, C5H8, to the roster of biosignature gases. We found that isoprene geochemical formation is highly thermodynamically disfavored and has no known abiotic false positives. The isoprene production rate on Earth rivals that of methane (CH4; ∼500 Tg/year). Unlike methane, on Earth isoprene is rapidly destroyed by oxygen-containing radicals. Although isoprene is predominantly produced by deciduous trees, isoprene production is ubiquitous to a diverse array of evolutionary distant organisms, from bacteria to plants and animals-few, if any, volatile secondary metabolites have a larger evolutionary reach. Although non-photochemical sinks of isoprene may exist, such as degradation of isoprene by life or other high deposition rates, destruction of isoprene in an anoxic atmosphere is mainly driven by photochemistry. Motivated by the concept that isoprene might accumulate in anoxic environments, we model the photochemistry and spectroscopic detection of isoprene in habitable temperature, rocky exoplanet anoxic atmospheres with a variety of atmosphere compositions under different host star ultraviolet fluxes. Limited by an assumed 10 ppm instrument noise floor, habitable atmosphere characterization when using James Webb Space Telescope (JWST) is only achievable with a transit signal similar or larger than that for a super-Earth-sized exoplanet transiting an M dwarf star with an H2-dominated atmosphere. Unfortunately, isoprene cannot accumulate to detectable abundance without entering a run-away phase, which occurs at a very high production rate, ∼100 times the Earth's production rate. In this run-away scenario, isoprene will accumulate to >100 ppm, and its spectral features are detectable with ∼20 JWST transits. One caveat is that some isoprene spectral features are hard to distinguish from those of methane and also from other hydrocarbons containing the isoprene substructure. Despite these challenges, isoprene is worth adding to the menu of potential biosignature gases.
Collapse
Affiliation(s)
- Zhuchang Zhan
- Department of Earth, Atmospheric, and Planetary Sciences, MIT, Cambridge, Massachusetts, USA
| | - Sara Seager
- Department of Earth, Atmospheric, and Planetary Sciences, MIT, Cambridge, Massachusetts, USA
- Department of Physics, MIT, Cambridge, Massachusetts, USA
- Department of Aeronautics and Astronautics, and MIT, Cambridge, Massachusetts, USA
| | - Janusz Jurand Petkowski
- Department of Earth, Atmospheric, and Planetary Sciences, MIT, Cambridge, Massachusetts, USA
| | - Clara Sousa-Silva
- Department of Earth, Atmospheric, and Planetary Sciences, MIT, Cambridge, Massachusetts, USA
| | - Sukrit Ranjan
- Department of Earth, Atmospheric, and Planetary Sciences, MIT, Cambridge, Massachusetts, USA
| | | | - William Bains
- Department of Earth, Atmospheric, and Planetary Sciences, MIT, Cambridge, Massachusetts, USA
- Rufus Scientific, Royston, United Kingdom
| |
Collapse
|
18
|
Krause T, Reichelt M, Gershenzon J, Schmidt A. Analysis of the isoprenoid pathway intermediates, dimethylallyl diphosphate and isopentenyl diphosphate, from crude plant extracts by liquid chromatography tandem mass spectrometry. PHYTOCHEMICAL ANALYSIS : PCA 2020; 31:770-777. [PMID: 32337807 DOI: 10.1002/pca.2941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE We sought to develop a sensitive and accurate analytical method for the detection and quantification of IDP and DMADP as well as their monophosphate derivatives in crude plant extracts. METHODS A liquid chromatography method coupled to tandem mass spectrometry (LC-MS/MS) with multiple reaction monitoring (MRM) was established to measure the amounts of IDP and DMADP down to low picogram levels, which was linear over at least three orders of magnitude. Extracts were enriched using an anion exchanger, and chromatographic separation was achieved using a β-cyclodextrin column. A S-thiolodiphosphate analog of DMADP was employed as an internal standard. RESULTS Dilution series of authentic compounds were used to determine the limits of detection and quantification for IDP, DMADP and their corresponding monophosphates. A survey of plant species producing varying amounts of isoprenoids showed a corresponding variation in IDP and DMADP with the ratio of DMADP/IDP ranging from 4:1 to 2:1. Trace levels of isopentenyl monophosphate (IP) and dimethylallyl monophosphate (DMAP) were also detected. CONCLUSION The LC-MS/MS method described enables absolute quantification of in planta levels of IDP and DMADP for the first time. The method is also suitable for analysing bacterial and animal samples as well as enzyme assays.
Collapse
Affiliation(s)
- Toni Krause
- Department of Biochemistry, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Michael Reichelt
- Department of Biochemistry, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Jonathan Gershenzon
- Department of Biochemistry, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Axel Schmidt
- Department of Biochemistry, Max Planck Institute for Chemical Ecology, Jena, Germany
| |
Collapse
|
19
|
Bateman A. Division of labour in a matrix, rather than phagocytosis or endosymbiosis, as a route for the origin of eukaryotic cells. Biol Direct 2020; 15:8. [PMID: 32345370 PMCID: PMC7187495 DOI: 10.1186/s13062-020-00260-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 02/25/2020] [Indexed: 12/13/2022] Open
Abstract
Abstract Two apparently irreconcilable models dominate research into the origin of eukaryotes. In one model, amitochondrial proto-eukaryotes emerged autogenously from the last universal common ancestor of all cells. Proto-eukaryotes subsequently acquired mitochondrial progenitors by the phagocytic capture of bacteria. In the second model, two prokaryotes, probably an archaeon and a bacterial cell, engaged in prokaryotic endosymbiosis, with the species resident within the host becoming the mitochondrial progenitor. Both models have limitations. A search was therefore undertaken for alternative routes towards the origin of eukaryotic cells. The question was addressed by considering classes of potential pathways from prokaryotic to eukaryotic cells based on considerations of cellular topology. Among the solutions identified, one, called here the “third-space model”, has not been widely explored. A version is presented in which an extracellular space (the third-space), serves as a proxy cytoplasm for mixed populations of archaea and bacteria to “merge” as a transitionary complex without obligatory endosymbiosis or phagocytosis and to form a precursor cell. Incipient nuclei and mitochondria diverge by division of labour. The third-space model can accommodate the reorganization of prokaryote-like genomes to a more eukaryote-like genome structure. Nuclei with multiple chromosomes and mitosis emerge as a natural feature of the model. The model is compatible with the loss of archaeal lipid biochemistry while retaining archaeal genes and provides a route for the development of membranous organelles such as the Golgi apparatus and endoplasmic reticulum. Advantages, limitations and variations of the “third-space” models are discussed. Reviewers This article was reviewed by Damien Devos, Buzz Baum and Michael Gray.
Collapse
Affiliation(s)
- Andrew Bateman
- Division of Experimental Medicine, Department of Medicine, McGill University, Glen Site Pavilion E, 1001 Boulevard Decarie, Montreal, Quebec, H4A 3J1, Canada. .,Centre for Translational Biology, Research Institute of McGill University Health Centre, Glen Site Pavilion E, 1001 Boulevard Decarie, Montreal, Quebec, H4A 3J1, Canada.
| |
Collapse
|
20
|
Abstract
Isoprenoids and their derivatives represent the largest group of organic compounds in nature and are distributed universally in the three domains of life. Isoprenoids are biosynthesized from isoprenyl diphosphate units, generated by two distinctive biosynthetic pathways: mevalonate pathway and methylerthritol 4-phosphate pathway. Archaea and eukaryotes exclusively have the former pathway, while most bacteria have the latter. Some bacteria, however, are known to possess the mevalonate pathway genes. Understanding the evolutionary history of these two isoprenoid biosynthesis pathways in each domain of life is critical since isoprenoids are so interweaved in the architecture of life that they would have had indispensable roles in the early evolution of life. Our study provides a detailed phylogenetic analysis of enzymes involved in the mevalonate pathway and sheds new light on its evolutionary history. The results suggest that a potential mevalonate pathway is present in the recently discovered superphylum Candidate Phyla Radiation (CPR), and further suggest a strong evolutionary relationship exists between archaea and CPR. Interestingly, CPR harbors the characteristics of both the bacterial-type and archaeal-type mevalonate pathways and may retain signatures regarding the ancestral isoprenoid biosynthesis pathway in the last universal common ancestor. Our study supports the ancient origin of the mevalonate pathway in the three domains of life as previously inferred, but concludes that the evolution of the mevalonate pathway was more complex.
Collapse
Affiliation(s)
- Yosuke Hoshino
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA
| | - Eric A Gaucher
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA.,School of Chemistry and Biochemistry, and Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA.,Department of Biology, Georgia State University, Atlanta, GA
| |
Collapse
|
21
|
Verdaguer IB, Zafra CA, Crispim M, Sussmann RA, Kimura EA, Katzin AM. Prenylquinones in Human Parasitic Protozoa: Biosynthesis, Physiological Functions, and Potential as Chemotherapeutic Targets. Molecules 2019; 24:molecules24203721. [PMID: 31623105 PMCID: PMC6832408 DOI: 10.3390/molecules24203721] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/25/2019] [Accepted: 10/01/2019] [Indexed: 12/19/2022] Open
Abstract
Human parasitic protozoa cause a large number of diseases worldwide and, for some of these diseases, there are no effective treatments to date, and drug resistance has been observed. For these reasons, the discovery of new etiological treatments is necessary. In this sense, parasitic metabolic pathways that are absent in vertebrate hosts would be interesting research candidates for the identification of new drug targets. Most likely due to the protozoa variability, uncertain phylogenetic origin, endosymbiotic events, and evolutionary pressure for adaptation to adverse environments, a surprising variety of prenylquinones can be found within these organisms. These compounds are involved in essential metabolic reactions in organisms, for example, prevention of lipoperoxidation, participation in the mitochondrial respiratory chain or as enzymatic cofactors. This review will describe several prenylquinones that have been previously characterized in human pathogenic protozoa. Among all existing prenylquinones, this review is focused on ubiquinone, menaquinone, tocopherols, chlorobiumquinone, and thermoplasmaquinone. This review will also discuss the biosynthesis of prenylquinones, starting from the isoprenic side chains to the aromatic head group precursors. The isoprenic side chain biosynthesis maybe come from mevalonate or non-mevalonate pathways as well as leucine dependent pathways for isoprenoid biosynthesis. Finally, the isoprenic chains elongation and prenylquinone aromatic precursors origins from amino acid degradation or the shikimate pathway is reviewed. The phylogenetic distribution and what is known about the biological functions of these compounds among species will be described, as will the therapeutic strategies associated with prenylquinone metabolism in protozoan parasites.
Collapse
Affiliation(s)
- Ignasi B. Verdaguer
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (I.B.V.); (C.A.Z.); (M.C.); (E.A.K.)
| | - Camila A. Zafra
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (I.B.V.); (C.A.Z.); (M.C.); (E.A.K.)
| | - Marcell Crispim
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (I.B.V.); (C.A.Z.); (M.C.); (E.A.K.)
| | - Rodrigo A.C. Sussmann
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (I.B.V.); (C.A.Z.); (M.C.); (E.A.K.)
- Centro de Formação em Ciências Ambientais, Universidade Federal do Sul da Bahia, Porto Seguro 45810-000 Bahia, Brazil
| | - Emília A. Kimura
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (I.B.V.); (C.A.Z.); (M.C.); (E.A.K.)
| | - Alejandro M. Katzin
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (I.B.V.); (C.A.Z.); (M.C.); (E.A.K.)
- Correspondence: ; Tel.: +55-11-3091-7330; Fax: +5511-3091-7417
| |
Collapse
|
22
|
Engineering Pseudomonas putida for isoprenoid production by manipulating endogenous and shunt pathways supplying precursors. Microb Cell Fact 2019; 18:152. [PMID: 31500633 PMCID: PMC6734295 DOI: 10.1186/s12934-019-1204-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/03/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The soil bacterium Pseudomonas putida is a promising platform for the production of industrially valuable natural compounds. In the case of isoprenoids, the availability of biosynthetic precursors is a major limiting factor. In P. putida and most other bacteria, these precursors are produced from pyruvate and glyceraldehyde 3-phosphate by the methylerythritol 4-phosphate (MEP) pathway, whereas other bacteria synthesize the same precursors from acetyl-CoA using the unrelated mevalonate (MVA) pathway. RESULTS Here we explored different strategies to increase the supply of isoprenoid precursors in P. putida cells using lycopene as a read-out. Because we were not aiming at producing high isoprenoid titers but were primarily interested in finding ways to enhance the metabolic flux to isoprenoids, we engineered the well-characterized P. putida strain KT2440 to produce low but detectable levels of lycopene under conditions in which MEP pathway steps were not saturated. Then, we compared lycopene production in cells expressing the Myxococcus xanthus MVA pathway genes or endogenous MEP pathway genes (dxs, dxr, idi) under the control of IPTG-induced and stress-regulated promoters. We also tested a shunt pathway producing isoprenoid precursors from ribulose 5-phosphate using a mutant version of the Escherichia coli ribB gene. CONCLUSIONS The most successful combination led to a 50-fold increase in lycopene levels, indicating that P. putida can be successfully engineered to substantially increase the supply of metabolic substrates for the production of industrially valuable isoprenoids.
Collapse
|
23
|
Fleitas Martínez O, Cardoso MH, Ribeiro SM, Franco OL. Recent Advances in Anti-virulence Therapeutic Strategies With a Focus on Dismantling Bacterial Membrane Microdomains, Toxin Neutralization, Quorum-Sensing Interference and Biofilm Inhibition. Front Cell Infect Microbiol 2019; 9:74. [PMID: 31001485 PMCID: PMC6454102 DOI: 10.3389/fcimb.2019.00074] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 03/05/2019] [Indexed: 12/11/2022] Open
Abstract
Antimicrobial resistance constitutes one of the major challenges facing humanity in the Twenty-First century. The spread of resistant pathogens has been such that the possibility of returning to a pre-antibiotic era is real. In this scenario, innovative therapeutic strategies must be employed to restrict resistance. Among the innovative proposed strategies, anti-virulence therapy has been envisioned as a promising alternative for effective control of the emergence and spread of resistant pathogens. This review presents some of the anti-virulence strategies that are currently being developed, it will cover strategies focused on quench pathogen quorum sensing (QS) systems, disassemble of bacterial functional membrane microdomains (FMMs), disruption of biofilm formation and bacterial toxin neutralization.
Collapse
Affiliation(s)
- Osmel Fleitas Martínez
- Programa de Pós-Graduação em Patologia Molecular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil.,Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil
| | - Marlon Henrique Cardoso
- Programa de Pós-Graduação em Patologia Molecular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil.,Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil.,S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Suzana Meira Ribeiro
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, Brazil
| | - Octavio Luiz Franco
- Programa de Pós-Graduação em Patologia Molecular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil.,Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil.,S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| |
Collapse
|
24
|
Chellapandi P, Prathiviraj R, Prisilla A. Molecular evolution and functional divergence of IspD homologs in malarial parasites. INFECTION GENETICS AND EVOLUTION 2018; 65:340-349. [DOI: 10.1016/j.meegid.2018.08.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 01/19/2023]
|
25
|
A Defective Undecaprenyl Pyrophosphate Synthase Induces Growth and Morphological Defects That Are Suppressed by Mutations in the Isoprenoid Pathway of Escherichia coli. J Bacteriol 2018; 200:JB.00255-18. [PMID: 29986944 DOI: 10.1128/jb.00255-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/05/2018] [Indexed: 01/10/2023] Open
Abstract
The peptidoglycan exoskeleton shapes bacteria and protects them against osmotic forces, making its synthesis the target of many current antibiotics. Peptidoglycan precursors are attached to a lipid carrier and flipped from the cytoplasm into the periplasm to be incorporated into the cell wall. In Escherichia coli, this carrier is undecaprenyl phosphate (Und-P), which is synthesized as a diphosphate by the enzyme undecaprenyl pyrophosphate synthase (UppS). E. coli MG1655 exhibits wild-type morphology at all temperatures, but one of our laboratory strains (CS109) was highly aberrant when grown at 42°C. This strain contained mutations affecting the Und-P synthetic pathway genes uppS, ispH, and idi Normal morphology was restored by overexpressing uppS or by replacing the mutant (uppS31) with the wild-type allele. Importantly, moving uppS31 into MG1655 was lethal even at 30°C, indicating that the altered enzyme was highly deleterious, but growth was restored by adding the CS109 versions of ispH and idi Purified UppSW31R was enzymatically defective at all temperatures, suggesting that it could not supply enough Und-P during rapid growth unless suppressor mutations were present. We conclude that cell wall synthesis is profoundly sensitive to changes in the pool of polyisoprenoids and that isoprenoid homeostasis exerts a particularly strong evolutionary pressure.IMPORTANCE Bacterial morphology is determined primarily by the overall structure of the semirigid macromolecule peptidoglycan. Not only does peptidoglycan contribute to cell shape, but it also protects cells against lysis caused by excess osmotic pressure. Because it is critical for bacterial survival, it is no surprise that many antibiotics target peptidoglycan biosynthesis. However, important gaps remain in our understanding about how this process is affected by peptidoglycan precursor availability. Here, we report that a mutation altering the enzyme that synthesizes Und-P prevents cells from growing at high temperatures and that compensatory mutations in enzymes functioning upstream of uppS can reverse this phenotype. The results highlight the importance of Und-P metabolism for maintaining normal cell wall synthesis and shape.
Collapse
|
26
|
Zhao J, Yang Y, Yu M, Yao K, Luo X, Qi H, Zhang G, Luo Y. Lanostane-type C 31 triterpenoid derivatives from the fruiting bodies of cultivated Fomitopsis palustris. PHYTOCHEMISTRY 2018; 152:10-21. [PMID: 29715599 DOI: 10.1016/j.phytochem.2018.04.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/15/2018] [Accepted: 04/23/2018] [Indexed: 06/08/2023]
Abstract
Fifteen undescribed and five known lanostane-type C31 triterpenoid derivatives were isolated from the aqueous EtOH extract of the fruiting bodies of cultivated Fomitopsis palustris. Their structures were identified from the spectroscopic data and chemical degradation studies. The structures of palustrisoic acids A and H were confirmed by X-ray crystallography. Polyporenic acid B showed strong cytotoxicity against the HCT116, A549, and HepG2 cell lines with IC50 values of 8.4, 12.1, and 12.2 μM, respectively. Palustrisolides A, C, and G displayed weak cytotoxicity.
Collapse
Affiliation(s)
- Jinzhi Zhao
- Chengdu Institute of Biology, Chinese Academy of Sciences, 9 Section 4, Renmin Road South, Chengdu, 610041, People's Republic of China; University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, People's Republic of China
| | - Yun Yang
- Chengdu Institute of Biology, Chinese Academy of Sciences, 9 Section 4, Renmin Road South, Chengdu, 610041, People's Republic of China
| | - Mengyao Yu
- Institute of Medicinal Fungi, Sichuan Academy of Chinese Medical Sciences, 51 Section 4, Renmin Road South, Chengdu, 610041, People's Republic of China
| | - Ke Yao
- Institute of Medicinal Fungi, Sichuan Academy of Chinese Medical Sciences, 51 Section 4, Renmin Road South, Chengdu, 610041, People's Republic of China
| | - Xia Luo
- Institute of Medicinal Fungi, Sichuan Academy of Chinese Medical Sciences, 51 Section 4, Renmin Road South, Chengdu, 610041, People's Republic of China
| | - Huayi Qi
- Chengdu Institute of Biology, Chinese Academy of Sciences, 9 Section 4, Renmin Road South, Chengdu, 610041, People's Republic of China
| | - Guolin Zhang
- Chengdu Institute of Biology, Chinese Academy of Sciences, 9 Section 4, Renmin Road South, Chengdu, 610041, People's Republic of China.
| | - Yinggang Luo
- Chengdu Institute of Biology, Chinese Academy of Sciences, 9 Section 4, Renmin Road South, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
27
|
Englund E, Shabestary K, Hudson EP, Lindberg P. Systematic overexpression study to find target enzymes enhancing production of terpenes in Synechocystis PCC 6803, using isoprene as a model compound. Metab Eng 2018; 49:164-177. [PMID: 30025762 DOI: 10.1016/j.ymben.2018.07.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/28/2018] [Accepted: 07/08/2018] [Indexed: 11/25/2022]
Abstract
Of the two natural metabolic pathways for making terpenoids, biotechnological utilization of the mevalonate (MVA) pathway has enabled commercial production of valuable compounds, while the more recently discovered but stoichiometrically more efficient methylerythritol phosphate (MEP) pathway is underdeveloped. We conducted a study on the overexpression of each enzyme in the MEP pathway in the unicellular cyanobacterium Synechocystis sp. PCC 6803, to identify potential targets for increasing flux towards terpenoid production, using isoprene as a reporter molecule. Results showed that the enzymes Ipi, Dxs and IspD had the biggest impact on isoprene production. By combining and creating operons out of those genes, isoprene production was increased 2-fold compared to the base strain. A genome-scale model was used to identify targets upstream of the MEP pathway that could redirect flux towards terpenoids. A total of ten reactions from the Calvin-Benson-Bassham cycle, lower glycolysis and co-factor synthesis pathways were probed for their effect on isoprene synthesis by co-expressing them with the MEP enzymes, resulting in a 60% increase in production from the best strain. Lastly, we studied two isoprene synthases with the highest reported catalytic rates. Only by expressing them together with Dxs and Ipi could we get stable strains that produced 2.8 mg/g isoprene per dry cell weight, a 40-fold improvement compared to the initial strain.
Collapse
Affiliation(s)
- Elias Englund
- Department of Chemistry - Ångström, Uppsala University, Box 523, SE-751 20 Uppsala, Sweden; School of Biotechnology, KTH - Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
| | - Kiyan Shabestary
- School of Biotechnology, KTH - Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
| | - Elton P Hudson
- School of Biotechnology, KTH - Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
| | - Pia Lindberg
- Department of Chemistry - Ångström, Uppsala University, Box 523, SE-751 20 Uppsala, Sweden.
| |
Collapse
|
28
|
Patchoulol Production with Metabolically Engineered Corynebacterium glutamicum. Genes (Basel) 2018; 9:genes9040219. [PMID: 29673223 PMCID: PMC5924561 DOI: 10.3390/genes9040219] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/10/2018] [Accepted: 04/16/2018] [Indexed: 12/25/2022] Open
Abstract
Patchoulol is a sesquiterpene alcohol and an important natural product for the perfume industry. Corynebacterium glutamicum is the prominent host for the fermentative production of amino acids with an average annual production volume of ~6 million tons. Due to its robustness and well established large-scale fermentation, C. glutamicum has been engineered for the production of a number of value-added compounds including terpenoids. Both C40 and C50 carotenoids, including the industrially relevant astaxanthin, and short-chain terpenes such as the sesquiterpene valencene can be produced with this organism. In this study, systematic metabolic engineering enabled construction of a patchoulol producing C. glutamicum strain by applying the following strategies: (i) construction of a farnesyl pyrophosphate-producing platform strain by combining genomic deletions with heterologous expression of ispA from Escherichia coli; (ii) prevention of carotenoid-like byproduct formation; (iii) overproduction of limiting enzymes from the 2-c-methyl-d-erythritol 4-phosphate (MEP)-pathway to increase precursor supply; and (iv) heterologous expression of the plant patchoulol synthase gene PcPS from Pogostemon cablin. Additionally, a proof of principle liter-scale fermentation with a two-phase organic overlay-culture medium system for terpenoid capture was performed. To the best of our knowledge, the patchoulol titers demonstrated here are the highest reported to date with up to 60 mg L−1 and volumetric productivities of up to 18 mg L−1 d−1.
Collapse
|
29
|
Ghavami M, Merino EF, Yao ZK, Elahi R, Simpson ME, Fernández-Murga ML, Butler JH, Casasanta MA, Krai PM, Totrov MM, Slade DJ, Carlier PR, Cassera MB. Biological Studies and Target Engagement of the 2- C-Methyl-d-Erythritol 4-Phosphate Cytidylyltransferase (IspD)-Targeting Antimalarial Agent (1 R,3 S)-MMV008138 and Analogs. ACS Infect Dis 2018; 4:549-559. [PMID: 29072835 DOI: 10.1021/acsinfecdis.7b00159] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Malaria continues to be one of the deadliest diseases worldwide, and the emergence of drug resistance parasites is a constant threat. Plasmodium parasites utilize the methylerythritol phosphate (MEP) pathway to synthesize isopentenyl pyrophosphate (IPP) and dimethylallyl pyrophosphate (DMAPP), which are essential for parasite growth. Previously, we and others identified that the Malaria Box compound MMV008138 targets the apicoplast and that parasite growth inhibition by this compound can be reversed by supplementation of IPP. Further work has revealed that MMV008138 targets the enzyme 2- C-methyl-d-erythritol 4-phosphate cytidylyltransferase (IspD) in the MEP pathway, which converts MEP and cytidine triphosphate (CTP) to cytidinediphosphate methylerythritol (CDP-ME) and pyrophosphate. In this work, we sought to gain insight into the structure-activity relationships by probing the ability of MMV008138 analogs to inhibit PfIspD recombinant enzyme. Here, we report PfIspD inhibition data for fosmidomycin (FOS) and 19 previously disclosed analogs and report parasite growth and PfIspD inhibition data for 27 new analogs of MMV008138. In addition, we show that MMV008138 does not target the recently characterized human IspD, reinforcing MMV008138 as a prototype of a new class of species-selective IspD-targeting antimalarial agents.
Collapse
Affiliation(s)
- Maryam Ghavami
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Hahn Hall South, 800 West Campus Drive, Blacksburg, Virginia 24061, United States
| | - Emilio F. Merino
- Department of Biochemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Engel Hall, 340 West Campus Drive, Blacksburg, Virginia 24061, United States
- Department of Biochemistry and Molecular Biology and Center for Tropical and Emerging Global Diseases (CTEGD), University of Georgia, 120 Green Street, Athens, Georgia 30602, United States
| | - Zhong-Ke Yao
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Hahn Hall South, 800 West Campus Drive, Blacksburg, Virginia 24061, United States
| | - Rubayet Elahi
- Department of Biochemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Engel Hall, 340 West Campus Drive, Blacksburg, Virginia 24061, United States
| | - Morgan E. Simpson
- Department of Biochemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Engel Hall, 340 West Campus Drive, Blacksburg, Virginia 24061, United States
| | - Maria L. Fernández-Murga
- Department of Biochemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Engel Hall, 340 West Campus Drive, Blacksburg, Virginia 24061, United States
| | - Joshua H. Butler
- Department of Biochemistry and Molecular Biology and Center for Tropical and Emerging Global Diseases (CTEGD), University of Georgia, 120 Green Street, Athens, Georgia 30602, United States
| | - Michael A. Casasanta
- Department of Biochemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Engel Hall, 340 West Campus Drive, Blacksburg, Virginia 24061, United States
| | - Priscilla M. Krai
- Department of Biochemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Engel Hall, 340 West Campus Drive, Blacksburg, Virginia 24061, United States
| | - Maxim M. Totrov
- Molsoft LLC, 11199 Sorrento Valley Road, San Diego, California 92121, United States
| | - Daniel J. Slade
- Department of Biochemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Engel Hall, 340 West Campus Drive, Blacksburg, Virginia 24061, United States
| | - Paul R. Carlier
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Hahn Hall South, 800 West Campus Drive, Blacksburg, Virginia 24061, United States
| | - Maria Belen Cassera
- Department of Biochemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Engel Hall, 340 West Campus Drive, Blacksburg, Virginia 24061, United States
- Department of Biochemistry and Molecular Biology and Center for Tropical and Emerging Global Diseases (CTEGD), University of Georgia, 120 Green Street, Athens, Georgia 30602, United States
| |
Collapse
|
30
|
Puri P, Liangpunsakul S, Christensen JE, Shah VH, Kamath PS, Gores GJ, Walker S, Comerford M, Katz B, Borst A, Yu Q, Kumar DP, Mirshahi F, Radaeva S, Chalasani NP, Crabb DW, Sanyal AJ, for the TREAT Consortium. The circulating microbiome signature and inferred functional metagenomics in alcoholic hepatitis. Hepatology 2018; 67:1284-1302. [PMID: 29083504 PMCID: PMC5867221 DOI: 10.1002/hep.29623] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 09/16/2017] [Accepted: 10/25/2017] [Indexed: 12/12/2022]
Abstract
Intestinal dysbiosis is implicated in alcoholic hepatitis (AH). However, changes in the circulating microbiome, its association with the presence and severity of AH, and its functional relevance in AH is unknown. Qualitative and quantitative assessment of changes in the circulating microbiome were performed by sequencing bacterial DNA in subjects with moderate AH (MAH) (n = 18) or severe AH (SAH) (n = 19). These data were compared with heavy drinking controls (HDCs) without obvious liver disease (n = 19) and non-alcohol-consuming controls (NACs, n = 20). The data were related to endotoxin levels and markers of monocyte activation. Linear discriminant analysis effect size (LEfSe) analysis, inferred metagenomics, and predictive functional analysis using PICRUSt were performed. There was a significant increase in 16S copies/ng DNA both in MAH (P < 0.01) and SAH (P < 0.001) subjects. Compared with NACs, the relative abundance of phylum Bacteroidetes was significantly decreased in HDCs, MAH, and SAH (P < 0.001). In contrast, all alcohol-consuming groups had enrichment with Fusobacteria; this was greatest for HDCs and decreased progressively in MAH and SAH. Subjects with SAH had significantly higher endotoxemia (P = 0.01). Compared with alcohol-consuming groups, predictive functional metagenomics indicated an enrichment of bacteria with genes related to methanogenesis and denitrification. Furthermore, both HDCs and SAH showed activation of a type III secretion system that has been linked to gram-negative bacterial virulence. Metagenomics in SAH versus NACs predicted increased isoprenoid synthesis via mevalonate and anthranilate degradation, known modulators of gram-positive bacterial growth and biofilm production, respectively. CONCLUSION Heavy alcohol consumption appears to be the primary driver of changes in the circulating microbiome associated with a shift in its inferred metabolic functions. (Hepatology 2018;67:1284-1302).
Collapse
Affiliation(s)
- Puneet Puri
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Virginia Commonwealth University, Richmond, VA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Roudebush Veterans Administration Medical Center, Indianapolis, IN
| | - Jeffrey E. Christensen
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), F-31432 Toulouse Cedex 4, France
| | - Vijay H. Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Patrick S. Kamath
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Susan Walker
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Virginia Commonwealth University, Richmond, VA
| | - Megan Comerford
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Barry Katz
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN
| | - Andrew Borst
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN
| | - Qigui Yu
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Divya P. Kumar
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Virginia Commonwealth University, Richmond, VA
| | - Faridoddin Mirshahi
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Virginia Commonwealth University, Richmond, VA
| | - Svetlana Radaeva
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD
| | - Naga P. Chalasani
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Roudebush Veterans Administration Medical Center, Indianapolis, IN
| | - David W. Crabb
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Roudebush Veterans Administration Medical Center, Indianapolis, IN
- Eskenazi Health, Indianapolis, IN
| | - Arun J. Sanyal
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Virginia Commonwealth University, Richmond, VA
| | | |
Collapse
|
31
|
Schempp FM, Drummond L, Buchhaupt M, Schrader J. Microbial Cell Factories for the Production of Terpenoid Flavor and Fragrance Compounds. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:2247-2258. [PMID: 28418659 DOI: 10.1021/acs.jafc.7b00473] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Terpenoid flavor and fragrance compounds are of high interest to the aroma industry. Microbial production offers an alternative sustainable access to the desired terpenoids independent of natural sources. Genetically engineered microorganisms can be used to synthesize terpenoids from cheap and renewable resources. Due to its modular architecture, terpenoid biosynthesis is especially well suited for the microbial cell factory concept: a platform host engineered for a high flux toward the central C5 prenyl diphosphate precursors enables the production of a broad range of target terpenoids just by varying the pathway modules converting the C5 intermediates to the product of interest. In this review typical terpenoid flavor and fragrance compounds marketed or under development by biotech and aroma companies are given, and the specificities of the aroma market are discussed. The main part of this work focuses on key strategies and recent advances to engineer microbes to become efficient terpenoid producers.
Collapse
Affiliation(s)
- Florence M Schempp
- DECHEMA-Forschungsinstitut, Industrial Biotechnology , Theodor-Heuss-Allee 25 , 60486 Frankfurt am Main , Germany
| | - Laura Drummond
- DECHEMA-Forschungsinstitut, Industrial Biotechnology , Theodor-Heuss-Allee 25 , 60486 Frankfurt am Main , Germany
| | - Markus Buchhaupt
- DECHEMA-Forschungsinstitut, Industrial Biotechnology , Theodor-Heuss-Allee 25 , 60486 Frankfurt am Main , Germany
| | - Jens Schrader
- DECHEMA-Forschungsinstitut, Industrial Biotechnology , Theodor-Heuss-Allee 25 , 60486 Frankfurt am Main , Germany
| |
Collapse
|
32
|
Biosynthesis and production of sabinene: current state and perspectives. Appl Microbiol Biotechnol 2017; 102:1535-1544. [DOI: 10.1007/s00253-017-8695-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/03/2017] [Accepted: 12/04/2017] [Indexed: 01/10/2023]
|
33
|
López-Lara IM, Geiger O. Bacterial lipid diversity. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1287-1299. [DOI: 10.1016/j.bbalip.2016.10.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 10/10/2016] [Accepted: 10/11/2016] [Indexed: 11/25/2022]
|
34
|
Remodeling of the Streptococcus mutans proteome in response to LrgAB and external stresses. Sci Rep 2017; 7:14063. [PMID: 29070798 PMCID: PMC5656683 DOI: 10.1038/s41598-017-14324-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/09/2017] [Indexed: 11/24/2022] Open
Abstract
The Streptococcus mutans Cid/Lrg system represents an ideal model to study how this organism withstands various stressors encountered in the oral cavity. Mutation of lrgAB renders S. mutans more sensitive to oxidative, heat, and vancomycin stresses. Here, we have performed a comprehensive proteomics experiment using label-free quantitative mass spectrometry to compare the proteome changes of wild type UA159 and lrgAB mutant strains in response to these same stresses. Importantly, many of identified proteins showed either a strikingly large fold-change, or were completely suppressed or newly induced in response to a particular stress condition. Notable stress proteome changes occurred in a variety of functional categories, including amino acid biosynthesis, energy metabolism, protein synthesis, transport/binding, and transcriptional/response regulators. In the non-stressed growth condition, mutation of lrgAB significantly altered the abundance of 76 proteins (a fold change >1.4, or <0.6, p-value <0.05) and several of these matched the stress proteome of the wild type strain. Interestingly, the statistical correlation between the proteome changes and corresponding RNA-seq transcriptomic studies was relatively low (rho(ρ) <0.16), suggesting that adaptation to a new environment may require radical proteome turnover or metabolic remodeling. Collectively, this study reinforces the importance of LrgAB to the S. mutans stress response.
Collapse
|
35
|
Hui X, Hua SH, Wu QQ, Li H, Gao WY. Antimicrobial mechanism of epigallocatechin gallate and gallocatechin gallate: They target 1-deoxy-d-xylulose 5-phosphate reductoisomerase, the key enzyme of the MEP terpenoid biosynthetic pathway. Arch Biochem Biophys 2017; 622:1-8. [PMID: 28428039 DOI: 10.1016/j.abb.2017.04.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 03/31/2017] [Accepted: 04/16/2017] [Indexed: 01/10/2023]
Abstract
The catechins EGCG and GCG show a variety of pharmacological activities, especially an antibacterial capacity, but their modes of antimicrobial action have not been fully elucidated. 1-Deoxy-d-xylulose 5-phosphate reductoisomerase (DXR), the first key enzyme in the MEP pathway for terpenoid biosynthesis, is a recently validated antimicrobial target. In order to disclose the antibacterial mechanism of EGCG and GCG, the DXR inhibitory activity of them was investigated in this study. The data show that EGCG and GCG both could specifically suppress the activity of DXR, with EGCG exhibiting relatively low effect against DXR (IC50 about 210 μM) and GCG displaying strong activity (IC50 27.5 μM). In addition, studies on inhibition kinetics of the catechins against DXR demonstrate that they are competitive inhibitors of DXR against DXP and uncompetitive inhibitors with respect to NADPH. Meanwhile, the possible interactions between DXR and the catechine, esyth onlols were simulated via docking experiments.
Collapse
Affiliation(s)
- Xian Hui
- National Engineering Research Center for Miniaturised Detection Systems and College of Life Sciences, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Shui-Hong Hua
- National Engineering Research Center for Miniaturised Detection Systems and College of Life Sciences, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Qian-Qian Wu
- National Engineering Research Center for Miniaturised Detection Systems and College of Life Sciences, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Heng Li
- National Engineering Research Center for Miniaturised Detection Systems and College of Life Sciences, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China
| | - Wen-Yun Gao
- National Engineering Research Center for Miniaturised Detection Systems and College of Life Sciences, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
36
|
Antimicrobial mechanism of the major active essential oil compounds and their structure–activity relationship. Med Chem Res 2016. [DOI: 10.1007/s00044-016-1762-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
37
|
Kirby J, Dietzel KL, Wichmann G, Chan R, Antipov E, Moss N, Baidoo EEK, Jackson P, Gaucher SP, Gottlieb S, LaBarge J, Mahatdejkul T, Hawkins KM, Muley S, Newman JD, Liu P, Keasling JD, Zhao L. Engineering a functional 1-deoxy-D-xylulose 5-phosphate (DXP) pathway in Saccharomyces cerevisiae. Metab Eng 2016; 38:494-503. [PMID: 27989805 DOI: 10.1016/j.ymben.2016.10.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/19/2016] [Accepted: 10/25/2016] [Indexed: 01/28/2023]
Abstract
Isoprenoids are used in many commercial applications and much work has gone into engineering microbial hosts for their production. Isoprenoids are produced either from acetyl-CoA via the mevalonate pathway or from pyruvate and glyceraldehyde 3-phosphate via the 1-deoxy-D-xylulose 5-phosphate (DXP) pathway. Saccharomyces cerevisiae exclusively utilizes the mevalonate pathway to synthesize native isoprenoids and in fact the alternative DXP pathway has never been found or successfully reconstructed in the eukaryotic cytosol. There are, however, several advantages to isoprenoid synthesis via the DXP pathway, such as a higher theoretical yield, and it has long been a goal to transplant the pathway into yeast. In this work, we investigate and address barriers to DXP pathway functionality in S. cerevisiae using a combination of synthetic biology, biochemistry and metabolomics. We report, for the first time, functional expression of the DXP pathway in S. cerevisiae. Under low aeration conditions, an engineered strain relying solely on the DXP pathway for isoprenoid biosynthesis achieved an endpoint biomass 80% of that of the same strain using the mevalonate pathway.
Collapse
Affiliation(s)
- James Kirby
- California Institute of Quantitative Biosciences (QB3), University of California, Berkeley, CA 94702, USA; Joint BioEnergy Institute, Emeryville, CA 94608, USA
| | - Kevin L Dietzel
- Amyris, Inc., 5885 Hollis Street, Suite 100, Emeryville, CA 94608, USA
| | - Gale Wichmann
- Amyris, Inc., 5885 Hollis Street, Suite 100, Emeryville, CA 94608, USA
| | - Rossana Chan
- California Institute of Quantitative Biosciences (QB3), University of California, Berkeley, CA 94702, USA; Joint BioEnergy Institute, Emeryville, CA 94608, USA
| | - Eugene Antipov
- Amyris, Inc., 5885 Hollis Street, Suite 100, Emeryville, CA 94608, USA
| | - Nathan Moss
- Amyris, Inc., 5885 Hollis Street, Suite 100, Emeryville, CA 94608, USA
| | | | - Peter Jackson
- Amyris, Inc., 5885 Hollis Street, Suite 100, Emeryville, CA 94608, USA
| | - Sara P Gaucher
- Amyris, Inc., 5885 Hollis Street, Suite 100, Emeryville, CA 94608, USA
| | - Shayin Gottlieb
- Amyris, Inc., 5885 Hollis Street, Suite 100, Emeryville, CA 94608, USA
| | - Jeremy LaBarge
- Amyris, Inc., 5885 Hollis Street, Suite 100, Emeryville, CA 94608, USA
| | - Tina Mahatdejkul
- Amyris, Inc., 5885 Hollis Street, Suite 100, Emeryville, CA 94608, USA
| | - Kristy M Hawkins
- Amyris, Inc., 5885 Hollis Street, Suite 100, Emeryville, CA 94608, USA
| | - Sheela Muley
- Amyris, Inc., 5885 Hollis Street, Suite 100, Emeryville, CA 94608, USA
| | - Jack D Newman
- Amyris, Inc., 5885 Hollis Street, Suite 100, Emeryville, CA 94608, USA
| | - Pinghua Liu
- Department of Chemistry, Boston University, Boston, MA 02215, USA
| | - Jay D Keasling
- California Institute of Quantitative Biosciences (QB3), University of California, Berkeley, CA 94702, USA; Joint BioEnergy Institute, Emeryville, CA 94608, USA; Departments of Chemical & Biomolecular Engineering and of Bioengineering, University of California, Berkeley, CA 94702, USA; Biological Systems & Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94702, USA; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kogle Allé, DK2970 Hørsholm, Denmark
| | - Lishan Zhao
- Amyris, Inc., 5885 Hollis Street, Suite 100, Emeryville, CA 94608, USA.
| |
Collapse
|
38
|
Lipko A, Swiezewska E. Isoprenoid generating systems in plants - A handy toolbox how to assess contribution of the mevalonate and methylerythritol phosphate pathways to the biosynthetic process. Prog Lipid Res 2016; 63:70-92. [PMID: 27133788 DOI: 10.1016/j.plipres.2016.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 01/07/2016] [Accepted: 04/22/2016] [Indexed: 12/21/2022]
Abstract
Isoprenoids comprise an astonishingly diverse group of metabolites with numerous potential and actual applications in medicine, agriculture and the chemical industry. Generation of efficient platforms producing isoprenoids is a target of numerous laboratories. Such efforts are generally enhanced if the native biosynthetic routes can be identified, and if the regulatory mechanisms responsible for the biosynthesis of the compound(s) of interest can be determined. In this review a critical summary of the techniques applied to establish the contribution of the two alternative routes of isoprenoid production operating in plant cells, the mevalonate and methylerythritol pathways, with a focus on their co-operation (cross-talk) is presented. Special attention has been paid to methodological aspects of the referred studies, in order to give the reader a deeper understanding for the nuances of these powerful techniques. This review has been designed as an organized toolbox, which might offer the researchers comments useful both for project design and for interpretation of results obtained.
Collapse
Affiliation(s)
- Agata Lipko
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland.
| | - Ewa Swiezewska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland.
| |
Collapse
|
39
|
Muskal SM, Sliman J, Kokai-Kun J, Pimentel M, Wacher V, Gottlieb K. Lovastatin lactone may improve irritable bowel syndrome with constipation (IBS-C) by inhibiting enzymes in the archaeal methanogenesis pathway. F1000Res 2016; 5:606. [PMID: 27347377 PMCID: PMC4909102 DOI: 10.12688/f1000research.8406.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2016] [Indexed: 12/14/2023] Open
Abstract
UNLABELLED Methane produced by the methanoarchaeon Methanobrevibacter smithii ( M. smithii) has been linked to constipation, irritable bowel syndrome with constipation (IBS-C), and obesity. Lovastatin, which demonstrates a cholesterol-lowering effect by the inhibition of HMG-CoA reductase, may also have an anti-methanogenesis effect through direct inhibition of enzymes in the archaeal methanogenesis pathway. We conducted protein-ligand docking experiments to evaluate this possibility. Results are consistent with recent clinical findings. METHODS F420-dependent methylenetetrahydromethanopterin dehydrogenase ( mtd), a key methanogenesis enzyme was modeled for two different methanogenic archaea: M. smithii and Methanopyrus kandleri. Once protein models were developed, ligand-binding sites were identified. Multiple ligands and their respective protonation, isomeric and tautomeric representations were docked into each site, including F420-coenzyme (natural ligand), lactone and β-hydroxyacid forms of lovastatin and simvastatin, and other co-complexed ligands found in related crystal structures. RESULTS 1) Generally, for each modeled site the lactone form of the statins had more favorable site interactions compared to F420; 2) The statin lactone forms generally had the most favorable docking scores, even relative to the native template PDB ligands; and 3) The statin β-hydroxyacid forms had less favorable docking scores, typically scoring in the middle with some of the F420 tautomeric forms. Consistent with these computational results were those from a recent phase II clinical trial ( NCT02495623) with a proprietary, modified-release lovastatin-lactone (SYN-010) in patients with IBS-C, which showed a reduction in symptoms and breath methane levels, compared to placebo. CONCLUSION The lactone form of lovastatin exhibits preferential binding over the native-F420 coenzyme ligand in silico and thus could inhibit the activity of the key M. smithii methanogenesis enzyme mtd in vivo. Statin lactones may thus exert a methane-reducing effect that is distinct from cholesterol lowering activity, which requires HMGR inhibition by statin β-hydroxyacid forms.
Collapse
|
40
|
Muskal SM, Sliman J, Kokai-Kun J, Pimentel M, Wacher V, Gottlieb K. Lovastatin lactone may improve irritable bowel syndrome with constipation (IBS-C) by inhibiting enzymes in the archaeal methanogenesis pathway. F1000Res 2016; 5:606. [PMID: 27347377 PMCID: PMC4909102 DOI: 10.12688/f1000research.8406.3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2016] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED Methane produced by the methanoarchaeon Methanobrevibacter smithii ( M. smithii) has been linked to constipation, irritable bowel syndrome with constipation (IBS-C), and obesity. Lovastatin, which demonstrates a cholesterol-lowering effect by the inhibition of HMG-CoA reductase, may also have an anti-methanogenesis effect through direct inhibition of enzymes in the archaeal methanogenesis pathway. We conducted protein-ligand docking experiments to evaluate this possibility. Results are consistent with recent clinical findings. METHODS F420-dependent methylenetetrahydromethanopterin dehydrogenase ( mtd), a key methanogenesis enzyme was modeled for two different methanogenic archaea: M. smithii and Methanopyrus kandleri. Once protein models were developed, ligand-binding sites were identified. Multiple ligands and their respective protonation, isomeric and tautomeric representations were docked into each site, including F420-coenzyme (natural ligand), lactone and β-hydroxyacid forms of lovastatin and simvastatin, and other co-complexed ligands found in related crystal structures. RESULTS 1) Generally, for each modeled site the lactone form of the statins had more favorable site interactions compared to F420; 2) The statin lactone forms generally had the most favorable docking scores, even relative to the native template PDB ligands; and 3) The statin β-hydroxyacid forms had less favorable docking scores, typically scoring in the middle with some of the F420 tautomeric forms. Consistent with these computational results were those from a recent phase II clinical trial ( NCT02495623) with a proprietary, modified-release lovastatin-lactone (SYN-010) in patients with IBS-C, which showed a reduction in symptoms and breath methane levels, compared to placebo. CONCLUSION The lactone form of lovastatin exhibits preferential binding over the native-F420 coenzyme ligand in silico and thus could inhibit the activity of the key M. smithii methanogenesis enzyme mtd in vivo. Statin lactones may thus exert a methane-reducing effect that is distinct from cholesterol lowering activity, which requires HMGR inhibition by statin β-hydroxyacid forms.
Collapse
|
41
|
Gottlieb K, Wacher V, Sliman J, Pimentel M. Review article: inhibition of methanogenic archaea by statins as a targeted management strategy for constipation and related disorders. Aliment Pharmacol Ther 2016; 43:197-212. [PMID: 26559904 PMCID: PMC4737270 DOI: 10.1111/apt.13469] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 09/29/2015] [Accepted: 10/20/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND Observational studies show a strong association between delayed intestinal transit and the production of methane. Experimental data suggest a direct inhibitory activity of methane on the colonic and ileal smooth muscle and a possible role for methane as a gasotransmitter. Archaea are the only confirmed biological sources of methane in nature and Methanobrevibacter smithii is the predominant methanogen in the human intestine. AIM To review the biosynthesis and composition of archaeal cell membranes, archaeal methanogenesis and the mechanism of action of statins in this context. METHODS Narrative review of the literature. RESULTS Statins can inhibit archaeal cell membrane biosynthesis without affecting bacterial numbers as demonstrated in livestock and humans. This opens the possibility of a therapeutic intervention that targets a specific aetiological factor of constipation while protecting the intestinal microbiome. While it is generally believed that statins inhibit methane production via their effect on cell membrane biosynthesis, mediated by inhibition of the HMG-CoA reductase, there is accumulating evidence for an alternative or additional mechanism of action where statins inhibit methanogenesis directly. It appears that this other mechanism may predominate when the lactone form of statins, particularly lovastatin lactone, is administered. CONCLUSIONS Clinical development appears promising. A phase 2 clinical trial is currently in progress that evaluates the effect of lovastatin lactone on methanogenesis and symptoms in patients with irritable bowel syndrome with constipation. The review concludes with an outlook for the future and subsequent work that needs to be done.
Collapse
Affiliation(s)
| | - V. Wacher
- Synthetic BiologicsInc.RockvilleMDUSA
| | - J. Sliman
- Synthetic BiologicsInc.RockvilleMDUSA
| | - M. Pimentel
- GastroenterologyCedars‐Sinai Medical CenterLos AngelesCAUSA
| |
Collapse
|
42
|
Ma T, Zhou Y, Li X, Zhu F, Cheng Y, Liu Y, Deng Z, Liu T. Genome mining of astaxanthin biosynthetic genes from Sphingomonas sp. ATCC 55669 for heterologous overproduction in Escherichia coli. Biotechnol J 2015; 11:228-37. [PMID: 26580858 PMCID: PMC5064606 DOI: 10.1002/biot.201400827] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/07/2015] [Accepted: 10/19/2015] [Indexed: 02/01/2023]
Abstract
As a highly valued keto-carotenoid, astaxanthin is widely used in nutritional supplements and pharmaceuticals. Therefore, the demand for biosynthetic astaxanthin and improved efficiency of astaxanthin biosynthesis has driven the investigation of metabolic engineering of native astaxanthin producers and heterologous hosts. However, microbial resources for astaxanthin are limited. In this study, we found that the α-Proteobacterium Sphingomonas sp. ATCC 55669 could produce astaxanthin naturally. We used whole-genome sequencing to identify the astaxanthin biosynthetic pathway using a combined PacBio-Illumina approach. The putative astaxanthin biosynthetic pathway in Sphingomonas sp. ATCC 55669 was predicted. For further confirmation, a high-efficiency targeted engineering carotenoid synthesis platform was constructed in E. coli for identifying the functional roles of candidate genes. All genes involved in astaxanthin biosynthesis showed discrete distributions on the chromosome. Moreover, the overexpression of exogenous E. coli idi in Sphingomonas sp. ATCC 55669 increased astaxanthin production by 5.4-fold. This study described a new astaxanthin producer and provided more biosynthesis components for bioengineering of astaxanthin in the future.
Collapse
Affiliation(s)
- Tian Ma
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Yuanjie Zhou
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Xiaowei Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Fayin Zhu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Yongbo Cheng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Yi Liu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Zixin Deng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Tiangang Liu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan, China. .,Hubei Engineering Laboratory for Synthetic Microbiology, Wuhan Institute of Biotechnology, Wuhan, China. .,Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Wuhan, China.
| |
Collapse
|
43
|
Grazziotin AL, Vidal NM, Venancio TM. Uncovering major genomic features of essential genes in Bacteria and a methanogenic Archaea. FEBS J 2015; 282:3395-3411. [PMID: 26084810 DOI: 10.1111/febs.13350] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 06/02/2015] [Accepted: 06/15/2015] [Indexed: 12/19/2022]
Abstract
Identification of essential genes is critical to understanding the physiology of a species, proposing novel drug targets and uncovering minimal gene sets required for life. Although essential gene sets of several organisms have been determined using large-scale mutagenesis techniques, systematic studies addressing their conservation, genomic context and functions remain scant. Here we integrate 17 essential gene sets from genome-wide in vitro screenings and three gene collections required for growth in vivo, encompassing 15 Bacteria and one Archaea. We refine and generalize important theories proposed using Escherichia coli. Essential genes are typically monogenic and more conserved than nonessential genes. Genes required in vivo are less conserved than those essential in vitro, suggesting that more divergent strategies are deployed when the organism is stressed by the host immune system and unstable nutrient availability. We identified essential analogous pathways that would probably be missed by orthology-based essentiality prediction strategies. For example, Streptococcus sanguinis carries horizontally transferred isoprenoid biosynthesis genes that are widespread in Archaea. Genes specifically essential in Mycobacterium tuberculosis and Burkholderia pseudomallei are reported as potential drug targets. Moreover, essential genes are not only preferentially located in operons, but also occupy the first position therein, supporting the influence of their regulatory regions in driving transcription of whole operons. Finally, these important genomic features are shared between Bacteria and at least one Archaea, suggesting that high order properties of gene essentiality and genome architecture were probably present in the last universal common ancestor or evolved independently in the prokaryotic domains.
Collapse
Affiliation(s)
- Ana Laura Grazziotin
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil.,National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Newton Medeiros Vidal
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil.,National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Thiago Motta Venancio
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil
| |
Collapse
|
44
|
Sohlenkamp C, Geiger O. Bacterial membrane lipids: diversity in structures and pathways. FEMS Microbiol Rev 2015; 40:133-59. [DOI: 10.1093/femsre/fuv008] [Citation(s) in RCA: 571] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2015] [Indexed: 12/22/2022] Open
|
45
|
Nonselective mevalonate kinase inhibitor as a novel class of antibacterial agents. CHOLESTEROL 2015; 2015:147601. [PMID: 25692035 PMCID: PMC4321089 DOI: 10.1155/2015/147601] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/08/2014] [Accepted: 01/01/2015] [Indexed: 11/18/2022]
Abstract
Introduction. There are a few evidences about targeting isoprenoids biosynthesis pathway in bacteria for finding new antibiotics. This study was conducted to assess antibacterial effects of vanadyl sulfate (VS), one of the mevalonate kinase inhibitors to find a new target for killing bacteria. Materials and Methods. Antibacterial effect of VS alone and in combination with glycine or EDTA was assessed on Escherichia coli and Pseudomonas aeruginosa as Gram-negative and Staphylococcus aureus and Enterococcus faecalis as Gram-positive bacteria using serial dilution method and minimum inhibitory concentrations (MICs) identified. Result. MICs for S. aureus and E. coli were 4 and 8 mg/mL, respectively. VS could not affect the growth of two other bacteria. However, VS in combination with glycine not only inhibited the growth of E. faecalis and P. aeruginosa, but also reduced MICs for VS-sensitive bacteria (S. aureus and E. coli). EDTA could reduce MIC for E. coli and P. aeruginosa. Conclusion. VS could inhibit the growth of S. aurous and E. coli, and adding glycine or EDTA improved VS antibacterial activity presumably via instability of the cell wall and enhanced transport of VS through bacterial cell wall. Inhibition of the isoprenoid pathway might provide new tools to overcome bacterial resistance.
Collapse
|
46
|
Enhancing Terpene yield from sugars via novel routes to 1-deoxy-d-xylulose 5-phosphate. Appl Environ Microbiol 2014; 81:130-8. [PMID: 25326299 DOI: 10.1128/aem.02920-14] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Terpene synthesis in the majority of bacterial species, together with plant plastids, takes place via the 1-deoxy-d-xylulose 5-phosphate (DXP) pathway. The first step of this pathway involves the condensation of pyruvate and glyceraldehyde 3-phosphate by DXP synthase (Dxs), with one-sixth of the carbon lost as CO2. A hypothetical novel route from a pentose phosphate to DXP (nDXP) could enable a more direct pathway from C5 sugars to terpenes and also circumvent regulatory mechanisms that control Dxs, but there is no enzyme known that can convert a sugar into its 1-deoxy equivalent. Employing a selection for complementation of a dxs deletion in Escherichia coli grown on xylose as the sole carbon source, we uncovered two candidate nDXP genes. Complementation was achieved either via overexpression of the wild-type E. coli yajO gene, annotated as a putative xylose reductase, or via various mutations in the native ribB gene. In vitro analysis performed with purified YajO and mutant RibB proteins revealed that DXP was synthesized in both cases from ribulose 5-phosphate (Ru5P). We demonstrate the utility of these genes for microbial terpene biosynthesis by engineering the DXP pathway in E. coli for production of the sesquiterpene bisabolene, a candidate biodiesel. To further improve flux into the pathway from Ru5P, nDXP enzymes were expressed as fusions to DXP reductase (Dxr), the second enzyme in the DXP pathway. Expression of a Dxr-RibB(G108S) fusion improved bisabolene titers more than 4-fold and alleviated accumulation of intracellular DXP.
Collapse
|
47
|
Genetically engineering cyanobacteria to convert CO2, water, and light into the long-chain hydrocarbon farnesene. Appl Microbiol Biotechnol 2014; 98:9869-77. [DOI: 10.1007/s00253-014-6118-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/23/2014] [Accepted: 09/25/2014] [Indexed: 01/05/2023]
|
48
|
Workalemahu G, Wang H, Puan KJ, Nada MH, Kuzuyama T, Jones BD, Jin C, Morita CT. Metabolic engineering of Salmonella vaccine bacteria to boost human Vγ2Vδ2 T cell immunity. THE JOURNAL OF IMMUNOLOGY 2014; 193:708-21. [PMID: 24943221 DOI: 10.4049/jimmunol.1302746] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Human Vγ2Vδ2 T cells monitor isoprenoid metabolism by recognizing foreign (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP), a metabolite in the 2-C-methyl-D-erythritol-4-phosphate pathway used by most eubacteria and apicomplexan parasites, and self isopentenyl pyrophosphate, a metabolite in the mevalonate pathway used by humans. Whereas microbial infections elicit prolonged expansion of memory Vγ2Vδ2 T cells, immunization with prenyl pyrophosphates or aminobisphosphonates elicit short-term Vγ2Vδ2 expansion with rapid anergy and deletion upon subsequent immunizations. We hypothesized that a live, attenuated bacterial vaccine that overproduces HMBPP would elicit long-lasting Vγ2Vδ2 T cell immunity by mimicking a natural infection. Therefore, we metabolically engineered the avirulent aroA(-) Salmonella enterica serovar Typhimurium SL7207 strain by deleting the gene for LytB (the downstream enzyme from HMBPP) and functionally complementing for this loss with genes encoding mevalonate pathway enzymes. LytB(-) Salmonella SL7207 had high HMBPP levels, infected human cells as efficiently as did the wild-type bacteria, and stimulated large ex vivo expansions of Vγ2Vδ2 T cells from human donors. Importantly, vaccination of a rhesus monkey with live lytB(-) Salmonella SL7207 stimulated a prolonged expansion of Vγ2Vδ2 T cells without significant side effects or anergy induction. These studies provide proof-of-principle that metabolic engineering can be used to derive live bacterial vaccines that boost Vγ2Vδ2 T cell immunity. Similar engineering of metabolic pathways to produce lipid Ags or B vitamin metabolite Ags could be used to derive live bacterial vaccine for other unconventional T cells that recognize nonpeptide Ags.
Collapse
Affiliation(s)
- Grefachew Workalemahu
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246
| | - Hong Wang
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246
| | - Kia-Joo Puan
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648
| | - Mohanad H Nada
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246; Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Tomohisa Kuzuyama
- Biotechnology Research Center, The University of Tokyo, Tokyo 113-8657, Japan
| | - Bradley D Jones
- Department of Microbiology, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Interdisciplinary Graduate Program in Genetics, University of Iowa Carver College of Medicine, Iowa City, IA 52242; and Inflammation Program, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Chenggang Jin
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246
| | - Craig T Morita
- Division of Immunology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Department of Veterans Affairs, Iowa City Health Care System, Iowa City, IA 52246; Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242;
| |
Collapse
|
49
|
The effect of chain length and unsaturation on Mtb Dxr inhibition and antitubercular killing activity of FR900098 analogs. Bioorg Med Chem Lett 2013; 24:649-53. [PMID: 24360562 DOI: 10.1016/j.bmcl.2013.11.067] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 11/20/2013] [Accepted: 11/25/2013] [Indexed: 11/20/2022]
Abstract
Inhibition of the nonmevalonate pathway (NMP) of isoprene biosynthesis has been examined as a source of new antibiotics with novel mechanisms of action. Dxr is the best studied of the NMP enzymes and several reports have described potent Dxr inhibitors. Many of these compounds are structurally related to natural products fosmidomycin and FR900098, each bearing retrohydroxamate and phosphonate groups. We synthesized a series of compounds with two to five methylene units separating these groups to examine what linker length was optimal and tested for inhibition against Mtb Dxr. We synthesized ethyl and pivaloyl esters of these compounds to increase lipophilicity and improve inhibition of Mtb growth. Our results show that propyl or propenyl linker chains are optimal. Propenyl analog 22 has an IC50 of 1.07 μM against Mtb Dxr. The pivaloyl ester of 22, compound 26, has an MIC of 9.4 μg/mL, representing a significant improvement in antitubercular potency in this class of compounds.
Collapse
|
50
|
Carretero-Paulet L, Lipska A, Pérez-Gil J, Sangari FJ, Albert VA, Rodríguez-Concepción M. Evolutionary diversification and characterization of the eubacterial gene family encoding DXR type II, an alternative isoprenoid biosynthetic enzyme. BMC Evol Biol 2013; 13:180. [PMID: 24004839 PMCID: PMC3847144 DOI: 10.1186/1471-2148-13-180] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 08/16/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Isoprenoids constitute a vast family of natural compounds performing diverse and essential functions in all domains of life. In most eubacteria, isoprenoids are synthesized through the methylerythritol 4-phosphate (MEP) pathway. The production of MEP is usually catalyzed by deoxyxylulose 5-phosphate reductoisomerase (DXR-I) but a few organisms use an alternative DXR-like enzyme (DXR-II). RESULTS Searches through 1498 bacterial complete proteomes detected 130 sequences with similarity to DXR-II. Phylogenetic analysis identified three well-resolved clades: the DXR-II family (clustering 53 sequences including eleven experimentally verified as functional enzymes able to produce MEP), and two previously uncharacterized NAD(P)-dependent oxidoreductase families (designated DLO1 and DLO2 for DXR-II-like oxidoreductases 1 and 2). Our analyses identified amino acid changes critical for the acquisition of DXR-II biochemical function through type-I functional divergence, two of them mapping onto key residues for DXR-II activity. DXR-II showed a markedly discontinuous distribution, which was verified at several levels: taxonomic (being predominantly found in Alphaproteobacteria and Firmicutes), metabolic (being mostly found in bacteria with complete functional MEP pathways with or without DXR-I), and phenotypic (as no biological/phenotypic property was found to be preferentially distributed among DXR-II-containing strains, apart from pathogenicity in animals). By performing a thorough comparative sequence analysis of GC content, 3:1 dinucleotide frequencies, codon usage and codon adaptation indexes (CAI) between DXR-II sequences and their corresponding genomes, we examined the role of horizontal gene transfer (HGT), as opposed to an scenario of massive gene loss, in the evolutionary origin and diversification of the DXR-II subfamily in bacteria. CONCLUSIONS Our analyses support a single origin of the DXR-II family through functional divergence, in which constitutes an exceptional model of acquisition and maintenance of redundant gene functions between non-homologous genes as a result of convergent evolution. Subsequently, although old episodic events of HGT could not be excluded, the results supported a prevalent role of gene loss in explaining the distribution of DXR-II in specific pathogenic eubacteria. Our results highlight the importance of the functional characterization of evolutionary shortcuts in isoprenoid biosynthesis for screening specific antibacterial drugs and for regulating the production of isoprenoids of human interest.
Collapse
Affiliation(s)
- Lorenzo Carretero-Paulet
- Institute for Plant Molecular and Cell Biology - IBMCP (CSIC-UPV), Integrative Systems Biology Group, C/ Ingeniero Fausto Elio s/n., Valencia 46022, Spain
- Department of Biological Sciences, SUNY-University at Buffalo, North Campus. 109 Cooke Hall, Buffalo, NY 14260, USA
| | - Agnieszka Lipska
- Institute for Plant Molecular and Cell Biology - IBMCP (CSIC-UPV), Integrative Systems Biology Group, C/ Ingeniero Fausto Elio s/n., Valencia 46022, Spain
| | - Jordi Pérez-Gil
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Barcelona 08193, Spain
| | - Félix J Sangari
- Department of Molecular Biology, Universidad de Cantabria and Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), UC-CSIC-SODERCAN, Avda. de los Castros s/n, Santander E-39005, Cantabria, Spain
| | - Victor A Albert
- Institute for Plant Molecular and Cell Biology - IBMCP (CSIC-UPV), Integrative Systems Biology Group, C/ Ingeniero Fausto Elio s/n., Valencia 46022, Spain
| | - Manuel Rodríguez-Concepción
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Barcelona 08193, Spain
| |
Collapse
|