1
|
Nestor BJ, Bird T, Severn-Ellis AA, Bayer PE, Ranathunge K, Prodhan MA, Dassanayake M, Batley J, Edwards D, Lambers H, Finnegan PM. Identification and expression analysis of Phosphate Transporter 1 (PHT1) genes in the highly phosphorus-use-efficient Hakea prostrata (Proteaceae). PLANT, CELL & ENVIRONMENT 2024; 47:5021-5038. [PMID: 39136390 DOI: 10.1111/pce.15088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/10/2024] [Accepted: 08/02/2024] [Indexed: 11/06/2024]
Abstract
Heavy and costly use of phosphorus (P) fertiliser is often needed to achieve high crop yields, but only a small amount of applied P fertiliser is available to most crop plants. Hakea prostrata (Proteaceae) is endemic to the P-impoverished landscape of southwest Australia and has several P-saving traits. We identified 16 members of the Phosphate Transporter 1 (PHT1) gene family (HpPHT1;1-HpPHT1;12d) in a long-read genome assembly of H. prostrata. Based on phylogenetics, sequence structure and expression patterns, we classified HpPHT1;1 as potentially involved in Pi uptake from soil and HpPHT1;8 and HpPHT1;9 as potentially involved in Pi uptake and root-to-shoot translocation. Three genes, HpPHT1;4, HpPHT1;6 and HpPHT1;8, lacked regulatory PHR1-binding sites (P1BS) in the promoter regions. Available expression data for HpPHT1;6 and HpPHT1;8 indicated they are not responsive to changes in P supply, potentially contributing to the high P sensitivity of H. prostrata. We also discovered a Proteaceae-specific clade of closely-spaced PHT1 genes that lacked conserved genetic architecture among genera, indicating an evolutionary hot spot within the genome. Overall, the genome assembly of H. prostrata provides a much-needed foundation for understanding the genetic mechanisms of novel adaptations to low P soils in southwest Australian plants.
Collapse
Affiliation(s)
- Benjamin J Nestor
- School of Biological Sciences, University of Western Australia, Perth, Western Australia, Australia
- Centre for Applied Bioinformatics, School of Biological Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Toby Bird
- School of Biological Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Anita A Severn-Ellis
- School of Biological Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Philipp E Bayer
- School of Biological Sciences, University of Western Australia, Perth, Western Australia, Australia
- Centre for Applied Bioinformatics, School of Biological Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Kosala Ranathunge
- School of Biological Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - M Asaduzzaman Prodhan
- School of Biological Sciences, University of Western Australia, Perth, Western Australia, Australia
- Centre for Applied Bioinformatics, School of Biological Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Maheshi Dassanayake
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Jacqueline Batley
- School of Biological Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - David Edwards
- School of Biological Sciences, University of Western Australia, Perth, Western Australia, Australia
- Centre for Applied Bioinformatics, School of Biological Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Hans Lambers
- School of Biological Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Patrick M Finnegan
- School of Biological Sciences, University of Western Australia, Perth, Western Australia, Australia
- Centre for Applied Bioinformatics, School of Biological Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
2
|
Çöl B, Kürkçü MS, Di Bek E. Genome-Wide Screens Identify Genes Responsible for Intrinsic Boric Acid Resistance in Escherichia coli. Biol Trace Elem Res 2024; 202:5771-5793. [PMID: 38466471 PMCID: PMC11502571 DOI: 10.1007/s12011-024-04129-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 02/24/2024] [Indexed: 03/13/2024]
Abstract
Boric acid (BA) has antimicrobial properties and is used to combat bacterial infections, including Enterobacteria. However, the molecular mechanisms and cellular responses to BA are still unknown. This genomics study aims to provide new information on the genes and molecular mechanisms related to the antimicrobial effect of BA in Escherichia coli. The Keio collection of E. coli was used to screen 3985 single-gene knockout strains in order to identify mutant strains that were sensitive or hypersensitive to BA at certain concentrations. The mutant strains were exposed to different concentrations of BA ranging from 0 to 120 mM in LB media. Through genome-wide screens, 92 mutants were identified that were relatively sensitive to BA at least at one concentration tested. The related biological processes in the particular cellular system were listed. This study demonstrates that intrinsic BA resistance is the result of various mechanisms acting together. Additionally, we identified eighteen out of ninety-two mutant strains (Delta_aceF, aroK, cheZ, dinJ, galS, garP, glxK, nohA, talB, torR, trmU, trpR, yddE, yfeS, ygaV, ylaC, yoaC, yohN) that exhibited sensitivity using other methods. To increase sensitivity to BA, we constructed double and triple knockout mutants of the selected sensitive mutants. In certain instances, engineered double and triple mutants exhibited significantly amplified effects. Overall, our analysis of these findings offers further understanding of the mechanisms behind BA toxicity and intrinsic resistance in E. coli.
Collapse
Affiliation(s)
- Bekir Çöl
- Faculty of Science, Department of Biology, Mugla Sitki Kocman University, Mugla, Turkey.
- Research Laboratories Center, Biotechnology Research Center, Mugla Sitki Kocman University, Mugla, Turkey.
| | - Merve Sezer Kürkçü
- Research Laboratories Center, Biotechnology Research Center, Mugla Sitki Kocman University, Mugla, Turkey
- Research and Application Center For Research Laboratories, Mugla Sitki Kocman University, Mugla, Turkey
| | - Esra Di Bek
- Research Laboratories Center, Biotechnology Research Center, Mugla Sitki Kocman University, Mugla, Turkey
- Köyceğiz Vocational School of Health Services, Department of Pharmacy Services, Mugla Sitki Kocman University, Mugla, Turkey
| |
Collapse
|
3
|
He S, Taher NM, Simard AR, Hvorecny KL, Ragusa MJ, Bahl CD, Hickman AB, Dyda F, Madden DR. Molecular basis for the transcriptional regulation of an epoxide-based virulence circuit in Pseudomonas aeruginosa. Nucleic Acids Res 2024; 52:12727-12747. [PMID: 39413156 DOI: 10.1093/nar/gkae889] [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: 01/15/2024] [Revised: 08/30/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
The opportunistic pathogen Pseudomonas aeruginosa infects the airways of people with cystic fibrosis (CF) and produces a virulence factor Cif that is associated with worse outcomes. Cif is an epoxide hydrolase that reduces cell-surface abundance of the cystic fibrosis transmembrane conductance regulator (CFTR) and sabotages pro-resolving signals. Its expression is regulated by a divergently transcribed TetR family transcriptional repressor. CifR represents the first reported epoxide-sensing bacterial transcriptional regulator, but neither its interaction with cognate operator sequences nor the mechanism of activation has been investigated. Using biochemical and structural approaches, we uncovered the molecular mechanisms controlling this complex virulence operon. We present here the first molecular structures of CifR alone and in complex with operator DNA, resolved in a single crystal lattice. Significant conformational changes between these two structures suggest how CifR regulates the expression of the virulence gene cif. Interactions between the N-terminal extension of CifR with the DNA minor groove of the operator play a significant role in the operator recognition of CifR. We also determined that cysteine residue Cys107 is critical for epoxide sensing and DNA release. These results offer new insights into the stereochemical regulation of an epoxide-based virulence circuit in a critically important clinical pathogen.
Collapse
Affiliation(s)
- Susu He
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Noor M Taher
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Adam R Simard
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Kelli L Hvorecny
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Michael J Ragusa
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
- Department of Chemistry, Dartmouth, Hanover, NH 03755, USA
| | - Christopher D Bahl
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Alison B Hickman
- Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fred Dyda
- Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dean R Madden
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
- Department of Chemistry, Dartmouth, Hanover, NH 03755, USA
| |
Collapse
|
4
|
Fakhimi N, Grossman AR. Photosynthetic Electron Flows and Networks of Metabolite Trafficking to Sustain Metabolism in Photosynthetic Systems. PLANTS (BASEL, SWITZERLAND) 2024; 13:3015. [PMID: 39519934 PMCID: PMC11548211 DOI: 10.3390/plants13213015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/03/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Photosynthetic eukaryotes have metabolic pathways that occur in distinct subcellular compartments. However, because metabolites synthesized in one compartment, including fixed carbon compounds and reductant generated by photosynthetic electron flows, may be integral to processes in other compartments, the cells must efficiently move metabolites among the different compartments. This review examines the various photosynthetic electron flows used to generate ATP and fixed carbon and the trafficking of metabolites in the green alga Chlamydomomas reinhardtii; information on other algae and plants is provided to add depth and nuance to the discussion. We emphasized the trafficking of metabolites across the envelope membranes of the two energy powerhouse organelles of the cell, the chloroplast and mitochondrion, the nature and roles of the major mobile metabolites that move among these compartments, and the specific or presumed transporters involved in that trafficking. These transporters include sugar-phosphate (sugar-P)/inorganic phosphate (Pi) transporters and dicarboxylate transporters, although, in many cases, we know little about the substrate specificities of these transporters, how their activities are regulated/coordinated, compensatory responses among transporters when specific transporters are compromised, associations between transporters and other cellular proteins, and the possibilities for forming specific 'megacomplexes' involving interactions between enzymes of central metabolism with specific transport proteins. Finally, we discuss metabolite trafficking associated with specific biological processes that occur under various environmental conditions to help to maintain the cell's fitness. These processes include C4 metabolism in plants and the carbon concentrating mechanism, photorespiration, and fermentation metabolism in algae.
Collapse
Affiliation(s)
- Neda Fakhimi
- Department of Biosphere Sciences and Engineering, The Carnegie Institution for Science, 260 Panama Street, Stanford, CA 94305, USA;
| | - Arthur R. Grossman
- Department of Biosphere Sciences and Engineering, The Carnegie Institution for Science, 260 Panama Street, Stanford, CA 94305, USA;
- Courtesy Appointment, Department of Biology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
5
|
Nath A, Ghosh S, Bandyopadhyay D. Role of melatonin in mitigation of insulin resistance and ensuing diabetic cardiomyopathy. Life Sci 2024; 355:122993. [PMID: 39154810 DOI: 10.1016/j.lfs.2024.122993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
Addressing insulin resistance or hyperinsulinemia might offer a viable treatment approach to stop the onset of diabetic cardiomyopathy, as these conditions independently predispose to the development of the disease, which is initially characterized by diastolic abnormalities. The development of diabetic cardiomyopathy appears to be driven mainly by insulin resistance or impaired insulin signalling and/or hyperinsulinemia. Oxidative stress, hypertrophy, fibrosis, cardiac diastolic dysfunction, and, ultimately, systolic heart failure are the outcomes of these pathophysiological alterations. Melatonin is a ubiquitous indoleamine, a widely distributed compound secreted mainly by the pineal gland, and serves a variety of purposes in almost every living creature. Melatonin is found to play a leading role by improving myocardial cell metabolism, decreasing vascular endothelial cell death, reversing micro-circulation disorders, reducing myocardial fibrosis, decreasing oxidative and endoplasmic reticulum stress, regulating cell autophagy and apoptosis, and enhancing mitochondrial function. This review highlights a relationship between insulin resistance and associated cardiomyopathy. It explores the potential therapeutic strategies offered by the neurohormone melatonin, an important antioxidant that plays a leading role in maintaining glucose homeostasis by influencing the glucose transporters independently and through its receptors. The vast distribution of melatonin receptors in the body, including beta cells of pancreatic islets, asserts the role of this indole molecule in maintaining glucose homeostasis. Melatonin controls the production of GLUT4 and/or the phosphorylation process of the receptor for insulin and its intracellular substrates, activating the insulin-signalling pathway through its G-protein-coupled membrane receptors.
Collapse
Affiliation(s)
- Anupama Nath
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, University College of Science, Technology and Agriculture, 92 APC Road, Kolkata 700 009, India
| | - Songita Ghosh
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, University College of Science, Technology and Agriculture, 92 APC Road, Kolkata 700 009, India
| | - Debasish Bandyopadhyay
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, University College of Science, Technology and Agriculture, 92 APC Road, Kolkata 700 009, India.
| |
Collapse
|
6
|
Toepfer S, Keniya MV, Lackner M, Monk BC. Azole Combinations and Multi-Targeting Drugs That Synergistically Inhibit Candidozyma auris. J Fungi (Basel) 2024; 10:698. [PMID: 39452650 PMCID: PMC11508803 DOI: 10.3390/jof10100698] [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: 09/03/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024] Open
Abstract
Limited antifungal treatment options and drug resistance require innovative approaches to effectively combat fungal infections. Combination therapy is a promising strategy that addresses these pressing issues by concurrently targeting multiple cellular sites. The drug targets usually selected for combination therapy are from different cellular pathways with the goals of increasing treatment options and reducing development of resistance. However, some circumstances can prevent the implementation of combination therapy in clinical practice. These could include the increased risk of adverse effects, drug interactions, and even the promotion of drug resistance. Furthermore, robust clinical evidence supporting the superiority of combination therapy over monotherapy is limited and underscores the need for further research. Despite these challenges, synergies detected with different antifungal classes, such as the azoles and echinocandins, suggest that treatment strategies can be optimized by better understanding the underlying mechanisms. This review provides an overview of multi-targeting combination strategies with a primary focus on Candidozyma auris infections.
Collapse
Affiliation(s)
- Stephanie Toepfer
- Sir John Walsh Research Institute, University of Otago, Dunedin 9016, New Zealand;
- Institute of Hygiene and Medical Microbiology, Medical University Innsbruck, 6020 Innsbruck, Austria;
| | - Mikhail V. Keniya
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA;
| | - Michaela Lackner
- Institute of Hygiene and Medical Microbiology, Medical University Innsbruck, 6020 Innsbruck, Austria;
| | - Brian C. Monk
- Sir John Walsh Research Institute, University of Otago, Dunedin 9016, New Zealand;
| |
Collapse
|
7
|
Niu W, Vu T, Du G, Bogdanov M, Zheng L. Lysophospholipid remodeling mediated by the LplT and Aas protein complex in the bacterial envelope. J Biol Chem 2024; 300:107704. [PMID: 39173951 PMCID: PMC11416262 DOI: 10.1016/j.jbc.2024.107704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
Lysophospholipid transporter LplT and acyltransferase Aas consist of a lysophospholipid-remodeling system ubiquitously found in gram-negative microorganisms. LplT flips lysophospholipid across the inner membrane which is subsequently acylated by Aas on the cytoplasmic membrane surface. Our previous study showed that the proper functioning of this system is important to protecting Escherichia coli from phospholipase-mediated host attack by maintaining the integrity of the bacterial cell envelope. However, the working mechanism of this system is still unclear. Herein, we report that LplT and Aas form a membrane protein complex in E. coli which allows these two enzymes to cooperate efficiently to move lysophospholipids across the bacterial membrane and catalyze their acylation. The direct interaction of LplT and Aas was demonstrated both in vivo and in vitro with a binding affinity of 2.3 μM. We found that a cytoplasmic loop of LplT adjacent to the exit of the substrate translocation pathway plays an important role in maintaining its interaction with Aas. Aas contains an acyl-acyl carrier protein synthase domain and an acyl-transferase domain. Its interaction with LplT is mediated exclusively by its transferase domain. Mutations within the three loops near the putative catalytic site of the transferase domain, respectively, disrupt its interaction with LplT and lysophospholipid acylation activity. These results support a hypothesis of the functional coupling mechanism, in which LplT directly interacts with the transferase domain of Aas for specific substrate membrane migration, providing synchronization of substrate translocation and biosynthetic events.
Collapse
Affiliation(s)
- Wei Niu
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas, USA
| | - Trung Vu
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas, USA
| | - Guangwei Du
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas, USA
| | - Mikhail Bogdanov
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas, USA
| | - Lei Zheng
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas, USA.
| |
Collapse
|
8
|
Lee SS, Kim S, Jin MS. Cryo-EM structure of the human glucose transporter GLUT7. Biochem Biophys Res Commun 2024; 738:150544. [PMID: 39163817 DOI: 10.1016/j.bbrc.2024.150544] [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: 08/07/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024]
Abstract
GLUT7 is a Class II glucose transporter predominantly expressed at the apical membrane of enterocytes in the small intestine. Here, we report the cryo-EM structure of nanodisc-reconstituted human GLUT7 in the apo state at 3.3 Å resolution. Our atomic model reveals a typical major facilitator superfamily fold, with the substrate-binding site open to the extracellular side of the membrane. Despite the nearly identical conformation to its closest family member, rat GLUT5, our structure unveils distinct features of the substrate-binding cavity that may influence substrate specificity and binding mode. A homology model of the inward-open human GLUT7 indicates that similar to other members of the GLUT family, it may undergo a global rocker-switch-like reorientation of the transmembrane bundles to facilitate substrate translocation across the membrane. Our work enhances the current structural understanding of the GLUT family, and lays a foundation for rational design of regulators of GLUTs and other sugar transporters.
Collapse
Affiliation(s)
- Sang Soo Lee
- School of Life Sciences, GIST, 123 Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Subin Kim
- School of Life Sciences, GIST, 123 Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Mi Sun Jin
- School of Life Sciences, GIST, 123 Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
9
|
Mélida H, Kappel L, Ullah SF, Bulone V, Srivastava V. Quantitative proteomic analysis of plasma membranes from the fish pathogen Saprolegnia parasitica reveals promising targets for disease control. Microbiol Spectr 2024; 12:e0034824. [PMID: 38888349 PMCID: PMC11302233 DOI: 10.1128/spectrum.00348-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/30/2024] [Indexed: 06/20/2024] Open
Abstract
The phylum Oomycota contains economically important pathogens of animals and plants, including Saprolegnia parasitica, the causal agent of the fish disease saprolegniasis. Due to intense fish farming and banning of the most effective control measures, saprolegniasis has re-emerged as a major challenge for the aquaculture industry. Oomycete cells are surrounded by a polysaccharide-rich cell wall matrix that, in addition to being essential for cell growth, also functions as a protective "armor." Consequently, the enzymes responsible for cell wall synthesis provide potential targets for disease control. Oomycete cell wall biosynthetic enzymes are predicted to be plasma membrane proteins. To identify these proteins, we applied a quantitative (iTRAQ) mass spectrometry-based proteomics approach to the plasma membrane of the hyphal cells of S. parasitica, providing the first complete plasma membrane proteome of an oomycete species. Of significance is the identification of 65 proteins enriched in detergent-resistant microdomains (DRMs). In silico analysis showed that DRM-enriched proteins are mainly involved in molecular transport and β-1,3-glucan synthesis, potentially contributing to pathogenesis. Moreover, biochemical characterization of the glycosyltransferase activity in these microdomains further supported their role in β-1,3-glucan synthesis. Altogether, the knowledge gained in this study provides a basis for developing disease control measures targeting specific plasma membrane proteins in S. parasitica.IMPORTANCEThe significance of this research lies in its potential to combat saprolegniasis, a detrimental fish disease, which has resurged due to intensive fish farming and regulatory restrictions. By targeting enzymes responsible for cell wall synthesis in Saprolegnia parasitica, this study uncovers potential avenues for disease control. Particularly noteworthy is the identification of several proteins enriched in membrane microdomains, offering insights into molecular mechanisms potentially involved in pathogenesis. Understanding the role of these proteins provides a foundation for developing targeted disease control measures. Overall, this research holds promise for safeguarding the aquaculture industry against the challenges posed by saprolegniasis.
Collapse
Affiliation(s)
- Hugo Mélida
- Division of Glycoscience, Department of Chemistry, CBH School, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, Sweden
| | - Lisa Kappel
- Division of Glycoscience, Department of Chemistry, CBH School, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, Sweden
| | - Sadia Fida Ullah
- Division of Glycoscience, Department of Chemistry, CBH School, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, Sweden
| | - Vincent Bulone
- Division of Glycoscience, Department of Chemistry, CBH School, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, Sweden
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Vaibhav Srivastava
- Division of Glycoscience, Department of Chemistry, CBH School, KTH Royal Institute of Technology, AlbaNova University Centre, Stockholm, Sweden
| |
Collapse
|
10
|
Toorop MMA, Hoogendijk IV, Dogterom-Ballering HCM, Boers SA, Kraakman MEM, van Prehn J, Wessels E, Bekker V, Veldkamp KE. Implications of ad-hoc molecular typing for infection control measures in a multi-cluster, multi-phenotypic Serratia marcescens outbreak in a neonatal intensive care unit. J Hosp Infect 2024; 150:26-33. [PMID: 38782059 DOI: 10.1016/j.jhin.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Serratia marcescens is known to cause outbreaks in neonatal intensive care units (NICUs). Traditionally epidemiological data, antimicrobial resistance patterns and epidemiological typing have been used to guide infection prevention methods. Whole-genome sequencing (WGS) applications such as core-genome multi-locus sequence typing (cgMLST) applied during an outbreak would potentially yield more information. AIM To use cgMLST to acquire detailed information on the source and spread of bacteria, enabling more efficient control measures during an S. marcescens outbreak at a NICU. METHODS Neonates admitted to the NICU of the Leiden University Medical Center (LUMC) during an outbreak between September 2023 and January 2024, with S. marcescens being cultured, were included. Environmental samples were taken to search for a common source, antibiotic susceptibility testing was performed, and antimicrobial resistance genes were analysed. FINDINGS S. marcescens strains from 17 of the 20 positive patients were available for molecular typing. The cgMLST scheme revealed five different complex types consisting of four separate clusters. Multiple clusters made an unidentified persistent environmental source as cause of the outbreak less likely, leading to a quick downscaling of infection prevention measures. Differences were shown in aminoglycoside resistance patterns of isolates within the same complex types and patients. CONCLUSION The use of ad-hoc cgMLST provided timely data for rational decision-making during an S. marcescens outbreak at the NICU. Antibiotic phenotyping alone was found not to be suitable for studying clonal spread during this outbreak with S. marcescens.
Collapse
Affiliation(s)
- M M A Toorop
- Leiden University Center for Infectious Diseases (LU-CID) Medical Microbiology and Infection Control, Leiden University Medical Center, Leiden, The Netherlands
| | - I V Hoogendijk
- Leiden University Center for Infectious Diseases (LU-CID) Medical Microbiology and Infection Control, Leiden University Medical Center, Leiden, The Netherlands
| | - H C M Dogterom-Ballering
- Leiden University Center for Infectious Diseases (LU-CID) Medical Microbiology and Infection Control, Leiden University Medical Center, Leiden, The Netherlands
| | - S A Boers
- Leiden University Center for Infectious Diseases (LU-CID) Medical Microbiology and Infection Control, Leiden University Medical Center, Leiden, The Netherlands
| | - M E M Kraakman
- Leiden University Center for Infectious Diseases (LU-CID) Medical Microbiology and Infection Control, Leiden University Medical Center, Leiden, The Netherlands
| | - J van Prehn
- Leiden University Center for Infectious Diseases (LU-CID) Medical Microbiology and Infection Control, Leiden University Medical Center, Leiden, The Netherlands
| | - E Wessels
- Leiden University Center for Infectious Diseases (LU-CID) Medical Microbiology and Infection Control, Leiden University Medical Center, Leiden, The Netherlands
| | - V Bekker
- Division of Neonatology, Department of Pediatrics, Willem Alexander Children's Hospital - Leiden University Medical Center, Leiden, The Netherlands
| | - K E Veldkamp
- Leiden University Center for Infectious Diseases (LU-CID) Medical Microbiology and Infection Control, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
11
|
Crandall JG, Zhou X, Rokas A, Hittinger CT. Specialization restricts the evolutionary paths available to yeast sugar transporters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.604696. [PMID: 39091816 PMCID: PMC11291069 DOI: 10.1101/2024.07.22.604696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Functional innovation at the protein level is a key source of evolutionary novelties. The constraints on functional innovations are likely to be highly specific in different proteins, which are shaped by their unique histories and the extent of global epistasis that arises from their structures and biochemistries. These contextual nuances in the sequence-function relationship have implications both for a basic understanding of the evolutionary process and for engineering proteins with desirable properties. Here, we have investigated the molecular basis of novel function in a model member of an ancient, conserved, and biotechnologically relevant protein family. These Major Facilitator Superfamily sugar porters are a functionally diverse group of proteins that are thought to be highly plastic and evolvable. By dissecting a recent evolutionary innovation in an α-glucoside transporter from the yeast Saccharomyces eubayanus, we show that the ability to transport a novel substrate requires high-order interactions between many protein regions and numerous specific residues proximal to the transport channel. To reconcile the functional diversity of this family with the constrained evolution of this model protein, we generated new, state-of-the-art genome annotations for 332 Saccharomycotina yeast species spanning approximately 400 million years of evolution. By integrating phylogenetic and phenotypic analyses across these species, we show that the model yeast α-glucoside transporters likely evolved from a multifunctional ancestor and became subfunctionalized. The accumulation of additive and epistatic substitutions likely entrenched this subfunction, which made the simultaneous acquisition of multiple interacting substitutions the only reasonably accessible path to novelty.
Collapse
Affiliation(s)
- Johnathan G. Crandall
- Laboratory of Genetics, J. F. Crow Institute for the Study of Evolution, Center for Genomic Science Innovation, DOE Great Lakes Bioenergy Research Center, Wisconsin Energy Institute, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Xiaofan Zhou
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Center, South China Agricultural University, Guangzhou 510642, China
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN 37235, USA
| | - Antonis Rokas
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN 37235, USA
| | - Chris Todd Hittinger
- Laboratory of Genetics, J. F. Crow Institute for the Study of Evolution, Center for Genomic Science Innovation, DOE Great Lakes Bioenergy Research Center, Wisconsin Energy Institute, University of Wisconsin-Madison, Madison, WI 53726, USA
| |
Collapse
|
12
|
Debbiche R, Elbahnsi A, Uguen K, Ka C, Callebaut I, Le Gac G. Insights into the role of glycerophospholipids on the iron export function of SLC40A1 and the molecular mechanisms of ferroportin disease. FASEB J 2024; 38:e23725. [PMID: 38959016 DOI: 10.1096/fj.202400337r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 07/04/2024]
Abstract
SLC40A1 is the sole iron export protein reported in mammals. In humans, its dysfunction is responsible for ferroportin disease, an inborn error of iron metabolism transmitted as an autosomal dominant trait and observed in different ethnic groups. As a member of the major facilitator superfamily, SLC40A1 requires a series of conformational changes to enable iron translocation across the plasma membrane. The influence of lipids on protein stability and its conformational changes has been little investigated to date. Here, we combine molecular dynamics simulations of SLC40A1 embedded in membrane bilayers with experimental alanine scanning mutagenesis to analyze the specific role of glycerophospholipids. We identify four basic residues (Lys90, Arg365, Lys366, and Arg371) that are located at the membrane-cytosol interface and consistently interact with 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine (POPE) molecules. These residues surround a network of salt bridges and hydrogens bonds that play a critical role in stabilizing SLC40A1 in its basal outward-facing conformation. More deeply embedded in the plasma membrane, we identify Arg179 as a charged amino acid residue also tightly interacting with lipid polar heads. This results in a local deformation of the lipid bilayer. Interestingly, Arg179 is adjacent to Arg178, which forms a functionally important salt-bridge with Asp473 and is a recurrently associated with ferroportin disease when mutated to glutamine. We demonstrate that the two p.Arg178Gln and p.Arg179Thr missense variants have similar functional behaviors. These observations provide insights into the role of phospholipids in the formation/disruption of the SLC40A1 inner gate, and give a better understanding of the diversity of molecular mechanisms of ferroportin disease.
Collapse
Affiliation(s)
- Rim Debbiche
- University of Brest, Inserm, EFS, UMR 1078, GGB, Brest, France
| | - Ahmad Elbahnsi
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, IRD, Paris, France
| | - Kévin Uguen
- University of Brest, Inserm, EFS, UMR 1078, GGB, Brest, France
- CHU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Brest, France
| | - Chandran Ka
- University of Brest, Inserm, EFS, UMR 1078, GGB, Brest, France
- CHU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Brest, France
| | - Isabelle Callebaut
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, IRD, Paris, France
| | - Gérald Le Gac
- University of Brest, Inserm, EFS, UMR 1078, GGB, Brest, France
- CHU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Brest, France
- Laboratory of Excellence GR-Ex, Paris, France
| |
Collapse
|
13
|
Harder CB, Miyauchi S, Virágh M, Kuo A, Thoen E, Andreopoulos B, Lu D, Skrede I, Drula E, Henrissat B, Morin E, Kohler A, Barry K, LaButti K, Salamov A, Lipzen A, Merényi Z, Hegedüs B, Baldrian P, Stursova M, Weitz H, Taylor A, Koriabine M, Savage E, Grigoriev IV, Nagy LG, Martin F, Kauserud H. Extreme overall mushroom genome expansion in Mycena s.s. irrespective of plant hosts or substrate specializations. CELL GENOMICS 2024; 4:100586. [PMID: 38942024 PMCID: PMC11293592 DOI: 10.1016/j.xgen.2024.100586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 01/28/2024] [Accepted: 05/28/2024] [Indexed: 06/30/2024]
Abstract
Mycena s.s. is a ubiquitous mushroom genus whose members degrade multiple dead plant substrates and opportunistically invade living plant roots. Having sequenced the nuclear genomes of 24 Mycena species, we find them to defy the expected patterns for fungi based on both their traditionally perceived saprotrophic ecology and substrate specializations. Mycena displayed massive genome expansions overall affecting all gene families, driven by novel gene family emergence, gene duplications, enlarged secretomes encoding polysaccharide degradation enzymes, transposable element (TE) proliferation, and horizontal gene transfers. Mainly due to TE proliferation, Arctic Mycena species display genomes of up to 502 Mbp (2-8× the temperate Mycena), the largest among mushroom-forming Agaricomycetes, indicating a possible evolutionary convergence to genomic expansions sometimes seen in Arctic plants. Overall, Mycena show highly unusual, varied mosaic-like genomic structures adaptable to multiple lifestyles, providing genomic illustration for the growing realization that fungal niche adaptations can be far more fluid than traditionally believed.
Collapse
Affiliation(s)
- Christoffer Bugge Harder
- Department of Biosciences, University of Oslo, Box 1066 Blindern, 0316 Oslo, Norway; Department of Biology, Microbial Ecology Group, Biology Department, Lund University, Lund, Sweden; University of Copenhagen, Department of Biology, Section of Terrestrial Ecology, 2100 Copenhagen Ø, Denmark.
| | - Shingo Miyauchi
- Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0495, Japan; Université de Lorraine, INRAE, UMR Interactions Arbres/Microorganismes, Centre INRAE Grand Est Nancy, 54280 Champenoux, France
| | - Máté Virágh
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, HUN-REN Szeged, 6726 Szeged, Hungary
| | - Alan Kuo
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Ella Thoen
- Department of Biosciences, University of Oslo, Box 1066 Blindern, 0316 Oslo, Norway
| | - Bill Andreopoulos
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Dabao Lu
- Department of Biosciences, University of Oslo, Box 1066 Blindern, 0316 Oslo, Norway
| | - Inger Skrede
- Department of Biosciences, University of Oslo, Box 1066 Blindern, 0316 Oslo, Norway
| | - Elodie Drula
- Architecture et Fonction des Macromolécules Biologiques (AFMB), CNRS, Aix Marseille Université, 163 avenue de Luminy, 13288 Marseille, France; INRAE, UMR 1163, Biodiversité et Biotechnologie Fongiques, 13009 Marseille, France
| | - Bernard Henrissat
- Architecture et Fonction des Macromolécules Biologiques (AFMB), CNRS, Aix Marseille Université, 163 avenue de Luminy, 13288 Marseille, France
| | - Emmanuelle Morin
- Université de Lorraine, INRAE, UMR Interactions Arbres/Microorganismes, Centre INRAE Grand Est Nancy, 54280 Champenoux, France
| | - Annegret Kohler
- Université de Lorraine, INRAE, UMR Interactions Arbres/Microorganismes, Centre INRAE Grand Est Nancy, 54280 Champenoux, France
| | - Kerrie Barry
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Kurt LaButti
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Asaf Salamov
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Anna Lipzen
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Zsolt Merényi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, HUN-REN Szeged, 6726 Szeged, Hungary
| | - Botond Hegedüs
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, HUN-REN Szeged, 6726 Szeged, Hungary
| | - Petr Baldrian
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague 4, Czech Republic
| | - Martina Stursova
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague 4, Czech Republic
| | - Hedda Weitz
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Andy Taylor
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK; The James Hutton Institute, Aberdeen, UK
| | - Maxim Koriabine
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Emily Savage
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Igor V Grigoriev
- U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Department of Plant and Microbial Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - László G Nagy
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, HUN-REN Szeged, 6726 Szeged, Hungary
| | - Francis Martin
- Université de Lorraine, INRAE, UMR Interactions Arbres/Microorganismes, Centre INRAE Grand Est Nancy, 54280 Champenoux, France.
| | - Håvard Kauserud
- Department of Biosciences, University of Oslo, Box 1066 Blindern, 0316 Oslo, Norway
| |
Collapse
|
14
|
Kim SH, Jang HW, Park JJ, Nam DG, Lee SJ, Yeo SH, Kim SY. Antibiotic Resistance in Acetic Acid Bacteria Originating from Vinegar. Antibiotics (Basel) 2024; 13:626. [PMID: 39061308 PMCID: PMC11274321 DOI: 10.3390/antibiotics13070626] [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: 05/30/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Acetic acid bacteria (AAB) are major contributors to the production of fermented vinegar, offering various cultural, culinary, and health benefits. Although the residual unpasteurized AAB after vinegar production are not pathogens, these are necessary and require safety evaluations, including antibiotic resistance, before use as a starter. In this research, we investigated the antibiotic resistance profiles of 26 AAB strains, including various species of Komagataeibacter and Acetobacter, against 10 different antibiotics using the E-test method. All strains exhibited resistance to aztreonam and clindamycin. Komagataeibacter species demonstrated a 50% resistance rate to ciprofloxacin, analogous to Acetobacter species, but showed twice the resistance rates to chloramphenicol and erythromycin. Genomic analysis of K. saccharivorans CV1 identified intrinsic resistance mechanisms, such as multidrug efflux pumps, thereby enhancing our understanding of antibiotic resistance in acetic acid-producing bacteria. These findings enhance understanding of antibiotic resistance in AAB for food safety and new antimicrobial strategies, suggesting the need for standardized testing methods and molecular genetic study.
Collapse
Affiliation(s)
- Sun-Hee Kim
- Fermented and Processed Food Science Division, Department of Agrofood Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Republic of Korea; (S.-H.K.); (H.-W.J.); (J.-J.P.); (S.-J.L.)
| | - Hyun-Wook Jang
- Fermented and Processed Food Science Division, Department of Agrofood Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Republic of Korea; (S.-H.K.); (H.-W.J.); (J.-J.P.); (S.-J.L.)
| | - Jin-Ju Park
- Fermented and Processed Food Science Division, Department of Agrofood Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Republic of Korea; (S.-H.K.); (H.-W.J.); (J.-J.P.); (S.-J.L.)
| | - Dong-Geon Nam
- Division of Functional Food & Nutrition, Department of Agrofood Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Republic of Korea;
| | - Su-Jeong Lee
- Fermented and Processed Food Science Division, Department of Agrofood Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Republic of Korea; (S.-H.K.); (H.-W.J.); (J.-J.P.); (S.-J.L.)
| | - Soo-Hwan Yeo
- Fermented and Processed Food Science Division, Department of Agrofood Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Republic of Korea; (S.-H.K.); (H.-W.J.); (J.-J.P.); (S.-J.L.)
| | - So-Young Kim
- Fermented and Processed Food Science Division, Department of Agrofood Resources, National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Republic of Korea; (S.-H.K.); (H.-W.J.); (J.-J.P.); (S.-J.L.)
| |
Collapse
|
15
|
Hammes UZ, Pedersen BP. Structure and Function of Auxin Transporters. ANNUAL REVIEW OF PLANT BIOLOGY 2024; 75:185-209. [PMID: 38211951 DOI: 10.1146/annurev-arplant-070523-034109] [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: 01/13/2024]
Abstract
Auxins, a group of central hormones in plant growth and development, are transported by a diverse range of transporters with distinct biochemical and structural properties. This review summarizes the current knowledge on all known auxin transporters with respect to their biochemical and biophysical properties and the methods used to characterize them. In particular, we focus on the recent advances that were made concerning the PIN-FORMED family of auxin exporters. Insights derived from solving their structures have improved our understanding of the auxin export process, and we discuss the current state of the art on PIN-mediated auxin transport, including the use of biophysical methods to examine their properties. Understanding the mechanisms of auxin transport is crucial for understanding plant growth and development, as well as for the development of more effective strategies for crop production and plant biotechnology.
Collapse
Affiliation(s)
- Ulrich Z Hammes
- School of Life Sciences, Plant Systems Biology, Technical University of Munich, Freising, Germany;
| | | |
Collapse
|
16
|
Zhao W, Hu X, Chen Z, Li X. Major Facilitator Superfamily Domain Containing 12 Is Overexpressed in Lung Cancer and Exhibits an Oncogenic Role in Lung Adenocarcinoma Cells. DNA Cell Biol 2024; 43:331-340. [PMID: 38687351 DOI: 10.1089/dna.2023.0378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
Major facilitator superfamily domain containing 12 (MFSD12) regulates lysosomal cysteine import and promotes the proliferation and survival of melanoma cells. However, the expression and function of MFSD12 in other cancers, particularly in lung cancer, remain unclear. The expression of MFSD12 across various types of cancers and corresponding control tissues was examined using TIMER. MFSD12 expression in lung adenocarcinoma (LUAD) and its correlation with distinct clinicopathological features of LUAD patients were analyzed with UALCAN. The correlation between MFSD12 expression and survival of LUAD patients was assessed using the R package, survival, and the relationship between MFSD12 expression and immune infiltration status in LUAD was investigated using CIBERSORT. In addition, MFSD12 expression was knocked down in PC9 LUAD cells and their proliferation, capacity for expansion, cell cycle, apoptosis, and migration/invasion were evaluated through CCK-8 assays, colony formation assays, 7-AAD staining, Annexin V/PI staining, and Transwell assays, respectively. The stemness of these PC9 cells was determined through Western blotting, flow cytometry, and tumor sphere formation assays. MFSD12 mRNA levels were significantly elevated in multiple types of cancers, including LUAD. MFSD12 expression was also positively correlated with cancer stage, nodal metastasis, and infiltration of various immune cells in LUAD, and high MFSD12 levels predicted poor survival among LUAD patients. Knockdown of MFSD12 in PC9 cells resulted in decreased proliferation, attenuated colony formation capacity, cell cycle arrest, elevated apoptosis, impaired migration/invasion, and reduced stemness in PC9 cells. MFSD12 is an oncogene in LUAD.
Collapse
Affiliation(s)
- Weijun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Xilin Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Zixuan Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Xinjian Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
17
|
Jungnickel KEJ, Guelle O, Iguchi M, Dong W, Kotov V, Gabriel F, Debacker C, Dairou J, McCort-Tranchepain I, Laqtom NN, Chan SH, Ejima A, Sato K, Massa López D, Saftig P, Mehdipour AR, Abu-Remaileh M, Gasnier B, Löw C, Damme M. MFSD1 with its accessory subunit GLMP functions as a general dipeptide uniporter in lysosomes. Nat Cell Biol 2024; 26:1047-1061. [PMID: 38839979 PMCID: PMC11252000 DOI: 10.1038/s41556-024-01436-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024]
Abstract
The lysosomal degradation of macromolecules produces diverse small metabolites exported by specific transporters for reuse in biosynthetic pathways. Here we deorphanized the major facilitator superfamily domain containing 1 (MFSD1) protein, which forms a tight complex with the glycosylated lysosomal membrane protein (GLMP) in the lysosomal membrane. Untargeted metabolomics analysis of MFSD1-deficient mouse lysosomes revealed an increase in cationic dipeptides. Purified MFSD1 selectively bound diverse dipeptides, while electrophysiological, isotope tracer and fluorescence-based studies in Xenopus oocytes and proteoliposomes showed that MFSD1-GLMP acts as a uniporter for cationic, neutral and anionic dipeptides. Cryoelectron microscopy structure of the dipeptide-bound MFSD1-GLMP complex in outward-open conformation characterized the heterodimer interface and, in combination with molecular dynamics simulations, provided a structural basis for its selectivity towards diverse dipeptides. Together, our data identify MFSD1 as a general lysosomal dipeptide uniporter, providing an alternative route to recycle lysosomal proteolysis products when lysosomal amino acid exporters are overloaded.
Collapse
Affiliation(s)
| | - Océane Guelle
- Saints-Pères Paris Institute for the Neurosciences, Université Paris Cité, Centre National de la Recherche Scientifique, Paris, France
| | - Miharu Iguchi
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health, Stanford University, Stanford, CA, USA
| | - Wentao Dong
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health, Stanford University, Stanford, CA, USA
| | - Vadim Kotov
- Centre for Structural Systems Biology, Hamburg, Germany
- European Molecular Biology Laboratory Hamburg, Hamburg, Germany
| | - Florian Gabriel
- Centre for Structural Systems Biology, Hamburg, Germany
- European Molecular Biology Laboratory Hamburg, Hamburg, Germany
| | - Cécile Debacker
- Saints-Pères Paris Institute for the Neurosciences, Université Paris Cité, Centre National de la Recherche Scientifique, Paris, France
| | - Julien Dairou
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Cité, Paris, France
| | - Isabelle McCort-Tranchepain
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR 8601, Université Paris Cité, Paris, France
| | - Nouf N Laqtom
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health, Stanford University, Stanford, CA, USA
| | - Sze Ham Chan
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Akika Ejima
- Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Kenji Sato
- Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - David Massa López
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | | | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health, Stanford University, Stanford, CA, USA
| | - Bruno Gasnier
- Saints-Pères Paris Institute for the Neurosciences, Université Paris Cité, Centre National de la Recherche Scientifique, Paris, France.
| | - Christian Löw
- Centre for Structural Systems Biology, Hamburg, Germany.
- European Molecular Biology Laboratory Hamburg, Hamburg, Germany.
| | - Markus Damme
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany.
| |
Collapse
|
18
|
Shen Z, Xu L, Wu T, Wang H, Wang Q, Ge X, Kong F, Huang G, Pan X. Structural basis for urate recognition and apigenin inhibition of human GLUT9. Nat Commun 2024; 15:5039. [PMID: 38866775 PMCID: PMC11169512 DOI: 10.1038/s41467-024-49420-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024] Open
Abstract
Urate, the physiological form of uric acid and a potent antioxidant in serum, plays a pivotal role in scavenging reactive oxygen species. Yet excessive accumulation of urate, known as hyperuricemia, is the primary risk factor for the development of gout. The high-capacity urate transporter GLUT9 represents a promising target for gout treatment. Here, we present cryo-electron microscopy structures of human GLUT9 in complex with urate or its inhibitor apigenin at overall resolutions of 3.5 Å and 3.3 Å, respectively. In both structures, GLUT9 exhibits an inward open conformation, wherein the substrate binding pocket faces the intracellular side. These structures unveil the molecular basis for GLUT9's substrate preference of urate over glucose, and show that apigenin acts as a competitive inhibitor by occupying the substrate binding site. Our findings provide critical information for the development of specific inhibitors targeting GLUT9 as potential therapeutics for gout and hyperuricemia.
Collapse
Affiliation(s)
- Zilin Shen
- Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Li Xu
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, 518107, Guangdong, China
| | - Tong Wu
- Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Huan Wang
- Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Qifan Wang
- Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Xiaofei Ge
- Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Fang Kong
- Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Gaoxingyu Huang
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, 310024, Zhejiang, China
| | - Xiaojing Pan
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
19
|
Lekshmi M, Ortiz-Alegria A, Kumar S, Varela MF. Major facilitator superfamily efflux pumps in human pathogens: Role in multidrug resistance and beyond. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100248. [PMID: 38974671 PMCID: PMC11225705 DOI: 10.1016/j.crmicr.2024.100248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024] Open
Abstract
The major facilitator superfamily (MFS) of proteins constitutes a large group of related solute transporters found across all known living taxa of organisms. The transporters of the MFS contain an extremely diverse array of substrates, including ions, molecules of intermediary metabolism, and structurally different antimicrobial agents. First discovered over 30 years ago, the MFS represents an important collection of integral membrane transporters. Bacterial microorganisms expressing multidrug efflux pumps belonging to the MFS are considered serious pathogens, accounting for alarming morbidity and mortality numbers annually. This review article considers recent advances in the structure-function relationships, the transport mechanism, and modulation of MFS multidrug efflux pumps within the context of drug resistance mechanisms of bacterial pathogens of public health concerns.
Collapse
Affiliation(s)
- Manjusha Lekshmi
- QC Laboratory, Post Harvest Technology, ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India
| | - Anely Ortiz-Alegria
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, United States
| | - Sanath Kumar
- QC Laboratory, Post Harvest Technology, ICAR-Central Institute of Fisheries Education (CIFE), Mumbai 400061, India
| | - Manuel F. Varela
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, United States
| |
Collapse
|
20
|
Lu L, Delrot S, Liang Z. From acidity to sweetness: a comprehensive review of carbon accumulation in grape berries. MOLECULAR HORTICULTURE 2024; 4:22. [PMID: 38835095 DOI: 10.1186/s43897-024-00100-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024]
Abstract
Most of the carbon found in fruits at harvest is imported by the phloem. Imported carbon provide the material needed for the accumulation of sugars, organic acids, secondary compounds, in addition to the material needed for the synthesis of cell walls. The accumulation of sugars during fruit development influences not only sweetness but also various parameters controlling fruit composition (fruit "quality"). The accumulation of organic acids and sugar in grape berry flesh cells is a key process for berry development and ripening. The present review presents an update of the research on grape berry development, anatomical structure, sugar and acid metabolism, sugar transporters, and regulatory factors.
Collapse
Affiliation(s)
- Lizhen Lu
- State Key Laboratory of Plant Diversity and Prominent Crop, Beijing Key Laboratory of Grape Science and Oenology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China
- China National Botanical Garden, Beijing, 100093, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Serge Delrot
- Bordeaux University, Bordeaux Sciences Agro, INRAE, UMR EGFV, ISVV, Villenave d'Ornon, 33882, France
| | - Zhenchang Liang
- State Key Laboratory of Plant Diversity and Prominent Crop, Beijing Key Laboratory of Grape Science and Oenology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China.
- China National Botanical Garden, Beijing, 100093, China.
| |
Collapse
|
21
|
Chen Z, Yu S, Liu J, Guo L, Wu T, Duan P, Yan D, Huang C, Huo Y. Concentration Recognition-Based Auto-Dynamic Regulation System (CRUISE) Enabling Efficient Production of Higher Alcohols. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310215. [PMID: 38626358 PMCID: PMC11187965 DOI: 10.1002/advs.202310215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/12/2024] [Indexed: 04/18/2024]
Abstract
Microbial factories lacking the ability of dynamically regulating the pathway enzymes overexpression, according to in situ metabolite concentrations, are suboptimal, especially when the metabolic intermediates are competed by growth and chemical production. The production of higher alcohols (HAs), which hijacks the amino acids (AAs) from protein biosynthesis, minimizes the intracellular concentration of AAs and thus inhibits the host growth. To balance the resource allocation and maintain stable AA flux, this work utilizes AA-responsive transcriptional attenuator ivbL and HA-responsive transcriptional activator BmoR to establish a concentration recognition-based auto-dynamic regulation system (CRUISE). This system ultimately maintains the intracellular homeostasis of AA and maximizes the production of HA. It is demonstrated that ivbL-driven enzymes overexpression can dynamically regulate the AA-to-HA conversion while BmoR-driven enzymes overexpression can accelerate the AA biosynthesis during the HA production in a feedback activation mode. The AA flux in biosynthesis and conversion pathways is balanced via the intracellular AA concentration, which is vice versa stabilized by the competition between AA biosynthesis and conversion. The CRUISE, further aided by scaffold-based self-assembly, enables 40.4 g L-1 of isobutanol production in a bioreactor. Taken together, CRUISE realizes robust HA production and sheds new light on the dynamic flux control during the process of chemical production.
Collapse
Affiliation(s)
- Zhenya Chen
- Key Laboratory of Molecular Medicine and BiotherapyAerospace Center HospitalSchool of Life ScienceBeijing Institute of TechnologyHaidian DistrictNo. 5 South Zhongguancun StreetBeijing100081China
- Tangshan Research InstituteBeijing Institute of Technology, No. 57, South Jianshe Road, Lubei DistrictTangshanHebei063000China
| | - Shengzhu Yu
- Key Laboratory of Molecular Medicine and BiotherapyAerospace Center HospitalSchool of Life ScienceBeijing Institute of TechnologyHaidian DistrictNo. 5 South Zhongguancun StreetBeijing100081China
| | - Jing Liu
- Key Laboratory of Molecular Medicine and BiotherapyAerospace Center HospitalSchool of Life ScienceBeijing Institute of TechnologyHaidian DistrictNo. 5 South Zhongguancun StreetBeijing100081China
| | - Liwei Guo
- Key Laboratory of Molecular Medicine and BiotherapyAerospace Center HospitalSchool of Life ScienceBeijing Institute of TechnologyHaidian DistrictNo. 5 South Zhongguancun StreetBeijing100081China
| | - Tong Wu
- Key Laboratory of Molecular Medicine and BiotherapyAerospace Center HospitalSchool of Life ScienceBeijing Institute of TechnologyHaidian DistrictNo. 5 South Zhongguancun StreetBeijing100081China
| | - Peifeng Duan
- Key Laboratory of Molecular Medicine and BiotherapyAerospace Center HospitalSchool of Life ScienceBeijing Institute of TechnologyHaidian DistrictNo. 5 South Zhongguancun StreetBeijing100081China
| | - Dongli Yan
- Key Laboratory of Molecular Medicine and BiotherapyAerospace Center HospitalSchool of Life ScienceBeijing Institute of TechnologyHaidian DistrictNo. 5 South Zhongguancun StreetBeijing100081China
| | - Chaoyong Huang
- Key Laboratory of Molecular Medicine and BiotherapyAerospace Center HospitalSchool of Life ScienceBeijing Institute of TechnologyHaidian DistrictNo. 5 South Zhongguancun StreetBeijing100081China
| | - Yi‐Xin Huo
- Key Laboratory of Molecular Medicine and BiotherapyAerospace Center HospitalSchool of Life ScienceBeijing Institute of TechnologyHaidian DistrictNo. 5 South Zhongguancun StreetBeijing100081China
- Tangshan Research InstituteBeijing Institute of Technology, No. 57, South Jianshe Road, Lubei DistrictTangshanHebei063000China
| |
Collapse
|
22
|
Walter-Nuno AB, Taracena-Agarwal M, Oliveira MP, Oliveira MF, Oliveira PL, Paiva-Silva GO. Export of heme by the feline leukemia virus C receptor regulates mitochondrial biogenesis and redox balance in the hematophagous insect Rhodnius prolixus. FASEB J 2024; 38:e23691. [PMID: 38780525 DOI: 10.1096/fj.202301671rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 04/25/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024]
Abstract
Heme is a prosthetic group of proteins involved in vital physiological processes. It participates, for example, in redox reactions crucial for cell metabolism due to the variable oxidation state of its central iron atom. However, excessive heme can be cytotoxic due to its prooxidant properties. Therefore, the control of intracellular heme levels ensures the survival of organisms, especially those that deal with high concentrations of heme during their lives, such as hematophagous insects. The export of heme initially attributed to the feline leukemia virus C receptor (FLVCR) has recently been called into question, following the discovery of choline uptake by the same receptor in mammals. Here, we found that RpFLVCR is a heme exporter in the midgut of the hematophagous insect Rhodnius prolixus, a vector for Chagas disease. Silencing RpFLVCR decreased hemolymphatic heme levels and increased the levels of intracellular dicysteinyl-biliverdin, indicating heme retention inside midgut cells. FLVCR silencing led to increased expression of heme oxygenase (HO), ferritin, and mitoferrin mRNAs while downregulating the iron importers Malvolio 1 and 2. In contrast, HO gene silencing increased FLVCR and Malvolio expression and downregulated ferritin, revealing crosstalk between heme degradation/export and iron transport/storage pathways. Furthermore, RpFLVCR silencing strongly increased oxidant production and lipid peroxidation, reduced cytochrome c oxidase activity, and activated mitochondrial biogenesis, effects not observed in RpHO-silenced insects. These data support FLVCR function as a heme exporter, playing a pivotal role in heme/iron metabolism and maintenance of redox balance, especially in an organism adapted to face extremely high concentrations of heme.
Collapse
Affiliation(s)
- Ana Beatriz Walter-Nuno
- Instituto de Bioquimica Medica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Mabel Taracena-Agarwal
- Instituto de Bioquimica Medica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Matheus P Oliveira
- Instituto de Bioquimica Medica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcus F Oliveira
- Instituto de Bioquimica Medica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Pedro L Oliveira
- Instituto de Bioquimica Medica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Gabriela O Paiva-Silva
- Instituto de Bioquimica Medica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| |
Collapse
|
23
|
Khramov DE, Rostovtseva EI, Matalin DA, Konoshenkova AO, Nedelyaeva OI, Volkov VS, Balnokin YV, Popova LG. Novel Proteins of the High-Affinity Nitrate Transporter Family NRT2, SaNRT2.1 and SaNRT2.5, from the Euhalophyte Suaeda altissima: Molecular Cloning and Expression Analysis. Int J Mol Sci 2024; 25:5648. [PMID: 38891835 PMCID: PMC11171637 DOI: 10.3390/ijms25115648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
Two genes of nitrate transporters SaNRT2.1 and SaNRT2.5, putative orthologs of high-affinity nitrate transporter genes AtNRT2.1 and AtNRT2.5 from Arabidopsis thaliana, were cloned from the euhalophyte Suaeda altissima. Phylogenetic bioinformatic analysis demonstrated that the proteins SaNRT2.1 and SaNRT2.5 exhibited higher levels of homology to the corresponding proteins from the plants of family Amaranthaceae; the similarity of amino acid sequences between proteins SaNRT2.1 and SaNRT2.5 was lower (54%). Both SaNRT2.1 and SaNRT2.5 are integral membrane proteins forming 12 transmembrane helices as predicted by topological modeling. An attempt to demonstrate nitrate transporting activity of SaNRT2.1 or SaNRT2.5 by heterologous expression of the genes in the yeast Hansenula (Ogataea) polymorpha mutant strain Δynt1 lacking the only yeast nitrate transporter was not successful. The expression patterns of SaNRT2.1 and SaNRT2.5 were studied in S. altissima plants that were grown in hydroponics under either low (0.5 mM) or high (15 mM) nitrate and salinity from 0 to 750 mM NaCl. The growth of the plants was strongly inhibited by low nitrogen supply while stimulated by NaCl; it peaked at 250 mM NaCl for high nitrate and at 500 mM NaCl for low nitrate. Under low nitrate supply, nitrate contents in S. altissima roots, leaves and stems were reduced but increased in leaves and stems as salinity in the medium increased. Potassium contents remained stable under salinity treatment from 250 to 750 mM NaCl. Quantitative real-time PCR demonstrated that without salinity, SaNRT2.1 was expressed in all organs, its expression was not influenced by nitrate supply, while SaNRT2.5 was expressed exclusively in roots-its expression rose about 10-fold under low nitrate. Salinity increased expression of both SaNRT2.1 and SaNRT2.5 under low nitrate. SaNRT2.1 peaked in roots at 500 mM NaCl with 15-fold increase; SaNRT2.5 peaked in roots at 500 mM NaCl with 150-fold increase. It is suggested that SaNRT2.5 ensures effective nitrate uptake by roots and functions as an essential high-affinity nitrate transporter to support growth of adult S. altissima plants under nitrogen deficiency.
Collapse
Affiliation(s)
| | | | | | | | | | - Vadim S. Volkov
- K.A. Timiryazev Institute of Plant Physiology, Russian Academy of Sciences, Moscow 127276, Russia; (D.E.K.); (E.I.R.); (D.A.M.); (A.O.K.); (O.I.N.); (Y.V.B.)
| | | | - Larissa G. Popova
- K.A. Timiryazev Institute of Plant Physiology, Russian Academy of Sciences, Moscow 127276, Russia; (D.E.K.); (E.I.R.); (D.A.M.); (A.O.K.); (O.I.N.); (Y.V.B.)
| |
Collapse
|
24
|
Li J, Zou R, Varrone A, Nag S, Halldin C, Ågren H. Exploring the Interactions between two Ligands, UCB-J and UCB-F, and Synaptic Vesicle Glycoprotein 2 Isoforms. ACS Chem Neurosci 2024; 15:2018-2027. [PMID: 38701380 PMCID: PMC11099911 DOI: 10.1021/acschemneuro.4c00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 05/05/2024] Open
Abstract
In silico modeling was applied to study the efficiency of two ligands, namely, UCB-J and UCB-F, to bind to isoforms of the synaptic vesicle glycoprotein 2 (SV2) that are involved in the regulation of synaptic function in the nerve terminals, with the ultimate goal to understand the selectivity of the interaction between UCB-J and UCB-F to different isoforms of SV2. Docking and large-scale molecular dynamics simulations were carried out to unravel various binding patterns, types of interactions, and binding free energies, covering hydrogen bonding and nonspecific hydrophobic interactions, water bridge, π-π, and cation-π interactions. The overall preference for bonding types of UCB-J and UCB-F with particular residues in the protein pockets can be disclosed in detail. A unique interaction fingerprint, namely, hydrogen bonding with additional cation-π interaction with the pyridine moiety of UCB-J, could be established as an explanation for its high selectivity over the SV2 isoform A (SV2A). Other molecular details, primarily referring to the presence of π-π interactions and hydrogen bonding, could also be analyzed as sources of selectivity of the UCB-F tracer for the three isoforms. The simulations provide atomic details to support future development of new selective tracers targeting synaptic vesicle glycoproteins and their associated diseases.
Collapse
Affiliation(s)
- Junhao Li
- Department
of Physics and Astronomy, Uppsala University, Box 516, Uppsala SE-751 20, Sweden
| | - Rongfeng Zou
- Department
of Physics and Astronomy, Uppsala University, Box 516, Uppsala SE-751 20, Sweden
| | - Andrea Varrone
- Department
of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Stockholm 171 77, Sweden
| | - Sangram Nag
- Department
of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Stockholm 171 77, Sweden
| | - Christer Halldin
- Department
of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Stockholm 171 77, Sweden
| | - Hans Ågren
- Department
of Physics and Astronomy, Uppsala University, Box 516, Uppsala SE-751 20, Sweden
| |
Collapse
|
25
|
Hou X, Lu Z, Yu T, Zhang Y, Yao Q, Zhang C, Niu Y, Liang Q. Two maize homologs of mammalian proton-coupled folate transporter, ZmMFS_1-62 and ZmMFS_1-73, are essential to salt and drought tolerance. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2024; 210:108623. [PMID: 38626656 DOI: 10.1016/j.plaphy.2024.108623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 04/18/2024]
Abstract
Folates are essential to the maintenance of normal life activities in almost all organisms. Proton-coupled folate transporter (PCFT), belonging to the major facilitator superfamily, is one of the three major folate transporter types widely studied in mammals. However, information about plant PCFTs is limited. Here, a genome-wide identification of maize PCFTs was performed, and two PCFTs, ZmMFS_1-62 and ZmMFS_1-73, were functionally investigated. Both proteins contained the typical 12 transmembrane helixes with N- and C-termini located in the cytoplasm, and were localized in the plasma membrane. Molecular docking analysis indicated their binding activity with folates via hydrogen bonding. Interference with ZmMFS_1-62 and ZmMFS_1-73 in maize seedlings through virus-induced gene silencing disrupted folate homeostasis, mainly in the roots, and reduced tolerance to drought and salt stresses. Moreover, a molecular chaperone protein, ZmHSP20, was found to interact with ZmMFS_1-62 and ZmMFS_1-73, and interference with ZmHSP20 in maize seedlings also led to folate disruption and increased sensitivity to drought and salt stresses. Overall, this is the first report of functional identification of maize PCFTs, which play essential roles in salt and drought stress tolerance, thereby linking folate metabolism with abiotic stress responses in maize.
Collapse
Affiliation(s)
- Xiaowan Hou
- Biotechnology Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, China; Key Laboratory of Hainan Province for Postharvest Physiology and Technology of Tropical Horticultural Products, South Subtropical Crops Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, China; Key Laboratory of Forage and Endemic Crop Biotechnology, Ministry of Education, School of Life Sciences, Inner Mongolia University, Hohhot, China.
| | - Zhiwei Lu
- Key Laboratory of Hainan Province for Postharvest Physiology and Technology of Tropical Horticultural Products, South Subtropical Crops Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, China.
| | - Taifei Yu
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, the "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha 410219, China.
| | - Yuanyuan Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, China; Key Laboratory of Hainan Province for Postharvest Physiology and Technology of Tropical Horticultural Products, South Subtropical Crops Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, China.
| | - Quansheng Yao
- Key Laboratory of Hainan Province for Postharvest Physiology and Technology of Tropical Horticultural Products, South Subtropical Crops Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, China.
| | - Chunyi Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, China; National Nanfan Research Institute, Chinese Academy of Agricultural Sciences, Sanya, 572000, China.
| | - Yiding Niu
- Key Laboratory of Forage and Endemic Crop Biotechnology, Ministry of Education, School of Life Sciences, Inner Mongolia University, Hohhot, China.
| | - Qiuju Liang
- Biotechnology Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, China; National Nanfan Research Institute, Chinese Academy of Agricultural Sciences, Sanya, 572000, China.
| |
Collapse
|
26
|
Fiorito V, Tolosano E. Unearthing FLVCR1a: tracing the path to a vital cellular transporter. Cell Mol Life Sci 2024; 81:166. [PMID: 38581583 PMCID: PMC10998817 DOI: 10.1007/s00018-024-05205-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/08/2024] [Accepted: 03/13/2024] [Indexed: 04/08/2024]
Abstract
The Feline Leukemia Virus Subgroup C Receptor 1a (FLVCR1a) is a member of the SLC49 Major Facilitator Superfamily of transporters. Initially recognized as the receptor for the retrovirus responsible of pure red cell aplasia in cats, nearly two decades since its discovery, FLVCR1a remains a puzzling transporter, with ongoing discussions regarding what it transports and how its expression is regulated. Nonetheless, despite this, the substantial body of evidence accumulated over the years has provided insights into several critical processes in which this transporter plays a complex role, and the health implications stemming from its malfunction. The present review intends to offer a comprehensive overview and a critical analysis of the existing literature on FLVCR1a, with the goal of emphasising the vital importance of this transporter for the organism and elucidating the interconnections among the various functions attributed to this transporter.
Collapse
Affiliation(s)
- Veronica Fiorito
- Molecular Biotechnology Center (MBC) "Guido Tarone", Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Turin, Italy
| | - Emanuela Tolosano
- Molecular Biotechnology Center (MBC) "Guido Tarone", Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126, Turin, Italy.
| |
Collapse
|
27
|
Dong H, Yang J, He K, Zheng WB, Lai DH, Liu J, Ding HY, Wu RB, Brown KM, Hide G, Lun ZR, Zhu XQ, Long S. The Toxoplasma monocarboxylate transporters are involved in the metabolism within the apicoplast and are linked to parasite survival. eLife 2024; 12:RP88866. [PMID: 38502570 PMCID: PMC10950331 DOI: 10.7554/elife.88866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024] Open
Abstract
The apicoplast is a four-membrane plastid found in the apicomplexans, which harbors biosynthesis and organelle housekeeping activities in the matrix. However, the mechanism driving the flux of metabolites, in and out, remains unknown. Here, we used TurboID and genome engineering to identify apicoplast transporters in Toxoplasma gondii. Among the many novel transporters, we show that one pair of apicomplexan monocarboxylate transporters (AMTs) appears to have evolved from a putative host cell that engulfed a red alga. Protein depletion showed that AMT1 and AMT2 are critical for parasite growth. Metabolite analyses supported the notion that AMT1 and AMT2 are associated with biosynthesis of isoprenoids and fatty acids. However, stronger phenotypic defects were observed for AMT2, including in the inability to establish T. gondii parasite virulence in mice. This study clarifies, significantly, the mystery of apicoplast transporter composition and reveals the importance of the pair of AMTs in maintaining the apicoplast activity in apicomplexans.
Collapse
Affiliation(s)
- Hui Dong
- National Key Laboratory of Veterinary Public Health Safety, and College of Veterinary Medicine, China Agricultural UniversityBeijingChina
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural UniversityBeijingChina
| | - Jiong Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen UniversityGuangzhouChina
| | - Kai He
- National Key Laboratory of Veterinary Public Health Safety, and College of Veterinary Medicine, China Agricultural UniversityBeijingChina
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural UniversityBeijingChina
| | - Wen-Bin Zheng
- College of Veterinary Medicine, Shanxi Agricultural UniversityTaiguChina
| | - De-Hua Lai
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen UniversityGuangzhouChina
| | - Jing Liu
- National Key Laboratory of Veterinary Public Health Safety, and College of Veterinary Medicine, China Agricultural UniversityBeijingChina
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural UniversityBeijingChina
| | - Hui-Yong Ding
- National Key Laboratory of Veterinary Public Health Safety, and College of Veterinary Medicine, China Agricultural UniversityBeijingChina
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural UniversityBeijingChina
| | - Rui-Bin Wu
- National Key Laboratory of Veterinary Public Health Safety, and College of Veterinary Medicine, China Agricultural UniversityBeijingChina
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural UniversityBeijingChina
| | - Kevin M Brown
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Geoff Hide
- Biomedical Research and Innovation Centre and Environmental Research and Innovation Centre, School of Science, Engineering and Environment, University of SalfordSalfordUnited Kingdom
| | - Zhao-Rong Lun
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen UniversityGuangzhouChina
| | - Xing-Quan Zhu
- College of Veterinary Medicine, Shanxi Agricultural UniversityTaiguChina
| | - Shaojun Long
- National Key Laboratory of Veterinary Public Health Safety, and College of Veterinary Medicine, China Agricultural UniversityBeijingChina
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural UniversityBeijingChina
| |
Collapse
|
28
|
Ricci F, Leggat W, Pasella MM, Bridge T, Horowitz J, Girguis PR, Ainsworth T. Deep sea treasures - Insights from museum archives shed light on coral microbial diversity within deepest ocean ecosystems. Heliyon 2024; 10:e27513. [PMID: 38468949 PMCID: PMC10926130 DOI: 10.1016/j.heliyon.2024.e27513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 02/15/2024] [Accepted: 03/01/2024] [Indexed: 03/13/2024] Open
Abstract
Deep sea benthic habitats are low productivity ecosystems that host an abundance of organisms within the Cnidaria phylum. The technical limitations and the high cost of deep sea surveys have made exploring deep sea environments and the biology of the organisms that inhabit them challenging. In spite of the widespread recognition of Cnidaria's environmental importance in these ecosystems, the microbial assemblage and its role in coral functioning have only been studied for a few deep water corals. Here, we explored the microbial diversity of deep sea corals by recovering nucleic acids from museum archive specimens. Firstly, we amplified and sequenced the V1-V3 regions of the 16S rRNA gene of these specimens, then we utilized the generated sequences to shed light on the microbial diversity associated with seven families of corals collected from depth in the Coral Sea (depth range 1309 to 2959 m) and Southern Ocean (depth range 1401 to 2071 m) benthic habitats. Surprisingly, Cyanobacteria sequences were consistently associated with six out of seven coral families from both sampling locations, suggesting that these bacteria are potentially ubiquitous members of the microbiome within these cold and deep sea water corals. Additionally, we show that Cnidaria might benefit from symbiotic associations with a range of chemosynthetic bacteria including nitrite, carbon monoxide and sulfur oxidizers. Consistent with previous studies, we show that sequences associated with the bacterial phyla Proteobacteria, Verrucomicrobia, Planctomycetes and Acidobacteriota dominated the microbial community of corals in the deep sea. We also explored genomes of the bacterial genus Mycoplasma, which we identified as associated with specimens of three deep sea coral families, finding evidence that these bacteria may aid the host immune system. Importantly our results show that museum specimens retain components of host microbiome that can provide new insights into the diversity of deep sea coral microbiomes (and potentially other organisms), as well as the diversity of microbes writ large in deep sea ecosystems.
Collapse
Affiliation(s)
- Francesco Ricci
- University of New South Wales, School of Biological, Earth and Environmental Sciences, Kensington, NSW, Australia
- University of Melbourne, School of Biosciences, Parkville, VIC, Australia
- Monash University, Department of Microbiology, Biomedicine Discovery Institute, Clayton, VIC, Australia
| | - William Leggat
- University of Newcastle, School of Environmental and Life Sciences, Callaghan, NSW, Australia
| | - Marisa M. Pasella
- University of Melbourne, School of Biosciences, Parkville, VIC, Australia
| | - Tom Bridge
- Biodiversity and Geosciences Program, Museum of Tropical Queensland, Queensland Museum, Townsville, QLD, Australia
- ARC Centre of Excellence for Coral Reef Studies, James Cook University, Townsville, QLD, Australia
| | - Jeremy Horowitz
- ARC Centre of Excellence for Coral Reef Studies, James Cook University, Townsville, QLD, Australia
- Smithsonian Institution, National Museum of Natural History, Washington, DC, USA
| | - Peter R. Girguis
- University of Harvard, Department of Organismic and Evolutionary Biology, Cambridge, MA, USA
| | - Tracy Ainsworth
- University of New South Wales, School of Biological, Earth and Environmental Sciences, Kensington, NSW, Australia
| |
Collapse
|
29
|
Wang Q, Long H, Wang H, Lau Vetter MCY. Characterize the Growth and Metabolism of Acidithiobacillus ferrooxidans under Electroautotrophic and Chemoautotrophic Conditions. Microorganisms 2024; 12:590. [PMID: 38543641 PMCID: PMC10974421 DOI: 10.3390/microorganisms12030590] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 11/12/2024] Open
Abstract
Acidophiles are capable of surviving in extreme environments with low pH. Acidithiobacillus ferrooxidans is a typical acidophilic bacterium that has been extensively studied when grown chemoautotrophically, i.e., when it derives energy from oxidation of Fe2+ or reduced inorganic sulfur compounds (RISCs). Although it is also known to grow with electrons supplied by solid electrodes serving as the sole source of energy, the understanding of its electroautotrophic growth is still limited. This study aimed to compare the growth characteristics of A. ferrooxidans under electroautotrophic (ea) and chemoautotrophic (ca) conditions, with an attempt to elucidate the possible mechanism(s) of extracellular electron flow into the cells. Jarosite was identified by Raman spectroscopy, and it accumulated when A. ferrooxidans used Fe2+ as the electron donor, but negligible mineral deposition occurred during electroautotrophic growth. Scanning electron microscopy (SEM) showed that A. ferrooxidans possesses more pili and extracellular polymeric substances (EPSs) under electroautotrophic conditions. A total of 493 differentially expressed genes (DEGs) were identified, with 297 genes being down-regulated and 196 genes being up-regulated in ea versus ca conditions. The genes known to be essential for chemoautotrophic growth showed a decreased expression in the electroautotrophic condition; meanwhile, there was an increased expression of genes related to direct electron transfer across the cell's outer/inner membranes and transmembrane proteins such as pilin and porin. Joint analysis of DEGs and differentially expressed metabolites (DEMs) showed that galactose metabolism is enhanced during electroautotrophic growth, inducing A. ferrooxidans to produce more EPSs, which aids the cells in adhering to the solid electrode during their growth. These results suggested that electroautotrophy and chemoautotrophy of A. ferrooxidans have different extracellular electron uptake (EEU) pathways, and a model of EEU during electroautotrophic growth is proposed. The use of extracellular electrons as the sole energy source triggers A. ferrooxidans to adopt metabolic and subsequently phenotypic modifications.
Collapse
Affiliation(s)
- Quansheng Wang
- Laboratory of Extraterrestrial Ocean Systems (LEOS), Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya 572000, China; (Q.W.); (H.L.); (H.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Haijun Long
- Laboratory of Extraterrestrial Ocean Systems (LEOS), Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya 572000, China; (Q.W.); (H.L.); (H.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Huiqi Wang
- Laboratory of Extraterrestrial Ocean Systems (LEOS), Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya 572000, China; (Q.W.); (H.L.); (H.W.)
| | - Maggie C. Y. Lau Vetter
- Laboratory of Extraterrestrial Ocean Systems (LEOS), Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya 572000, China; (Q.W.); (H.L.); (H.W.)
| |
Collapse
|
30
|
Hale AT, Boudreau H, Devulapalli R, Duy PQ, Atchley TJ, Dewan MC, Goolam M, Fieggen G, Spader HL, Smith AA, Blount JP, Johnston JM, Rocque BG, Rozzelle CJ, Chong Z, Strahle JM, Schiff SJ, Kahle KT. The genetic basis of hydrocephalus: genes, pathways, mechanisms, and global impact. Fluids Barriers CNS 2024; 21:24. [PMID: 38439105 PMCID: PMC10913327 DOI: 10.1186/s12987-024-00513-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/25/2024] [Indexed: 03/06/2024] Open
Abstract
Hydrocephalus (HC) is a heterogenous disease characterized by alterations in cerebrospinal fluid (CSF) dynamics that may cause increased intracranial pressure. HC is a component of a wide array of genetic syndromes as well as a secondary consequence of brain injury (intraventricular hemorrhage (IVH), infection, etc.) that can present across the age spectrum, highlighting the phenotypic heterogeneity of the disease. Surgical treatments include ventricular shunting and endoscopic third ventriculostomy with or without choroid plexus cauterization, both of which are prone to failure, and no effective pharmacologic treatments for HC have been developed. Thus, there is an urgent need to understand the genetic architecture and molecular pathogenesis of HC. Without this knowledge, the development of preventive, diagnostic, and therapeutic measures is impeded. However, the genetics of HC is extraordinarily complex, based on studies of varying size, scope, and rigor. This review serves to provide a comprehensive overview of genes, pathways, mechanisms, and global impact of genetics contributing to all etiologies of HC in humans.
Collapse
Affiliation(s)
- Andrew T Hale
- Department of Neurosurgery, University of Alabama at Birmingham, FOT Suite 1060, 1720 2ndAve, Birmingham, AL, 35294, UK.
| | - Hunter Boudreau
- Department of Neurosurgery, University of Alabama at Birmingham, FOT Suite 1060, 1720 2ndAve, Birmingham, AL, 35294, UK
| | - Rishi Devulapalli
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, UK
| | - Phan Q Duy
- Department of Neurosurgery, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Travis J Atchley
- Department of Neurosurgery, University of Alabama at Birmingham, FOT Suite 1060, 1720 2ndAve, Birmingham, AL, 35294, UK
| | - Michael C Dewan
- Division of Pediatric Neurosurgery, Monroe Carell Jr. Children's Hospital, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Mubeen Goolam
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Graham Fieggen
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Division of Pediatric Neurosurgery, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
| | - Heather L Spader
- Department of Neurosurgery, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Anastasia A Smith
- Division of Pediatric Neurosurgery, Children's of Alabama, University of Alabama at Birmingham, Birmingham, AL, UK
| | - Jeffrey P Blount
- Division of Pediatric Neurosurgery, Children's of Alabama, University of Alabama at Birmingham, Birmingham, AL, UK
| | - James M Johnston
- Division of Pediatric Neurosurgery, Children's of Alabama, University of Alabama at Birmingham, Birmingham, AL, UK
| | - Brandon G Rocque
- Division of Pediatric Neurosurgery, Children's of Alabama, University of Alabama at Birmingham, Birmingham, AL, UK
| | - Curtis J Rozzelle
- Division of Pediatric Neurosurgery, Children's of Alabama, University of Alabama at Birmingham, Birmingham, AL, UK
| | - Zechen Chong
- Heflin Center for Genomics, University of Alabama at Birmingham, Birmingham, AL, UK
| | - Jennifer M Strahle
- Division of Pediatric Neurosurgery, St. Louis Children's Hospital, Washington University in St. Louis, St. Louis, MO, USA
| | - Steven J Schiff
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
31
|
Zhang C, Gu L, Xie H, Liu Y, Huang P, Zhang J, Luo D, Zhang J. Glucose transport, transporters and metabolism in diabetic retinopathy. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166995. [PMID: 38142757 DOI: 10.1016/j.bbadis.2023.166995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/02/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Diabetic retinopathy (DR) is the most common reason for blindness in working-age individuals globally. Prolonged high blood glucose is a main causative factor for DR development, and glucose transport is prerequisite for the disturbances in DR caused by hyperglycemia. Glucose transport is mediated by its transporters, including the facilitated transporters (glucose transporter, GLUTs), the "active" glucose transporters (sodium-dependent glucose transporters, SGLTs), and the SLC50 family of uniporters (sugars will eventually be exported transporters, SWEETs). Glucose transport across the blood-retinal barrier (BRB) is crucial for nourishing the neuronal retina in the context of retinal physiology. This physiological process primarily relies on GLUTs and SGLTs, which mediate the glucose transportation across both the cell membrane of retinal capillary endothelial cells and the retinal pigment epithelium (RPE). Under diabetic conditions, increased accumulation of extracellular glucose enhances the retinal cellular glucose uptake and metabolism via both glycolysis and glycolytic side branches, which activates several biochemical pathways, including the protein kinase C (PKC), advanced glycation end-products (AGEs), polyol pathway and hexosamine biosynthetic pathway (HBP). These activated biochemical pathways further increase the production of reactive oxygen species (ROS), leading to oxidative stress and activation of Poly (ADP-ribose) polymerase (PARP). The activated PARP further affects all the cellular components in the retina, and finally resulting in microangiopathy, neurodegeneration and low-to-moderate grade inflammation in DR. This review aims to discuss the changes of glucose transport, glucose transporters, as well as its metabolism in DR, which influences the retinal neurovascular unit (NVU) and implies the possible therapeutic strategies for treating DR.
Collapse
Affiliation(s)
- Chaoyang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Limin Gu
- Department of Ophthalmology, Shanghai Aier Eye Hospital, Shanghai, China.
| | - Hai Xie
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Yan Liu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Peirong Huang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Jingting Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Dawei Luo
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| |
Collapse
|
32
|
Liao M, Yao D, Wu L, Luo C, Wang Z, Zhang J, Liu B. Targeting the Warburg effect: A revisited perspective from molecular mechanisms to traditional and innovative therapeutic strategies in cancer. Acta Pharm Sin B 2024; 14:953-1008. [PMID: 38487001 PMCID: PMC10935242 DOI: 10.1016/j.apsb.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 03/17/2024] Open
Abstract
Cancer reprogramming is an important facilitator of cancer development and survival, with tumor cells exhibiting a preference for aerobic glycolysis beyond oxidative phosphorylation, even under sufficient oxygen supply condition. This metabolic alteration, known as the Warburg effect, serves as a significant indicator of malignant tumor transformation. The Warburg effect primarily impacts cancer occurrence by influencing the aerobic glycolysis pathway in cancer cells. Key enzymes involved in this process include glucose transporters (GLUTs), HKs, PFKs, LDHs, and PKM2. Moreover, the expression of transcriptional regulatory factors and proteins, such as FOXM1, p53, NF-κB, HIF1α, and c-Myc, can also influence cancer progression. Furthermore, lncRNAs, miRNAs, and circular RNAs play a vital role in directly regulating the Warburg effect. Additionally, gene mutations, tumor microenvironment remodeling, and immune system interactions are closely associated with the Warburg effect. Notably, the development of drugs targeting the Warburg effect has exhibited promising potential in tumor treatment. This comprehensive review presents novel directions and approaches for the early diagnosis and treatment of cancer patients by conducting in-depth research and summarizing the bright prospects of targeting the Warburg effect in cancer.
Collapse
Affiliation(s)
- Minru Liao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dahong Yao
- School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518118, China
| | - Lifeng Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chaodan Luo
- Department of Psychology, University of Southern California, Los Angeles, CA 90089, USA
| | - Zhiwen Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518118, China
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jin Zhang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Bo Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
33
|
Luna-Bulbarela A, Romero-Gutiérrez MT, Tinoco-Valencia R, Ortiz E, Martínez-Romero ME, Galindo E, Serrano-Carreón L. Response of Bacillus velezensis 83 to interaction with Colletotrichum gloeosporioides resembles a Greek phalanx-style formation: A stress resistant phenotype with antibiosis capacity. Microbiol Res 2024; 280:127592. [PMID: 38199003 DOI: 10.1016/j.micres.2023.127592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/06/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024]
Abstract
Plant growth-promoting rhizobacteria, such as Bacillus spp., establish beneficial associations with plants and may inhibit the growth of phytopathogenic fungi. However, these bacteria are subject to multiple biotic stimuli from their competitors, causing stress and modifying their development. This work is a study of an in vitro interaction between two model microorganisms of socioeconomic relevance, using population dynamics and transcriptomic approaches. Co-cultures of Bacillus velezensis 83 with the phytopathogenic fungus Colletotrichum gloeosporioides 09 were performed to evaluate the metabolic response of the bacteria under conditions of non-nutritional limitation. The bacterial response was associated with the induction of a stress-resistant phenotype, characterized by a lower specific growth rate, but with antimicrobial production capacity. About 12% of co-cultured B. velezensis 83 coding sequences were differentially expressed, including the up-regulation of the general stress response (sigB regulon), and the down-regulation of alternative carbon sources catabolism (glucose preference). Defense strategies in B. velezensis are a determining factor in order to preserve the long-term viability of its population. Mostly, the presence of the fungus does not affect the expression of antibiosis genes, except for those corresponding to surfactin/bacillomycin D production. Indeed, the up-regulation of antibiosis genes expression is associated with bacterial growth, regardless of the presence of the fungus. This behavior in B. velezensis 83 resembles the strategy used by the classical Greek phalanx formation: by sacrificing growth rate and metabolic versatility, resources can be redistributed to defense (stress resistant phenotype) while maintaining the attack (antibiosis capacity). The presented results are the first characterization of the molecular phenotype at the transcriptome level of a biological control agent under biotic stress caused by a phytopathogen without nutrient limitation.
Collapse
Affiliation(s)
- Agustín Luna-Bulbarela
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad #2001, Col. Chamilpa, CP 62210 Cuernavaca, Morelos, Mexico; Agro&Biotecnia S. de R.L. de C.V., Limones 8, Amate Redondo, 62334 Cuernavaca, Morelos, Mexico
| | - María Teresa Romero-Gutiérrez
- Technological Innovation Department, Tlajomulco University Center, University of Guadalajara, 45641 Tlajomulco de Zúñiga, Jalisco, Mexico; Translational Bioengineering Department, Exact Sciences and Engineering University Center, Universidad de Guadalajara, Blvd. Marcelino García Barragán #1421, 44430 Guadalajara, Jalisco, Mexico
| | - Raunel Tinoco-Valencia
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad #2001, Col. Chamilpa, CP 62210 Cuernavaca, Morelos, Mexico
| | - Ernesto Ortiz
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad #2001, Col. Chamilpa, CP 62210 Cuernavaca, Morelos, Mexico
| | - María Esperanza Martínez-Romero
- Ecología Genómica, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Av. Universidad s/n, Col. Chamilpa, CP 62210 Cuernavaca, Morelos, Mexico
| | - Enrique Galindo
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad #2001, Col. Chamilpa, CP 62210 Cuernavaca, Morelos, Mexico; Agro&Biotecnia S. de R.L. de C.V., Limones 8, Amate Redondo, 62334 Cuernavaca, Morelos, Mexico
| | - Leobardo Serrano-Carreón
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad #2001, Col. Chamilpa, CP 62210 Cuernavaca, Morelos, Mexico; Agro&Biotecnia S. de R.L. de C.V., Limones 8, Amate Redondo, 62334 Cuernavaca, Morelos, Mexico.
| |
Collapse
|
34
|
Kim JH, Mailloux L, Bloor D, Tae H, Nguyen H, McDowell M, Padilla J, DeWaard A. Multiple roles for the cytoplasmic C-terminal domains of the yeast cell surface receptors Rgt2 and Snf3 in glucose sensing and signaling. Sci Rep 2024; 14:4055. [PMID: 38374219 PMCID: PMC10876965 DOI: 10.1038/s41598-024-54628-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024] Open
Abstract
The plasma membrane proteins Rgt2 and Snf3 are glucose sensing receptors (GSRs) that generate an intracellular signal for the induction of gene expression in response to high and low extracellular glucose concentrations, respectively. The GSRs consist of a 12-transmembrane glucose recognition domain and a cytoplasmic C-terminal signaling tail. The GSR tails are dissimilar in length and sequence, but their distinct roles in glucose signal transduction are poorly understood. Here, we show that swapping the tails between Rgt2 and Snf3 does not alter the signaling activity of the GSRs, so long as their tails are phosphorylated in a Yck-dependent manner. Attachment of the GSR tails to Hxt1 converts the transporter into a glucose receptor; however, the tails attached to Hxt1 are not phosphorylated by the Ycks, resulting in only partial signaling. Moreover, in response to non-fermentable carbon substrates, Rgt2 and Hxt1-RT (RT, Rgt2-tail) are efficiently endocytosed, whereas Snf3 and Hxt1-ST (ST, Snf3-tail) are endocytosis-impaired. Thus, the tails are important regulatory domains required for the endocytosis of the Rgt2 and Snf3 glucose sensing receptors triggered by different cellular stimuli. Taken together, these results suggest multiple roles for the tail domains in GSR-mediated glucose sensing and signaling.
Collapse
Affiliation(s)
- Jeong-Ho Kim
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA.
| | - Levi Mailloux
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| | - Daniel Bloor
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| | - Haeun Tae
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| | - Han Nguyen
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| | - Morgan McDowell
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| | - Jaqueline Padilla
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| | - Anna DeWaard
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| |
Collapse
|
35
|
Currie MJ, Davies JS, Scalise M, Gulati A, Wright JD, Newton-Vesty MC, Abeysekera GS, Subramanian R, Wahlgren WY, Friemann R, Allison JR, Mace PD, Griffin MDW, Demeler B, Wakatsuki S, Drew D, Indiveri C, Dobson RCJ, North RA. Structural and biophysical analysis of a Haemophilus influenzae tripartite ATP-independent periplasmic (TRAP) transporter. eLife 2024; 12:RP92307. [PMID: 38349818 PMCID: PMC10942642 DOI: 10.7554/elife.92307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024] Open
Abstract
Tripartite ATP-independent periplasmic (TRAP) transporters are secondary-active transporters that receive their substrates via a soluble-binding protein to move bioorganic acids across bacterial or archaeal cell membranes. Recent cryo-electron microscopy (cryo-EM) structures of TRAP transporters provide a broad framework to understand how they work, but the mechanistic details of transport are not yet defined. Here we report the cryo-EM structure of the Haemophilus influenzae N-acetylneuraminate TRAP transporter (HiSiaQM) at 2.99 Å resolution (extending to 2.2 Å at the core), revealing new features. The improved resolution (the previous HiSiaQM structure is 4.7 Å resolution) permits accurate assignment of two Na+ sites and the architecture of the substrate-binding site, consistent with mutagenic and functional data. Moreover, rather than a monomer, the HiSiaQM structure is a homodimer. We observe lipids at the dimer interface, as well as a lipid trapped within the fusion that links the SiaQ and SiaM subunits. We show that the affinity (KD) for the complex between the soluble HiSiaP protein and HiSiaQM is in the micromolar range and that a related SiaP can bind HiSiaQM. This work provides key data that enhances our understanding of the 'elevator-with-an-operator' mechanism of TRAP transporters.
Collapse
Affiliation(s)
- Michael J Currie
- Biomolecular Interaction Centre, Maurice Wilkins Centre for Biodiscovery, MacDiarmid Institute for Advanced Materials and Nanotechnology, and School of Biological Sciences, University of CanterburyChristchurchNew Zealand
| | - James S Davies
- Biomolecular Interaction Centre, Maurice Wilkins Centre for Biodiscovery, MacDiarmid Institute for Advanced Materials and Nanotechnology, and School of Biological Sciences, University of CanterburyChristchurchNew Zealand
- Department of Biochemistry and Biophysics, Stockholm UniversityStockholmSweden
| | - Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of CalabriaArcavacata di RendeItaly
| | - Ashutosh Gulati
- Department of Biochemistry and Biophysics, Stockholm UniversityStockholmSweden
| | - Joshua D Wright
- Biomolecular Interaction Centre, Maurice Wilkins Centre for Biodiscovery, MacDiarmid Institute for Advanced Materials and Nanotechnology, and School of Biological Sciences, University of CanterburyChristchurchNew Zealand
| | - Michael C Newton-Vesty
- Biomolecular Interaction Centre, Maurice Wilkins Centre for Biodiscovery, MacDiarmid Institute for Advanced Materials and Nanotechnology, and School of Biological Sciences, University of CanterburyChristchurchNew Zealand
| | - Gayan S Abeysekera
- Biomolecular Interaction Centre, Maurice Wilkins Centre for Biodiscovery, MacDiarmid Institute for Advanced Materials and Nanotechnology, and School of Biological Sciences, University of CanterburyChristchurchNew Zealand
| | - Ramaswamy Subramanian
- Biological Sciences and Biomedical Engineering, Bindley Bioscience Center, Purdue University West LafayetteWest LafayetteUnited States
| | - Weixiao Y Wahlgren
- Department of Chemistry and Molecular Biology, Biochemistry and Structural Biology, University of GothenburgGothenburgSweden
| | - Rosmarie Friemann
- Centre for Antibiotic Resistance Research (CARe) at University of GothenburgGothenburgSweden
| | - Jane R Allison
- Biomolecular Interaction Centre, Digital Life Institute, Maurice Wilkins Centre for Molecular Biodiscovery, and School of Biological Sciences, University of AucklandAucklandNew Zealand
| | - Peter D Mace
- Biochemistry Department, School of Biomedical Sciences, University of OtagoDunedinNew Zealand
| | - Michael DW Griffin
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Bio Molecular Science and Biotechnology Institute, Department of Biochemistry and Pharmacology, University of MelbourneMelbourneAustralia
| | - Borries Demeler
- Department of Chemistry and Biochemistry, University of MontanaMissoulaUnited States
- Department of Chemistry and Biochemistry, University of LethbridgeLethbridgeCanada
| | - Soichi Wakatsuki
- Biological Sciences Division, SLAC National Accelerator LaboratoryMenlo ParkUnited States
- Department of Structural Biology, Stanford University School of MedicineStanfordUnited States
| | - David Drew
- Department of Biochemistry and Biophysics, Stockholm UniversityStockholmSweden
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of CalabriaArcavacata di RendeItaly
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM)BariItaly
| | - Renwick CJ Dobson
- Biomolecular Interaction Centre, Maurice Wilkins Centre for Biodiscovery, MacDiarmid Institute for Advanced Materials and Nanotechnology, and School of Biological Sciences, University of CanterburyChristchurchNew Zealand
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Bio Molecular Science and Biotechnology Institute, Department of Biochemistry and Pharmacology, University of MelbourneMelbourneAustralia
| | - Rachel A North
- Department of Biochemistry and Biophysics, Stockholm UniversityStockholmSweden
- School of Medical Sciences, Faculty of Medicine and Health, University of SydneySydneyAustralia
| |
Collapse
|
36
|
Wu D, Chen Q, Yu Z, Huang B, Zhao J, Wang Y, Su J, Zhou F, Yan R, Li N, Zhao Y, Jiang D. Transport and inhibition mechanisms of human VMAT2. Nature 2024; 626:427-434. [PMID: 38081299 DOI: 10.1038/s41586-023-06926-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/01/2023] [Indexed: 01/19/2024]
Abstract
Vesicular monoamine transporter 2 (VMAT2) accumulates monoamines in presynaptic vesicles for storage and exocytotic release, and has a vital role in monoaminergic neurotransmission1-3. Dysfunction of monoaminergic systems causes many neurological and psychiatric disorders, including Parkinson's disease, hyperkinetic movement disorders and depression4-6. Suppressing VMAT2 with reserpine and tetrabenazine alleviates symptoms of hypertension and Huntington's disease7,8, respectively. Here we describe cryo-electron microscopy structures of human VMAT2 complexed with serotonin and three clinical drugs at 3.5-2.8 Å, demonstrating the structural basis for transport and inhibition. Reserpine and ketanserin occupy the substrate-binding pocket and lock VMAT2 in cytoplasm-facing and lumen-facing states, respectively, whereas tetrabenazine binds in a VMAT2-specific pocket and traps VMAT2 in an occluded state. The structures in three distinct states also reveal the structural basis of the VMAT2 transport cycle. Our study establishes a structural foundation for the mechanistic understanding of substrate recognition, transport, drug inhibition and pharmacology of VMAT2 while shedding light on the rational design of potential therapeutic agents.
Collapse
Affiliation(s)
- Di Wu
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qihao Chen
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Biomacromolecules, Chinese Academy of Sciences, National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhuoya Yu
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Biomacromolecules, Chinese Academy of Sciences, National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Bo Huang
- Beijing StoneWise Technology, Beijing, China
| | - Jun Zhao
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences at Weifang, Weifang, China
| | - Yuhang Wang
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Biomacromolecules, Chinese Academy of Sciences, National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jiawei Su
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Biomacromolecules, Chinese Academy of Sciences, National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Feng Zhou
- Beijing StoneWise Technology, Beijing, China
| | - Rui Yan
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Na Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yan Zhao
- University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Biomacromolecules, Chinese Academy of Sciences, National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| | - Daohua Jiang
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Songshan Lake Materials Laboratory, Dongguan, China.
| |
Collapse
|
37
|
Li X, Ma Y, Zhang N, Li Y, Liang Z, Luo Y, Lin L, Zhang D, He Y, Wang Z, Zhang Z, Deng Y. Whole-genome sequencing of Fusarium spp. causing sugarcane root rot on both chewing cane and sugar-making cane. STRESS BIOLOGY 2024; 4:7. [PMID: 38270818 PMCID: PMC10811303 DOI: 10.1007/s44154-023-00145-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/24/2023] [Indexed: 01/26/2024]
Abstract
Previously we isolated three Fusarium strains (a F. sacchari strain namely GXUF-1, and another two F. commune strains namely GXUF-2 and GXUF-3), and we verified that GXUF-3 was able to cause sugarcane root rot to the chewing cane cultivar Badila. Considering that Fusarium spp. are a group of widely distributed fungal pathogens, we tested whether these three Fusarium isolates were able to cause root rot to Badila as well as sugar-making cane cultivar (Guitang42), using a suitable inoculation method established based on infection assays using Badila. We found that the three Fusarium strains were able to cause root rot symptoms to both Badila and Guitang42, to different extents. To better investigate the potential pathogenicity mechanisms, we performed Illumina high-throughput sequencing and analyzed the whole genomic sequence data of these three Fusarium strains. The results reveal that the assembly sizes of the three Fusarium strains were in a range of 44.7-48.2 Mb, with G + C contents of 48.0-48.5%, and 14,154-15,175 coding genes. The coding genes were annotated by multiple public databases, and potential pathogenic genes were predicted using proprietary databases (such as PHI, DFVF, CAZy, etc.). Furthermore, based on evolutionary analysis of the coding sequence, we found that contraction and expansion of gene families occurred in the three Fusarium strains. Overall, our results suggest a potential risk that the root rot disease may occur to the sugar-making canes although it was initially spotted from fruit cane, and provide clues to understand the pathogenic mechanisms of Fusarium spp. causing sugarcane root rot.
Collapse
Affiliation(s)
- Xinyang Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresource, Guangxi Key Laboratory of Sugarcane Biology, Guangxi University, Nanning, 530004, China
| | - Yuming Ma
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, South China Agricultural University, Guangzhou, 510642, China
| | - Na Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, South China Agricultural University, Guangzhou, 510642, China
| | - Yiming Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresource, Guangxi Key Laboratory of Sugarcane Biology, Guangxi University, Nanning, 530004, China
| | - Zhibin Liang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, South China Agricultural University, Guangzhou, 510642, China
| | - Yibao Luo
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresource, Guangxi Key Laboratory of Sugarcane Biology, Guangxi University, Nanning, 530004, China
| | - Longxin Lin
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresource, Guangxi Key Laboratory of Sugarcane Biology, Guangxi University, Nanning, 530004, China
| | - Dongliang Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresource, Guangxi Key Laboratory of Sugarcane Biology, Guangxi University, Nanning, 530004, China
| | - Yongqiang He
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresource, Guangxi Key Laboratory of Sugarcane Biology, Guangxi University, Nanning, 530004, China
| | - Ziting Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresource, Guangxi Key Laboratory of Sugarcane Biology, Guangxi University, Nanning, 530004, China
| | - Zhiquan Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresource, Guangxi Key Laboratory of Sugarcane Biology, Guangxi University, Nanning, 530004, China
| | - Yizhen Deng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
38
|
Zhou H, Jia S, Gao Y, Li X, Lin Y, Yang F, Ni K. Characterization of phyllosphere endophytic lactic acid bacteria reveals a potential novel route to enhance silage fermentation quality. Commun Biol 2024; 7:117. [PMID: 38253824 PMCID: PMC10803313 DOI: 10.1038/s42003-024-05816-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
The naturally attached phyllosphere microbiota play a crucial role in plant-derived fermentation, but the structure and function of phyllosphere endophytes remain largely unidentified. Here, we reveal the diversity, specificity, and functionality of phyllosphere endophytes in alfalfa (Medicago sativa L.) through combining typical microbial culture, high-throughput sequencing, and genomic comparative analysis. In comparison to phyllosphere bacteria (PB), the fermentation of alfalfa solely with endophytes (EN) enhances the fermentation characteristics, primarily due to the dominance of specific lactic acid bacteria (LAB) such as Lactiplantibacillus, Weissella, and Pediococcus. The inoculant with selected endophytic LAB strains also enhances the fermentation quality compared to epiphytic LAB treatment. Especially, one key endophytic LAB named Pediococcus pentosaceus EN5 shows enrichment of genes related to the mannose phosphotransferase system (Man-PTS) and carbohydrate-metabolizing enzymes and higher utilization of carbohydrates. Representing phyllosphere, endophytic LAB shows great potential of promoting ensiling and provides a novel direction for developing microbial inoculant.
Collapse
Affiliation(s)
- Hongzhang Zhou
- College of Grassland Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Shangang Jia
- College of Grassland Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yu Gao
- College of Grassland Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiaomei Li
- College of Grassland Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yanli Lin
- College of Grassland Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Fuyu Yang
- College of Grassland Science and Technology, China Agricultural University, Beijing, 100193, China
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Kuikui Ni
- College of Grassland Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
39
|
Wu PC, Choo YL, Wei SY, Yago JI, Chung KR. Contribution of Autophagy to Cellular Iron Homeostasis and Stress Adaptation in Alternaria alternata. Int J Mol Sci 2024; 25:1123. [PMID: 38256200 PMCID: PMC10816921 DOI: 10.3390/ijms25021123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/04/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
The tangerine pathotype of Alternaria alternata produces the Alternaria citri toxin (ACT), which elicits a host immune response characterized by the increase in harmful reactive oxygen species (ROS) production. ROS detoxification in A. alternata relies on the degradation of peroxisomes through autophagy and iron acquisition using siderophores. In this study, we investigated the role of autophagy in regulating siderophore and iron homeostasis in A. alternata. Our results showed that autophagy positively influences siderophore production and iron uptake. The A. alternata strains deficient in autophagy-related genes 1 and 8 (ΔAaatg1 and ΔAaatg8) could not thrive without iron, and their adaptability to high-iron environments was also reduced. Furthermore, the ability of autophagy-deficient strains to withstand ROS was compromised. Notably, autophagy deficiency significantly reduced the production of dimerumic acid (DMA), a siderophore in A. alternata, which may contribute to ROS detoxification. Compared to the wild-type strain, ΔAaatg8 was defective in cellular iron balances. We also observed iron-induced autophagy and lipid peroxidation in A. alternata. To summarize, our study indicates that autophagy and maintaining iron homeostasis are interconnected and contribute to the stress resistance and the virulence of A. alternata. These results provide new insights into the complex interplay connecting autophagy, iron metabolism, and fungal pathogenesis in A. alternata.
Collapse
Affiliation(s)
- Pei-Ching Wu
- Department of Plant Pathology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung 402202, Taiwan; (P.-C.W.); (Y.-L.C.); (S.-Y.W.)
| | - Yen-Ling Choo
- Department of Plant Pathology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung 402202, Taiwan; (P.-C.W.); (Y.-L.C.); (S.-Y.W.)
| | - Sian-Yong Wei
- Department of Plant Pathology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung 402202, Taiwan; (P.-C.W.); (Y.-L.C.); (S.-Y.W.)
| | - Jonar I. Yago
- Plant Science Department, College of Agriculture, Nueva Vizcaya State University, Bayombong 3700, Philippines;
| | - Kuang-Ren Chung
- Department of Plant Pathology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung 402202, Taiwan; (P.-C.W.); (Y.-L.C.); (S.-Y.W.)
| |
Collapse
|
40
|
He S, Taher NM, Hvorecny KL, Ragusa MJ, Bahl CD, Hickman AB, Dyda F, Madden DR. Molecular basis for the transcriptional regulation of an epoxide-based virulence circuit in Pseudomonas aeruginosa. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.16.572601. [PMID: 38293063 PMCID: PMC10827105 DOI: 10.1101/2024.01.16.572601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The opportunistic pathogen Pseudomonas aeruginosa infects cystic fibrosis (CF) patient airways and produces a virulence factor Cif that is associated with worse outcomes. Cif is an epoxide hydrolase that reduces cell-surface abundance of the cystic fibrosis transmembrane conductance regulator (CFTR) and sabotages pro-resolving signals. Its expression is regulated by a divergently transcribed TetR family transcriptional repressor. CifR represents the first reported epoxide-sensing bacterial transcriptional regulator, but neither its interaction with cognate operator sequences nor the mechanism of activation has been investigated. Using biochemical and structural approaches, we uncovered the molecular mechanisms controlling this complex virulence operon. We present here the first molecular structures of CifR alone and in complex with operator DNA, resolved in a single crystal lattice. Significant conformational changes between these two structures suggest how CifR regulates the expression of the virulence gene cif. Interactions between the N-terminal extension of CifR with the DNA minor groove of the operator play a significant role in the operator recognition of CifR. We also determined that cysteine residue Cys107 is critical for epoxide sensing and DNA release. These results offer new insights into the stereochemical regulation of an epoxide-based virulence circuit in a critically important clinical pathogen.
Collapse
Affiliation(s)
- Susu He
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755 USA
| | - Noor M. Taher
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755 USA
| | - Kelli L. Hvorecny
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755 USA
| | - Michael J. Ragusa
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755 USA
- Department of Chemistry, Dartmouth, Hanover, NH 03755 USA
| | - Christopher D. Bahl
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755 USA
| | - Alison B. Hickman
- Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892 USA
| | - Fred Dyda
- Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892 USA
| | - Dean R. Madden
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755 USA
- Department of Chemistry, Dartmouth, Hanover, NH 03755 USA
| |
Collapse
|
41
|
Antunes M, Sá-Correia I. The role of ion homeostasis in adaptation and tolerance to acetic acid stress in yeasts. FEMS Yeast Res 2024; 24:foae016. [PMID: 38658183 PMCID: PMC11092280 DOI: 10.1093/femsyr/foae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 04/26/2024] Open
Abstract
Maintenance of asymmetric ion concentrations across cellular membranes is crucial for proper yeast cellular function. Disruptions of these ionic gradients can significantly impact membrane electrochemical potential and the balance of other ions, particularly under stressful conditions such as exposure to acetic acid. This weak acid, ubiquitous to both yeast metabolism and industrial processes, is a major inhibitor of yeast cell growth in industrial settings and a key determinant of host colonization by pathogenic yeast. Acetic acid toxicity depends on medium composition, especially on the pH (H+ concentration), but also on other ions' concentrations. Regulation of ion fluxes is essential for effective yeast response and adaptation to acetic acid stress. However, the intricate interplay among ion balancing systems and stress response mechanisms still presents significant knowledge gaps. This review offers a comprehensive overview of the mechanisms governing ion homeostasis, including H+, K+, Zn2+, Fe2+/3+, and acetate, in the context of acetic acid toxicity, adaptation, and tolerance. While focus is given on Saccharomyces cerevisiae due to its extensive physiological characterization, insights are also provided for biotechnologically and clinically relevant yeast species whenever available.
Collapse
Affiliation(s)
- Miguel Antunes
- iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001, Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1049-001, Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, 1049-001, Lisbon, Portugal
| | - Isabel Sá-Correia
- iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001, Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1049-001, Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, 1049-001, Lisbon, Portugal
| |
Collapse
|
42
|
Zheng C, Liu D, Lu X, Wu H, Hua J, Zhang C, Liu K, Li C, He J, Du C. Trans-aconitic acid assimilation system as a widespread bacterial mechanism for environmental adaptation. THE ISME JOURNAL 2024; 18:wrae198. [PMID: 39375013 PMCID: PMC11495376 DOI: 10.1093/ismejo/wrae198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/18/2024] [Accepted: 10/03/2024] [Indexed: 10/09/2024]
Abstract
The ability of bacteria to use natural carbon sources greatly affects their growth and survival in the environment. Bacteria have evolved versatile abilities to use environmental carbon sources, but their diversity and assimilation pathways remain largely unexplored. Trans-aconitic acid (TAA), a geometric isomer of cis-aconitic acid involved in the tricarboxylic acid cycle, has long been considered a natural carbon source metabolizable by bacteria. However, its catabolism and ecological role in linking bacterial interactions with the environment remain unclear. Here, we identify a regulatory system in Bacillus velezensis FZB42 that is capable of sensing and catabolizing TAA. The system consists of a tar operon, an adjacent positive regulatory gene tarR, and a shared promoter. After receiving the TAA signal, the TarR protein interacts directly with the promoter, initiating the expression of the membrane transporter TarB and aconitate isomerase TarA encoded by the operon, which function in importing the TAA and isomerizing it into the central intermediate cis-aconitic acid. Subsequent soil colonization experiments reveal that TAA assimilating ability can give its coding bacteria a growth and competitive advantage. Bioinformatics analyses coupled with bacterial isolation experiments further show that the assimilation system of TAA is widely distributed in the bacterial domain, and its assimilating bacteria are also extensively distributed in nature, indicating an important role of TAA metabolism in bacterial carbon acquisition. This work emphasizes the importance of metabolic adaptation to environmental carbon sources for bacterial survival and may provide inspiration for engineering microbes with enhanced environmental competitiveness.
Collapse
Affiliation(s)
- Cao Zheng
- Hubei Province Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, College of Life Science and Technology, Hubei Engineering University, Xiaogan, Hubei 432000, China
| | - Dingqi Liu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xinyu Lu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Huijun Wu
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Jingyi Hua
- Hubei Province Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, College of Life Science and Technology, Hubei Engineering University, Xiaogan, Hubei 432000, China
| | - Chuang Zhang
- Hubei Province Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, College of Life Science and Technology, Hubei Engineering University, Xiaogan, Hubei 432000, China
| | - Kang Liu
- Hubei Province Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, College of Life Science and Technology, Hubei Engineering University, Xiaogan, Hubei 432000, China
| | - Changchun Li
- Hubei Province Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, College of Life Science and Technology, Hubei Engineering University, Xiaogan, Hubei 432000, China
| | - Jin He
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Cuiying Du
- Hubei Province Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, College of Life Science and Technology, Hubei Engineering University, Xiaogan, Hubei 432000, China
| |
Collapse
|
43
|
Paiva DS, Fernandes L, Portugal A, Trovão J. First Genome Sequence of the Microcolonial Black Fungus Saxispiralis lemnorum MUM 23.14: Insights into the Unique Genomic Traits of the Aeminiaceae Family. Microorganisms 2024; 12:104. [PMID: 38257931 PMCID: PMC10820743 DOI: 10.3390/microorganisms12010104] [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: 11/16/2023] [Revised: 12/15/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Saxispiralis lemnorum MUM 23.14 is an extremotolerant microcolonial black fungus, originally isolated from a biodeteriorated limestone artwork in Portugal. This recently introduced species belongs to the Aeminiaceae family, representing the second member of this monophyletic clade. This fungus exhibits a unique set of characteristics, including xerophily, cold tolerance, high UV radiation tolerance, and an exceptional ability to thrive in NaCl concentrations of up to 30% while also enduring pH levels ranging from 5 to 11. To gain insights into its genomic traits associated with stress resistance mechanisms, specialization, and their potential implications in stone biodeterioration, we conducted a comprehensive genome sequencing and analysis. This draft genome not only marks the first for the Saxispiralis genus but also the second for the Aeminiaceae family. Furthermore, we performed two comparative genomic analyses: one focusing on the closest relative within the Aeminiaceae family, Aeminium ludgeri, and another encompassing the genome of different extremotolerant black fungi. In this study, we successfully achieved high genome completeness for S. lemnorum and confirmed its close phylogenetic relationship to A. ludgeri. Our findings revealed traits contributing to its extremophilic nature and provided insights into potential mechanisms contributing to stone biodeterioration. Many traits are common to both Aeminiaceae species and are shared with other black fungi, while numerous unique traits may be attributed to species-specific characteristics.
Collapse
Affiliation(s)
- Diana S. Paiva
- Centre for Functional Ecology (CFE)—Science for People & the Planet, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal (J.T.)
| | - Luís Fernandes
- Centre for Functional Ecology (CFE)—Science for People & the Planet, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal (J.T.)
| | - António Portugal
- Centre for Functional Ecology (CFE)—Science for People & the Planet, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal (J.T.)
- FitoLab—Laboratory for Phytopathology, Instituto Pedro Nunes (IPN), Rua Pedro Nunes, 3030-199 Coimbra, Portugal
- TERRA—Associate Laboratory for Sustainable Land Use and Ecosystem Services, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - João Trovão
- Centre for Functional Ecology (CFE)—Science for People & the Planet, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal (J.T.)
| |
Collapse
|
44
|
Roxo I, Amaral A, Portugal A, Trovão J. Draft genome sequence and comparative genomic analysis of Penicillium pancosmium MUM 23.27 isolated from raw honey. Arch Microbiol 2023; 206:36. [PMID: 38142242 DOI: 10.1007/s00203-023-03766-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/13/2023] [Accepted: 11/22/2023] [Indexed: 12/25/2023]
Abstract
The draft genome sequence and main genomic features of Penicillium pancosmium MUM 23.27, isolated from Portuguese raw honey are reported. The genome size is 34.82 Mb, containing a 48.99% GC content, 11,394 genes, with 39 rRNAs and 147 tRNAs/tmRNAs. Twenty-six BGCs were predicted with four exhibiting significant similarities with YWA1, chaetoglobosin A/chaetoglobosin C, squalestatin S1, and nidulanin A. Moreover, the whole-genome sequencing and in silico genomic analysis, allowed to further understand some aspects of this species habitat, resistance, and evolutionary genomic events. Altogether, the results obtained also allow to dwell deeper on particular Penicillia biological characteristics and genomic traits, permitting them to thrive in these honey substrates. In addition, this resource represents the first genome for the species and one of the first for raw honeys filamentous fungi.
Collapse
Affiliation(s)
- Ivo Roxo
- FitoLab - Laboratory for Phytopathology, Instituto Pedro Nunes, Rua Pedro Nunes, Quinta da Nora, 3030-199, Coimbra, Portugal.
- Polytechnic Institute of Coimbra, Coimbra Institute of Engineering, Rua Pedro Nunes, Quinta da Nora, 3030-199, Coimbra, Portugal.
| | - António Amaral
- Polytechnic Institute of Coimbra, Coimbra Institute of Engineering, Rua Pedro Nunes, Quinta da Nora, 3030-199, Coimbra, Portugal
- CEB, Centre of Biological Engineering, Universidade Do Minho, Campus de Gualtar, 4710-057, Braga, Portugal
- LABBELS - Associate Laboratory, Centre of Biological Engineering, Universidade Do Minho, Campus de Gualtar, 4710-057, Braga/Guimarães, Portugal
- Laboratório SiSus, Instituto de Investigação Aplicada, Rua Pedro Nunes, Quinta da Nora, 3030-199, Coimbra, Portugal
| | - António Portugal
- FitoLab - Laboratory for Phytopathology, Instituto Pedro Nunes, Rua Pedro Nunes, Quinta da Nora, 3030-199, Coimbra, Portugal
- Centre for Functional Ecology - Science for People and the Planet, Department of Life Sciences, TERRA Associate Laboratory, University of Coimbra, Calçada Martim de Freitas 3000-456, Coimbra, Portugal
| | - João Trovão
- FitoLab - Laboratory for Phytopathology, Instituto Pedro Nunes, Rua Pedro Nunes, Quinta da Nora, 3030-199, Coimbra, Portugal.
- Centre for Functional Ecology - Science for People and the Planet, Department of Life Sciences, TERRA Associate Laboratory, University of Coimbra, Calçada Martim de Freitas 3000-456, Coimbra, Portugal.
| |
Collapse
|
45
|
Jormakka M. Structural insights into ferroportin mediated iron transport. Biochem Soc Trans 2023; 51:BST20230594. [PMID: 38115725 DOI: 10.1042/bst20230594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
Iron is a vital trace element for almost all organisms, and maintaining iron homeostasis is critical for human health. In mammals, the only known gatekeeper between intestinally absorbed iron and circulatory blood plasma is the membrane transporter ferroportin (Fpn). As such, dysfunction of Fpn or its regulation is a key driver of iron-related pathophysiology. This review focuses on discussing recent insights from high-resolution structural studies of the Fpn protein family. While these studies have unveiled crucial details of Fpn regulation and structural architecture, the associated functional studies have also at times provided conflicting data provoking more questions than answers. Here, we summarize key findings and illuminate important remaining questions and contradictions.
Collapse
Affiliation(s)
- Mika Jormakka
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
46
|
Zhang J, Huang X, Yang S, Huang A, Ren J, Luo X, Feng S, Li P, Li Z, Dong P. Endophytic Bacillus subtilis H17-16 effectively inhibits Phytophthora infestans, the pathogen of potato late blight, and its potential application. PEST MANAGEMENT SCIENCE 2023; 79:5073-5086. [PMID: 37572366 DOI: 10.1002/ps.7717] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 08/03/2023] [Accepted: 08/13/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND As a highly prevalent epidemic disease of potato, late blight caused by Phytophthora infestans poses a serious threat to potato yield and quality. At present, chemical fungicides are mainly used to control potato late blight, but long-term overuse of chemical fungicides may lead to environmental pollution and human health threats. Endophytes, natural resources for plant diseases control, can promote plant growth, enhance plant resistance, and secrete antifungal substances. Therefore, there is an urgent need to find some beneficial endophytes to control potato late blight. RESULTS We isolated a strain of Bacillus subtilis H17-16 from potato healthy roots. It can significantly inhibit mycelial growth, sporangia germination and the pathogenicity of Phytophthora infestans, induce the resistance of potato to late blight, and promote potato growth. In addition, H17-16 has the ability to produce protease, volatile compounds (VOCs) and form biofilms. After H17-16 treatment, most of the genes involved in metabolism, virulence and drug resistance of Phytophthora infestans were down-regulated significantly, and the genes related to ribosome biogenesis were mainly up-regulated. Moreover, field and postharvest application of H17-16 can effectively reduce the occurrence of potato late blight, and the combination of H17-16 with chitosan or chemical fungicides had a better effect than single H17-16. CONCLUSION Our results reveal that Bacillus subtilis H17-16 has great potential as a natural fungicide for controlling potato late blight, laying a theoretical basis for its development as a biological control agent. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jiaomei Zhang
- School of Life Sciences, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Biology and Genetic Breeding for Tuber and Root Crops, Chongqing, China
| | - Xiaoqing Huang
- School of Life Sciences, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Biology and Genetic Breeding for Tuber and Root Crops, Chongqing, China
| | - Shidong Yang
- Shandong Nongdeli Biotechnology Co., Ltd, Jinan, China
| | - Airong Huang
- School of Life Sciences, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Biology and Genetic Breeding for Tuber and Root Crops, Chongqing, China
| | - Jie Ren
- School of Life Sciences, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Biology and Genetic Breeding for Tuber and Root Crops, Chongqing, China
| | - Xunguang Luo
- School of Life Sciences, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Biology and Genetic Breeding for Tuber and Root Crops, Chongqing, China
| | - Shun Feng
- School of Life Sciences, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Biology and Genetic Breeding for Tuber and Root Crops, Chongqing, China
| | - Peihua Li
- College of Agronomy, Xichang University, Xichang, China
| | - Zhengguo Li
- School of Life Sciences, Chongqing University, Chongqing, China
- Key Laboratory of Plant Hormones and Development Regulation of Chongqing, Chongqing, China
| | - Pan Dong
- School of Life Sciences, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Biology and Genetic Breeding for Tuber and Root Crops, Chongqing, China
| |
Collapse
|
47
|
Kim JH, Mailloux L, Bloor D, Maddox B, Humble J. The role of salt bridge networks in the stability of the yeast hexose transporter 1. Biochim Biophys Acta Gen Subj 2023; 1867:130490. [PMID: 37844739 DOI: 10.1016/j.bbagen.2023.130490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/06/2023] [Accepted: 10/12/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND The yeast S. cerevisiae preferably metabolizes glucose through aerobic glycolysis. Glucose transport is facilitated by multiple hexose transporters (Hxts), and their expression and activity are tightly regulated by multiple mechanisms. However, detailed structural and functional analyses of Hxts remain limited, largely due to the lack of crystal structure. METHODS Homology modeling was used to build a 3D structural model for the yeast glucose transporter Hxt1 and investigate the effects of site directed mutations on Hxt1 stability and glucose transport activity. RESULTS The conserved salt bridge-forming residues observed in the human Glut4 and the yeast glucose receptor Rgt2 were identified within and between the two 6-transmembrane spanning segments of Hxt1. Most of the RGT2 mutations that disrupt the salt bridge networks were known to cause constitutive signal generation, whereas the corresponding substitutions in HXT1 were shown to decrease Hxt1 stability. While substitutions of the two residues in the salt bridge 2 in Glut4-E329Q and E393D-were reported to abolish glucose transport, the equivalent substitutions in Hxt1 (D382Q and E454D) did not affect Hxt1 glucose transport activity. CONCLUSIONS Substitutions of equivalent salt bridge-forming residues in Hxt1, Rgt2, and Glut4 are predicted to lock them in an inward-facing conformation but lead to different functional consequences. GENERAL SIGNIFICANCE The salt bridge networks in yeast and human glucose transporters and yeast glucose receptors may play different roles in maintaining their structural and functional integrity.
Collapse
Affiliation(s)
- Jeong-Ho Kim
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA 24502, USA.
| | - Levi Mailloux
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA 24502, USA
| | - Daniel Bloor
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA 24502, USA
| | - Bradley Maddox
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA 24502, USA
| | - Julia Humble
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA 24502, USA
| |
Collapse
|
48
|
Dorigan AF, Moreira SI, da Silva Costa Guimarães S, Cruz-Magalhães V, Alves E. Target and non-target site mechanisms of fungicide resistance and their implications for the management of crop pathogens. PEST MANAGEMENT SCIENCE 2023; 79:4731-4753. [PMID: 37592727 DOI: 10.1002/ps.7726] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/14/2023] [Accepted: 08/18/2023] [Indexed: 08/19/2023]
Abstract
Fungicides are indispensable for high-quality crops, but the rapid emergence and evolution of fungicide resistance have become the most important issues in modern agriculture. Hence, the sustainability and profitability of agricultural production have been challenged due to the limited number of fungicide chemical classes. Resistance to site-specific fungicides has principally been linked to target and non-target site mechanisms. These mechanisms change the structure or expression level, affecting fungicide efficacy and resulting in different and varying resistance levels. This review provides background information about fungicide resistance mechanisms and their implications for developing anti-resistance strategies in plant pathogens. Here, our purpose was to review changes at the target and non-target sites of quinone outside inhibitor (QoI) fungicides, methyl-benzimidazole carbamate (MBC) fungicides, demethylation inhibitor (DMI) fungicides, and succinate dehydrogenase inhibitor (SDHI) fungicides and to evaluate if they may also be associated with a fitness cost on crop pathogen populations. The current knowledge suggests that understanding fungicide resistance mechanisms can facilitate resistance monitoring and assist in developing anti-resistance strategies and new fungicide molecules to help solve this issue. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
| | | | | | | | - Eduardo Alves
- Department of Plant Pathology, Federal University of Lavras, Lavras, Brazil
| |
Collapse
|
49
|
Potenciano da Silva KL, Moraes D, Lechner B, Lindner H, Haas H, Almeida Soares CM, Silva-Bailão MG, Bailão AM. Fonsecaea pedrosoi produces ferricrocin and can utilize different host iron sources. Fungal Biol 2023; 127:1512-1523. [PMID: 38097325 DOI: 10.1016/j.funbio.2023.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 12/18/2023]
Abstract
The survival of living organisms depends on iron, one of the most abundant metals in the Earth's crust. Nevertheless, this micronutrient is poorly available in our aerobic atmosphere as well as inside the mammalian host. This problem is circumvented by the expression of high affinity iron uptake machineries, including the production of siderophores, in pathogenic fungi. Here we demonstrated that F. pedrosoi, the causative agent of the neglected tropical disease chromoblastomycosis, presents gene clusters for siderophore production. In addition, ten putative siderophore transporters were identified. Those genes are upregulated under iron starvation, a condition that induces the secretion of hydroxamates, as revealed by chrome azurol S assays. RP-HPLC and mass spectrometry analysis allowed the identification of ferricrocin as an intra- and extracellular siderophore. F. pedrosoi can grow in different iron sources, including the bacterial ferrioxamine B and the host proteins ferritin, hemoglobin and holotransferrin. Of note, addition of hemoglobin, lactoferrin and holotransferrin to the growth medium of macrophages infected with F. pedrosoi enhanced significantly fungal survival. The ability to produce siderophores in iron limited conditions added to the versatility to utilize different sources of iron are strategies that certainly may contribute to fungal survival inside the host.
Collapse
Affiliation(s)
| | - Dayane Moraes
- Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Goiás, Brazil.
| | - Beatrix Lechner
- Institute of Molecular Biology/ Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
| | - Herbert Lindner
- Institute of Medical Biochemistry/Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
| | - Hubertus Haas
- Institute of Molecular Biology/ Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
| | | | | | - Alexandre Melo Bailão
- Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Goiás, Brazil.
| |
Collapse
|
50
|
Alharake J, Bidard F, Aouam T, Sénamaud-Beaufort C, Margeot A, Heiss-Blanquet S. Effect of the res2 transcription factor gene deletion on protein secretion and stress response in the hyperproducer strain Trichoderma reesei Rut-C30. BMC Microbiol 2023; 23:374. [PMID: 38036984 PMCID: PMC10687790 DOI: 10.1186/s12866-023-03125-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND The fungus Trichoderma reesei is one of the most used industrial cellulase producers due to its high capacity of protein secretion. Strains of T. reesei with enhanced protein secretion capacity, such as Rut-C30, have been obtained after several rounds of random mutagenesis. The strain was shown to possess an expanded endoplasmic reticulum, but the genetic factors responsible for this phenotype remain still unidentified. Recently, three new transcription factors were described in Neurospora crassa which were demonstrated to be involved in protein secretion. One of them, RES2, was involved in upregulation of secretion-related genes. The aim of our present study was therefore to analyze the role of RES2, on protein secretion in the T. reesei Rut-C30 strain. RESULT Deletion of the res2 gene in Rut-C30 resulted in slightly slower growth on all substrates tested, and lower germination rate as well as lower protein secretion compared to the parental strain Rut-C30. Transcriptomic analysis of the Rut-C30 and the Δres2 mutant strain in secretion stress conditions showed remarkably few differences : 971 genes were differentially expressed (DE) in both strains while 192 genes out of 1163 (~ 16.5%) were DE in Rut-C30 only and 693 out of 1664 genes (~ 41.6%) displayed differential expression solely in Δres2. Notably, induction of protein secretion by cultivating on lactose and addition of secretion stress inducer DTT induced many genes of the secretion pathway similarly in both strains. Among the differentially expressed genes, those coding for amino acid biosynthesis genes, transporters and genes involved in lipid metabolism were found to be enriched specifically in the Δres2 strain upon exposure to lactose or DTT. Besides, redox homeostasis and DNA repair genes were specifically upregulated in the Δres2 strain, indicating an altered stress response. CONCLUSION These results indicate that in the T. reesei Rut-C30 strain, RES2 does not act as a master regulator of the secretion pathway, but it contributes to a higher protein secretion by adjusting the expression of genes involved in different steps of protein synthesis and the secretion pathway.
Collapse
Affiliation(s)
- Jawad Alharake
- IFP Energies Nouvelles, 1 et 4, avenue de Bois-Préau, Rueil-Malmaison Cedex, 92852, France
| | - Frédérique Bidard
- IFP Energies Nouvelles, 1 et 4, avenue de Bois-Préau, Rueil-Malmaison Cedex, 92852, France
| | - Thiziri Aouam
- IFP Energies Nouvelles, 1 et 4, avenue de Bois-Préau, Rueil-Malmaison Cedex, 92852, France
| | - Catherine Sénamaud-Beaufort
- Département de biologie, GenomiqueENS, Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Université PSL, École normale supérieure, Paris, 75005, France
| | - Antoine Margeot
- IFP Energies Nouvelles, 1 et 4, avenue de Bois-Préau, Rueil-Malmaison Cedex, 92852, France
| | - Senta Heiss-Blanquet
- IFP Energies Nouvelles, 1 et 4, avenue de Bois-Préau, Rueil-Malmaison Cedex, 92852, France.
| |
Collapse
|