1
|
My R, Gupte AP, Bizzotto E, Frizzarin M, Antoniali P, Campanaro S, Favaro L. Unveiling the fitness of Saccharomyces cerevisiae strains for lignocellulosic bioethanol: a genomic exploration through fermentation stress tests. N Biotechnol 2025; 85:63-74. [PMID: 39675422 DOI: 10.1016/j.nbt.2024.12.004] [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: 06/17/2024] [Revised: 12/02/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Lignocellulosic biomass holds significant promise as a substrate for bioethanol production, yet the financial viability of lignocellulosic fermentation poses challenges. The pre-treatment step needed for lignocellulosic substrates generates inhibitors that impede Saccharomyces cerevisiae growth, affecting the fermentation process and overall yield. In modern sugarcane-to-ethanol plants, a rapid succession of yeast strains occurs, with dominant strains prevailing. Therefore, yeast strains with both dominance potential and inhibitor tolerance are crucial towards the development of superior strains with industrial fitness. This study adopted a hybrid approach combining biotechnology and bioinformatics to explore a cluster of 20 S. cerevisiae strains, including industrial and oenological strains exhibiting diverse phenotypic features. In-depth genomic analyses focusing on gene copy number variations (CNVs) and single nucleotide polymorphisms (SNPs) were conducted and compared with results from fermentation tests once inoculated in multiple strains kinetics under stressing conditions such as low nitrogen availability and high formic or acetic acid levels. Some strains showed high resistance to biotic stress and acetic acid. Moreover, four out of 20 strains - namely S. cerevisiae YI30, Fp89, Fp90 and CESPLG05 - displayed promising resistance also to formic acid, the most impactful weak acids in pre-treated lignocellulosic biomass. These strains have the potential to be used for the development of superior S. cerevisiae strains tailored for lignocellulosic bioethanol production.
Collapse
Affiliation(s)
- Rebecca My
- Department of Agronomy, Food, Natural resources, Animals and the Environment (DAFNAE), University of Padova, Agripolis, Legnaro 35020, Italy
| | - Ameya Pankaj Gupte
- Department of Agronomy, Food, Natural resources, Animals and the Environment (DAFNAE), University of Padova, Agripolis, Legnaro 35020, Italy
| | - Edoardo Bizzotto
- Department of Biology, University of Padova, Padova 35131, Italy
| | | | | | | | - Lorenzo Favaro
- Department of Agronomy, Food, Natural resources, Animals and the Environment (DAFNAE), University of Padova, Agripolis, Legnaro 35020, Italy; Department of Microbiology, Stellenbosch University, Private Bag X1, South Africa 7602, South Africa.
| |
Collapse
|
2
|
Yu H, Wang Y, Wang Y, Niu Y, Zhou J, Li J, Chen J, Du G, Zhao X. Metabolic engineering of yeast to efficiently synthesize heme and hemoproteins: recent advance and prospects. FEMS Yeast Res 2025; 25:foaf019. [PMID: 40228812 PMCID: PMC12020473 DOI: 10.1093/femsyr/foaf019] [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: 01/30/2025] [Revised: 03/30/2025] [Accepted: 04/12/2025] [Indexed: 04/16/2025] Open
Abstract
Owing to the potential for commercialization, the recombinant production of hemoproteins has been heavily investigated. Yeast is a superior host for the synthesis of eukaryotic hemoproteins with optimal pathway to facilitate heme delivery and utilization, as well as suitable environment for the post-translational folding and modification. The efficient binding of heme is the critical determinant for the various functions of hemeproteins. Thus, many metabolic engineering strategies have been employed to modify heme synthetic pathways and balance the intracellular metabolic burden. This paper provides a comprehensive review on the improvement of heme supply, the enhancement of hemoprotein expression, and the current efforts to harmonize the synthesis of heme and the expression of protein components in yeast. These insights offer a solid foundation for the development of yeast chassis for the efficient production of high-active hemoproteins in the future.
Collapse
Affiliation(s)
- Haibo Yu
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yunpeng Wang
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yijie Wang
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yueheng Niu
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Jingwen Zhou
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Jianghua Li
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Jian Chen
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Guocheng Du
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Xinrui Zhao
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| |
Collapse
|
3
|
Ribeiro MO, Oliveira M, Nogueira V, Costa V, Teixeira V. N88S seipin-related seipinopathy is a lipidopathy associated with loss of iron homeostasis. Cell Commun Signal 2025; 23:10. [PMID: 39773523 PMCID: PMC11706183 DOI: 10.1186/s12964-024-02007-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Seipin is a protein encoded by the BSCL2 gene in humans and SEI1 gene in yeast, forming an Endoplasmic Reticulum (ER)-bound homo-oligomer. This oligomer is crucial in targeting ER-lipid droplet (LD) contact sites, facilitating the delivery of triacylglycerol (TG) to nascent LDs. Mutations in BSCL2, particularly N88S and S90L, lead to seipinopathies, which correspond to a cohort of motor neuron diseases (MNDs) characterized by the accumulation of misfolded N88S seipin into inclusion bodies (IBs) and cellular dysfunctions. METHODS Quantitative untargeted mass spectrometric proteomic and lipidomic analyses were conducted to examine changes in protein and lipid abundance in wild-type (WT) versus N88S seipin-expressing mutant cells. Differentially expressed proteins were categorized into functional networks to highlight altered protein functions and signaling pathways. Statistical comparisons were made using unpaired Student's t-tests or two-way ANOVA followed by Tukey´s / Šídák's multiple comparisons tests. P-values < 0.05 are considered significant. RESULTS In a well-established yeast model of N88S seipinopathy, misfolded N88S seipin forms IBs and exhibits higher levels of ER stress, leading to decreased cell viability due to increased reactive oxygen species (ROS), oxidative damage, lipid peroxidation, and reduced antioxidant activity. Proteomic and lipidomic analyses revealed alterations in phosphatidic acid (PA) levels, associated with disrupted inositol metabolism and decreased flux towards phospholipid biosynthesis. Importantly, deregulation of lipid metabolism contributed to ER stress beyond N88S seipin misfolding and IB formation. Additionally, the model exhibited deregulated iron (Fe) homeostasis during lifespan. N88S seipin-expressing cells showed impaired ability to cope with iron deficiency. This was linked to changes in the expression of Aft1p-controlled iron regulon genes, including the mRNA-binding protein CTH2 and the high-affinity iron transport system member FET3, in a p38/Hog1p- and Msn2p/Msn4p-dependent manner. Importantly, we unraveled a novel link between inositol metabolism and activation of the iron regulon in cells expressing the N88S seipin mutation. Despite iron accumulation, this was not associated with oxidative stress. CONCLUSIONS The study highlights that the effects of N88S seipin mutation extend beyond protein misfolding, with significant disruptions in lipid metabolism and iron homeostasis. This research marks a substantial advance in understanding and defining the roles of proteins and signaling pathways that contribute to human seipinopathy. Altered cellular processes, as well as potential therapeutic targets and biomarkers, were identified and can be explored in translational studies using human cell models.
Collapse
Affiliation(s)
- Mariana O Ribeiro
- IBMC - Instituto de Biologia Molecular E Celular, University of Porto, Porto, Portugal
- Yeast Signalling Networks (YSN), i3S - Instituto de Investigação E Inovação Em Saúde, University of Porto, Porto, Portugal
| | - Mafalda Oliveira
- IBMC - Instituto de Biologia Molecular E Celular, University of Porto, Porto, Portugal
- Yeast Signalling Networks (YSN), i3S - Instituto de Investigação E Inovação Em Saúde, University of Porto, Porto, Portugal
| | - Verónica Nogueira
- IBMC - Instituto de Biologia Molecular E Celular, University of Porto, Porto, Portugal
- Yeast Signalling Networks (YSN), i3S - Instituto de Investigação E Inovação Em Saúde, University of Porto, Porto, Portugal
| | - Vítor Costa
- IBMC - Instituto de Biologia Molecular E Celular, University of Porto, Porto, Portugal
- Yeast Signalling Networks (YSN), i3S - Instituto de Investigação E Inovação Em Saúde, University of Porto, Porto, Portugal
- Department of Molecular Biology, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, School of Medicine and Biomedical Sciences, Universidade Do Porto, Porto, Portugal
| | - Vitor Teixeira
- IBMC - Instituto de Biologia Molecular E Celular, University of Porto, Porto, Portugal.
- Yeast Signalling Networks (YSN), i3S - Instituto de Investigação E Inovação Em Saúde, University of Porto, Porto, Portugal.
| |
Collapse
|
4
|
Drozdova P, Gurkov A, Saranchina A, Vlasevskaya A, Zolotovskaya E, Indosova E, Timofeyev M, Borvinskaya E. Transcriptional response of Saccharomyces cerevisiae to lactic acid enantiomers. Appl Microbiol Biotechnol 2024; 108:121. [PMID: 38229303 PMCID: PMC10787881 DOI: 10.1007/s00253-023-12863-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/16/2023] [Accepted: 10/24/2023] [Indexed: 01/18/2024]
Abstract
The model yeast, Saccharomyces cerevisiae, is a popular object for both fundamental and applied research, including the development of biosensors and industrial production of pharmaceutical compounds. However, despite multiple studies exploring S. cerevisiae transcriptional response to various substances, this response is unknown for some substances produced in yeast, such as D-lactic acid (DLA). Here, we explore the transcriptional response of the BY4742 strain to a wide range of DLA concentrations (from 0.05 to 45 mM), and compare it to the response to 45 mM L-lactic acid (LLA). We recorded a response to 5 and 45 mM DLA (125 and 113 differentially expressed genes (DEGs), respectively; > 50% shared) and a less pronounced response to 45 mM LLA (63 DEGs; > 30% shared with at least one DLA treatment). Our data did not reveal natural yeast promoters quantitatively sensing DLA but provide the first description of the transcriptome-wide response to DLA and enrich our understanding of the LLA response. Some DLA-activated genes were indeed related to lactate metabolism, as well as iron uptake and cell wall structure. Additional analyses showed that at least some of these genes were activated only by acidic form of DLA but not its salt, revealing the role of pH. The list of LLA-responsive genes was similar to those published previously and also included iron uptake and cell wall genes, as well as genes responding to other weak acids. These data might be instrumental for optimization of lactate production in yeast and yeast co-cultivation with lactic acid bacteria. KEY POINTS: • We present the first dataset on yeast transcriptional response to DLA. • Differential gene expression was correlated with yeast growth inhibition. • The transcriptome response to DLA was richer in comparison to LLA.
Collapse
Affiliation(s)
- Polina Drozdova
- Irkutsk State University, Karl-Marx Str. 1, Irkutsk, 664025, Russian Federation.
- Baikal Research Centre, Rabochaya Str. 5V, Irkutsk, 664011, Russian Federation.
| | - Anton Gurkov
- Irkutsk State University, Karl-Marx Str. 1, Irkutsk, 664025, Russian Federation
- Baikal Research Centre, Rabochaya Str. 5V, Irkutsk, 664011, Russian Federation
| | | | | | - Elena Zolotovskaya
- Irkutsk State University, Karl-Marx Str. 1, Irkutsk, 664025, Russian Federation
| | - Elizaveta Indosova
- Irkutsk State University, Karl-Marx Str. 1, Irkutsk, 664025, Russian Federation
| | - Maxim Timofeyev
- Irkutsk State University, Karl-Marx Str. 1, Irkutsk, 664025, Russian Federation
| | | |
Collapse
|
5
|
Weyer Y, Schwabl SI, Tang X, Purwar A, Siegmann K, Ruepp A, Dunzendorfer-Matt T, Widerin MA, Niedrist V, Mutsters NJM, Tettamanti MG, Weys S, Sarg B, Kremser L, Liedl KR, Schmidt O, Teis D. The Dsc ubiquitin ligase complex identifies transmembrane degrons to degrade orphaned proteins at the Golgi. Nat Commun 2024; 15:9257. [PMID: 39461958 PMCID: PMC11513148 DOI: 10.1038/s41467-024-53676-6] [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: 03/24/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
The Golgi apparatus is essential for protein sorting, yet its quality control mechanisms are poorly understood. Here we show that the Dsc ubiquitin ligase complex uses its rhomboid pseudo-protease subunit, Dsc2, to assess the hydrophobic length of α-helical transmembrane domains (TMDs) at the Golgi. Thereby the Dsc complex likely interacts with orphaned ER and Golgi proteins that have shorter TMDs and ubiquitinates them for targeted degradation. Some Dsc substrates will be extracted by Cdc48 for endosome and Golgi associated proteasomal degradation (EGAD), while others will undergo ESCRT dependent vacuolar degradation. Some substrates are degraded by both, EGAD- or ESCRT pathways. The accumulation of Dsc substrates entails a specific increase in glycerophospholipids with shorter and asymmetric fatty acyl chains. Hence, the Dsc complex mediates the selective degradation of orphaned proteins at the sorting center of cells, which prevents their spreading across other organelles and thereby preserves cellular membrane protein and lipid composition.
Collapse
Affiliation(s)
- Yannick Weyer
- Institute of Molecular Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Sinead I Schwabl
- Institute of Molecular Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Xuechen Tang
- Department of General, Inorganic and Theoretical Chemistry, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Astha Purwar
- Institute of Molecular Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Konstantin Siegmann
- Institute of Molecular Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Angela Ruepp
- Institute of Molecular Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Michael A Widerin
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Veronika Niedrist
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Noa J M Mutsters
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Maria G Tettamanti
- Department of Molecular and Cell Biology, University of Geneva, Geneva, Switzerland
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Sabine Weys
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
- Institute of Science and Technology Austria (ISTA), Am Campus 1, Klosterneuburg, Austria
| | - Bettina Sarg
- Institute of Medical Biochemistry, Protein Core Facility, Medical University of Innsbruck, Innsbruck, Austria
| | - Leopold Kremser
- Institute of Medical Biochemistry, Protein Core Facility, Medical University of Innsbruck, Innsbruck, Austria
| | - Klaus R Liedl
- Institute of Medical Biochemistry, Protein Core Facility, Medical University of Innsbruck, Innsbruck, Austria
| | - Oliver Schmidt
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - David Teis
- Institute of Molecular Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
6
|
Lee HJ, Shin DJ, Nho SB, Lee KW, Kim SK. Metabolic Engineering of Saccharomyces cerevisiae for Fermentative Production of Heme. Biotechnol J 2024; 19:e202400351. [PMID: 39380497 DOI: 10.1002/biot.202400351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 10/10/2024]
Abstract
Heme is a key ingredient required to mimic the color and flavor of meat in plant-based alternatives. This study aimed to develop a yeast-based microbial cell factory for efficient and sustainable production of heme. To this end, first, Hem12p (uroporphyrinogen decarboxylase) was identified as the rate-limiting enzyme in the heme biosynthetic pathway present in Saccharomyces cerevisiae D452-2. Next, we investigated the effects of disruption of the genes involved in the competition for heme biosynthesis precursors, transcriptional repression, and heme degradation (HMX1) on heme production efficiency. Of the knock-out strains constructed in this study, only the HMX1-deficient strain produced heme at a higher concentration than the background strain without gene disruption. In addition, overexpression of PUG1 encoding a plasma membrane transporter involved in protoporphyrin IX (the precursor to heme biosynthesis) uptake led to a significant increase in intracellular heme concentration. As a result, among the various engineered strains constructed in this study, the ΔHMX1/H3&12 + PUG1 strain, the HMX1-deficient strain overexpressing HEM3, HEM12, and PUG1, produced the highest concentration of heme (4.6 mg/L) in batch fermentation, which was 3.9-fold higher than that produced by the wild-type D452-2 strain. In a glucose-limited fed-batch fermentation, the ΔHMX1/H3&12 + PUG1 strain produced 28 mg/L heme in 66 h.
Collapse
Affiliation(s)
- Hyun-Jae Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, Gyeonggi, Republic of Korea
- GreenTech-based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong, Gyeonggi, Republic of Korea
| | - Dong Joo Shin
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Soo Bin Nho
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, Gyeonggi, Republic of Korea
- GreenTech-based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong, Gyeonggi, Republic of Korea
| | - Ki Won Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Sun-Ki Kim
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, Gyeonggi, Republic of Korea
- GreenTech-based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong, Gyeonggi, Republic of Korea
| |
Collapse
|
7
|
Tang J, Chen Y, Li Q, Xin W, Xiao X, Chen X, Yang L, Mou B, Li J, Lu F, Fu C, Long W, Liao H, Han X, Feng P, Li W, Zhou K, Yang L, Yang Y, Ma M, Wang H. The response mechanism analysis of HMX1 knockout strain to levulinic acid in Saccharomyces cerevisiae. Front Microbiol 2024; 15:1416903. [PMID: 38989024 PMCID: PMC11233763 DOI: 10.3389/fmicb.2024.1416903] [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] [Received: 04/13/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Levulinic acid, a hydrolysis product of lignocellulose, can be metabolized into important compounds in the field of medicine and pesticides by engineered strains of Saccharomyces cerevisiae. Levulinic acid, as an intermediate product widely found in the conversion process of lignocellulosic biomass, has multiple applications. However, its toxicity to Saccharomyces cerevisiae reduces its conversion efficiency, so screening Saccharomyces cerevisiae genes that can tolerate levulinic acid becomes the key. By creating a whole-genome knockout library and bioinformatics analysis, this study used the phenotypic characteristics of cells as the basis for screening and found the HMX1 gene that is highly sensitive to levulinic acid in the oxidative stress pathway. After knocking out HMX1 and treating with levulinic acid, the omics data of the strain revealed that multiple affected pathways, especially the expression of 14 genes related to the cell wall and membrane system, were significantly downregulated. The levels of acetyl-CoA and riboflavin decreased by 1.02-fold and 1.44-fold, respectively, while the content of pantothenic acid increased. These findings indicate that the cell wall-membrane system, as well as the metabolism of acetyl-CoA and riboflavin, are important in improving the resistance of Saccharomyces cerevisiae to levulinic acid. They provide theoretical support for enhancing the tolerance of microorganisms to levulinic acid, which is significant for optimizing the conversion process of lignocellulosic biomass to levulinic acid.
Collapse
Affiliation(s)
- Jiaye Tang
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
- College of Resources, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yulei Chen
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
- College of Resources, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qian Li
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
- College of Resources, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Wenli Xin
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| | - Ximeng Xiao
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| | - Xuemei Chen
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| | - Lixi Yang
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| | - Borui Mou
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| | - Jialian Li
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| | - Fujia Lu
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| | - Chun Fu
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| | - Wencong Long
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| | - Hong Liao
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| | - Xuebing Han
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| | - Peng Feng
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| | - Wei Li
- Aba Prefecture Ecological Protection and Development Research Institute, Wenchuan, Sichuan, China
| | - Kedi Zhou
- Institute of Nature Conservation Area Planning, Sichuan Forestry and Grassland Survey and Planning Institute, Chengdu, Sichuan, China
| | - Liuyun Yang
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| | - Yaojun Yang
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| | - Menggen Ma
- College of Resources, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hanyu Wang
- Bamboo Diseases and Pests Control and Resources Development Key Laboratory of Sichuan Province, College of Life Science, Leshan Normal University, Leshan, Sichuan, China
| |
Collapse
|
8
|
Pijuan J, Moreno DF, Yahya G, Moisa M, Ul Haq I, Krukiewicz K, Mosbah R, Metwally K, Cavalu S. Regulatory and pathogenic mechanisms in response to iron deficiency and excess in fungi. Microb Biotechnol 2023; 16:2053-2071. [PMID: 37804207 PMCID: PMC10616654 DOI: 10.1111/1751-7915.14346] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/09/2023] Open
Abstract
Iron is an essential element for all eukaryote organisms because of its redox properties, which are important for many biological processes such as DNA synthesis, mitochondrial respiration, oxygen transport, lipid, and carbon metabolism. For this reason, living organisms have developed different strategies and mechanisms to optimally regulate iron acquisition, transport, storage, and uptake in different environmental responses. Moreover, iron plays an essential role during microbial infections. Saccharomyces cerevisiae has been of key importance for decrypting iron homeostasis and regulation mechanisms in eukaryotes. Specifically, the transcription factors Aft1/Aft2 and Yap5 regulate the expression of genes to control iron metabolism in response to its deficiency or excess, adapting to the cell's iron requirements and its availability in the environment. We also review which iron-related virulence factors have the most common fungal human pathogens (Aspergillus fumigatus, Cryptococcus neoformans, and Candida albicans). These factors are essential for adaptation in different host niches during pathogenesis, including different fungal-specific iron-uptake mechanisms. While being necessary for virulence, they provide hope for developing novel antifungal treatments, which are currently scarce and usually toxic for patients. In this review, we provide a compilation of the current knowledge about the metabolic response to iron deficiency and excess in fungi.
Collapse
Affiliation(s)
- Jordi Pijuan
- Laboratory of Neurogenetics and Molecular MedicineInstitut de Recerca Sant Joan de DéuBarcelonaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIIIMadridSpain
| | - David F. Moreno
- Department of Molecular Cellular and Developmental BiologyYale UniversityNew HavenConnecticutUSA
- Systems Biology InstituteYale UniversityWest HavenConnecticutUSA
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of PharmacyZagazig UniversityAl SharqiaEgypt
| | - Mihaela Moisa
- Faculty of Medicine and PharmacyUniversity of OradeaOradeaRomania
| | - Ihtisham Ul Haq
- Department of Physical Chemistry and Polymers TechnologySilesian University of TechnologyGliwicePoland
- Programa de Pós‐graduação em Inovação TecnológicaUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Katarzyna Krukiewicz
- Department of Physical Chemistry and Polymers TechnologySilesian University of TechnologyGliwicePoland
- Centre for Organic and Nanohybrid ElectronicsSilesian University of TechnologyGliwicePoland
| | - Rasha Mosbah
- Infection Control UnitHospitals of Zagazig UniversityZagazigEgypt
| | - Kamel Metwally
- Department of Medicinal Chemistry, Faculty of PharmacyUniversity of TabukTabukSaudi Arabia
- Department of Pharmaceutical Medicinal Chemistry, Faculty of PharmacyZagazig UniversityZagazigEgypt
| | - Simona Cavalu
- Faculty of Medicine and PharmacyUniversity of OradeaOradeaRomania
| |
Collapse
|
9
|
Pentjuss A, Bolmanis E, Suleiko A, Didrihsone E, Suleiko A, Dubencovs K, Liepins J, Kazaks A, Vanags J. Pichia pastoris growth-coupled heme biosynthesis analysis using metabolic modelling. Sci Rep 2023; 13:15816. [PMID: 37739976 PMCID: PMC10516909 DOI: 10.1038/s41598-023-42865-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023] Open
Abstract
Soy leghemoglobin is one of the most important and key ingredients in plant-based meat substitutes that can imitate the colour and flavour of the meat. To improve the high-yield production of leghemoglobin protein and its main component-heme in the yeast Pichia pastoris, glycerol and methanol cultivation conditions were studied. Additionally, in-silico metabolic modelling analysis of growth-coupled enzyme quantity, suggests metabolic gene up/down-regulation strategies for heme production. First, cultivations and metabolic modelling analysis of P. pastoris were performed on glycerol and methanol in different growth media. Glycerol cultivation uptake and production rates can be increased by 50% according to metabolic modelling results, but methanol cultivation-is near the theoretical maximum. Growth-coupled metabolic optimisation results revealed the best feasible upregulation (33 reactions) (1.47% of total reactions) and 66 downregulation/deletion (2.98% of total) reaction suggestions. Finally, we describe reaction regulation suggestions with the highest potential to increase heme production yields.
Collapse
Affiliation(s)
- Agris Pentjuss
- Microbiology and Biotechnology Institute, University of Latvia, Jelgavas Street 1, Riga, 1004, Latvia.
- Latvian State Institute of Wood Chemistry, Dzerbenes Street 27, Riga, 1006, Latvia.
| | - Emils Bolmanis
- Latvian Biomedical Research and Study Centre, Ratsupites Street 1 K-1, Riga, 1067, Latvia
| | - Anastasija Suleiko
- Latvian State Institute of Wood Chemistry, Dzerbenes Street 27, Riga, 1006, Latvia
- Bioreactors.Net AS, Dzerbenes Street 27, Riga, 1006, Latvia
| | - Elina Didrihsone
- Latvian State Institute of Wood Chemistry, Dzerbenes Street 27, Riga, 1006, Latvia
| | - Arturs Suleiko
- Latvian State Institute of Wood Chemistry, Dzerbenes Street 27, Riga, 1006, Latvia
- Bioreactors.Net AS, Dzerbenes Street 27, Riga, 1006, Latvia
| | - Konstantins Dubencovs
- Latvian State Institute of Wood Chemistry, Dzerbenes Street 27, Riga, 1006, Latvia
- Bioreactors.Net AS, Dzerbenes Street 27, Riga, 1006, Latvia
| | - Janis Liepins
- Microbiology and Biotechnology Institute, University of Latvia, Jelgavas Street 1, Riga, 1004, Latvia
| | - Andris Kazaks
- Latvian Biomedical Research and Study Centre, Ratsupites Street 1 K-1, Riga, 1067, Latvia
| | - Juris Vanags
- Latvian State Institute of Wood Chemistry, Dzerbenes Street 27, Riga, 1006, Latvia
- Bioreactors.Net AS, Dzerbenes Street 27, Riga, 1006, Latvia
| |
Collapse
|
10
|
Xue J, Zhou J, Li J, Du G, Chen J, Wang M, Zhao X. Systematic engineering of Saccharomyces cerevisiae for efficient synthesis of hemoglobins and myoglobins. BIORESOURCE TECHNOLOGY 2023; 370:128556. [PMID: 36586429 DOI: 10.1016/j.biortech.2022.128556] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 05/26/2023]
Abstract
Hemoglobin (Hb) and myoglobin (Mb) are kinds of heme-binding proteins that play crucial physiological roles in different organisms. With rapid application development in food processing and biocatalysis, the requirement of biosynthetic Hb and Mb is increasing. However, the production of Hb and Mb is limited by the lower expressional level of globins and insufficient or improper heme supply. After selecting an inducible strategy for the expression of globins, removing the spatial barrier during heme synthesis, increasing the synthesis of 5-aminolevulinate and moderately enhancing heme synthetic rate-limiting steps, the microbial synthesis of bovine and porcine Hb was firstly achieved. Furthermore, an engineered Saccharomyces cerevisiae obtained a higher titer of soybean (108.2 ± 3.5 mg/L) and clover (13.7 ± 0.5 mg/L) Hb and bovine (68.9 ± 1.6 mg/L) and porcine (85.9 ± 5.0 mg/L) Mb. Therefore, this systematic engineering strategy will be useful to produce other hemoproteins or hemoenzymes with high activities.
Collapse
Affiliation(s)
- Jike Xue
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Jingwen Zhou
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China; Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Jianghua Li
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China; Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Guocheng Du
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China; Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Jian Chen
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China; Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Miao Wang
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Xinrui Zhao
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China; Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
11
|
Lindahl PA, Vali SW. Mössbauer-based molecular-level decomposition of the Saccharomyces cerevisiae ironome, and preliminary characterization of isolated nuclei. Metallomics 2022; 14:mfac080. [PMID: 36214417 PMCID: PMC9624242 DOI: 10.1093/mtomcs/mfac080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/23/2022] [Indexed: 11/25/2022]
Abstract
One hundred proteins in Saccharomyces cerevisiae are known to contain iron. These proteins are found mainly in mitochondria, cytosol, nuclei, endoplasmic reticula, and vacuoles. Cells also contain non-proteinaceous low-molecular-mass labile iron pools (LFePs). How each molecular iron species interacts on the cellular or systems' level is underdeveloped as doing so would require considering the entire iron content of the cell-the ironome. In this paper, Mössbauer (MB) spectroscopy was used to probe the ironome of yeast. MB spectra of whole cells and isolated organelles were predicted by summing the spectral contribution of each iron-containing species in the cell. Simulations required input from published proteomics and microscopy data, as well as from previous spectroscopic and redox characterization of individual iron-containing proteins. Composite simulations were compared to experimentally determined spectra. Simulated MB spectra of non-proteinaceous iron pools in the cell were assumed to account for major differences between simulated and experimental spectra of whole cells and isolated mitochondria and vacuoles. Nuclei were predicted to contain ∼30 μM iron, mostly in the form of [Fe4S4] clusters. This was experimentally confirmed by isolating nuclei from 57Fe-enriched cells and obtaining the first MB spectra of the organelle. This study provides the first semi-quantitative estimate of all concentrations of iron-containing proteins and non-proteinaceous species in yeast, as well as a novel approach to spectroscopically characterizing LFePs.
Collapse
Affiliation(s)
- Paul A Lindahl
- Department of Chemistry, Texas A&M University, College Station, TX, USA
- Department of Biochemistry and Biophysics, Texas A&M University, College Station TX, USA
| | - Shaik Waseem Vali
- Department of Chemistry, Texas A&M University, College Station, TX, USA
| |
Collapse
|
12
|
Genome-scale modeling drives 70-fold improvement of intracellular heme production in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A 2022; 119:e2108245119. [PMID: 35858410 PMCID: PMC9335255 DOI: 10.1073/pnas.2108245119] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Heme availability in the cell enables the proper folding and function of enzymes, which carry heme as a cofactor. Using genome-scale modeling, we identified metabolic fluxes and genes that limit heme production. Our study experimentally validates ecYeast8 model predictions. Moreover, we developed an approach to predict gene combinations, which provides an in silico design of a viable strain able to overproduce the metabolite of interest. Using our approach, we constructed a yeast strain that produces 70-fold-higher levels of intracellular heme. With its high-capacity metabolic subnetwork, our engineered strain is a suitable platform for the production of additional heme enzymes. The heme ligand-binding biosensor (Heme-LBB) detects the cotranslational incorporation of heme into the heme-protein hemoglobin. Heme is an oxygen carrier and a cofactor of both industrial enzymes and food additives. The intracellular level of free heme is low, which limits the synthesis of heme proteins. Therefore, increasing heme synthesis allows an increased production of heme proteins. Using the genome-scale metabolic model (GEM) Yeast8 for the yeast Saccharomyces cerevisiae, we identified fluxes potentially important to heme synthesis. With this model, in silico simulations highlighted 84 gene targets for balancing biomass and increasing heme production. Of those identified, 76 genes were individually deleted or overexpressed in experiments. Empirically, 40 genes individually increased heme production (up to threefold). Heme was increased by modifying target genes, which not only included the genes involved in heme biosynthesis, but also those involved in glycolysis, pyruvate, Fe-S clusters, glycine, and succinyl-coenzyme A (CoA) metabolism. Next, we developed an algorithmic method for predicting an optimal combination of these genes by using the enzyme-constrained extension of the Yeast8 model, ecYeast8. The computationally identified combination for enhanced heme production was evaluated using the heme ligand-binding biosensor (Heme-LBB). The positive targets were combined using CRISPR-Cas9 in the yeast strain (IMX581-HEM15-HEM14-HEM3-Δshm1-HEM2-Δhmx1-FET4-Δgcv2-HEM1-Δgcv1-HEM13), which produces 70-fold-higher levels of intracellular heme.
Collapse
|
13
|
Imre A, Kovács R, Tóth Z, Majoros L, Benkő Z, Pfliegler WP, Pócsi I. Heme Oxygenase-1 ( HMX1) Loss of Function Increases the In-Host Fitness of the Saccharomyces 'boulardii' Probiotic Yeast in a Mouse Fungemia Model. J Fungi (Basel) 2022; 8:522. [PMID: 35628777 PMCID: PMC9146039 DOI: 10.3390/jof8050522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023] Open
Abstract
The use of yeast-containing probiotics is on the rise; however, these products occasionally cause fungal infections and possibly even fungemia among susceptible probiotic-treated patients. The incidence of such cases is probably underestimated, which is why it is important to delve deeper into the pathomechanism and the adaptive features of S. ‘boulardii’. Here in this study, the potential role of the gene heme oxygenase-1 (HMX1) in probiotic yeast bloodstream-derived infections was studied by generating marker-free HMX1 deletion mutants with CRISPR/Cas9 technology from both commercial and clinical S. ‘boulardii’ isolates. The six commercial and clinical yeasts used here represented closely related but different genetic backgrounds as revealed by comparative genomic analysis. We compared the wild-type isolates against deletion mutants for their tolerance of iron starvation, hemolytic activity, as well as kidney burden in immunosuppressed BALB/c mice after lateral tail vein injection. Our results reveal that the lack of HMX1 in S. ‘boulardii’ significantly (p < 0.0001) increases the kidney burden of the mice in most genetic backgrounds, while at the same time causes decreased growth in iron-deprived media in vitro. These findings indicate that even a single-gene loss-of-function mutation can, surprisingly, cause elevated fitness in the host during an opportunistic systemic infection. Our findings indicate that the safety assessment of S. ‘boulardii’ strains should not only take strain-to-strain variation into account, but also avoid extrapolating in vitro results to in vivo virulence factor determination.
Collapse
Affiliation(s)
- Alexandra Imre
- Department of Molecular Biotechnology and Microbiology, University of Debrecen, Egyetem tér 1., H4032 Debrecen, Hungary; (A.I.); (Z.B.); (W.P.P.)
- Kálmán Laki Doctoral School of Biomedical and Clinical Sciences, University of Debrecen, Egyetem tér 1., H4032 Debrecen, Hungary
| | - Renátó Kovács
- Department of Medical Microbiology, University of Debrecen, Egyetem tér 1., H4032 Debrecen, Hungary; (R.K.); (Z.T.); (L.M.)
- Faculty of Pharmacy, University of Debrecen, Egyetem tér 1., H4032 Debrecen, Hungary
| | - Zoltán Tóth
- Department of Medical Microbiology, University of Debrecen, Egyetem tér 1., H4032 Debrecen, Hungary; (R.K.); (Z.T.); (L.M.)
| | - László Majoros
- Department of Medical Microbiology, University of Debrecen, Egyetem tér 1., H4032 Debrecen, Hungary; (R.K.); (Z.T.); (L.M.)
| | - Zsigmond Benkő
- Department of Molecular Biotechnology and Microbiology, University of Debrecen, Egyetem tér 1., H4032 Debrecen, Hungary; (A.I.); (Z.B.); (W.P.P.)
| | - Walter P. Pfliegler
- Department of Molecular Biotechnology and Microbiology, University of Debrecen, Egyetem tér 1., H4032 Debrecen, Hungary; (A.I.); (Z.B.); (W.P.P.)
| | - István Pócsi
- Department of Molecular Biotechnology and Microbiology, University of Debrecen, Egyetem tér 1., H4032 Debrecen, Hungary; (A.I.); (Z.B.); (W.P.P.)
| |
Collapse
|
14
|
Chen X, Li X, Ji B, Wang Y, Ishchuk OP, Vorontsov E, Petranovic D, Siewers V, Engqvist MK. Suppressors of amyloid-β toxicity improve recombinant protein production in yeast by reducing oxidative stress and tuning cellular metabolism. Metab Eng 2022; 72:311-324. [DOI: 10.1016/j.ymben.2022.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 12/24/2022]
|
15
|
Pérez ALA, Piva LC, Fulber JPC, de Moraes LMP, De Marco JL, Vieira HLA, Coelho CM, Reis VCB, Torres FAG. Optogenetic strategies for the control of gene expression in yeasts. Biotechnol Adv 2021; 54:107839. [PMID: 34592347 DOI: 10.1016/j.biotechadv.2021.107839] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 09/07/2021] [Accepted: 09/22/2021] [Indexed: 12/18/2022]
Abstract
Optogenetics involves the use of light to control cellular functions and has become increasingly popular in various areas of research, especially in the precise control of gene expression. While this technology is already well established in neurobiology and basic research, its use in bioprocess development is still emerging. Some optogenetic switches have been implemented in yeasts for different purposes, taking advantage of a wide repertoire of biological parts and relatively easy genetic manipulation. In this review, we cover the current strategies used for the construction of yeast strains to be used in optogenetically controlled protein or metabolite production, as well as the operational aspects to be considered for the scale-up of this type of process. Finally, we discuss the main applications of optogenetic switches in yeast systems and highlight the main advantages and challenges of bioprocess development considering future directions for this field.
Collapse
Affiliation(s)
- Ana Laura A Pérez
- Instituto de Ciências Biológicas, Departamento de Biologia Celular, Bloco K, 1° andar, Universidade de Brasília, Brasília 70910-900, Brazil
| | - Luiza C Piva
- Instituto de Ciências Biológicas, Departamento de Biologia Celular, Bloco K, 1° andar, Universidade de Brasília, Brasília 70910-900, Brazil
| | - Julia P C Fulber
- Instituto de Ciências Biológicas, Departamento de Biologia Celular, Bloco K, 1° andar, Universidade de Brasília, Brasília 70910-900, Brazil
| | - Lidia M P de Moraes
- Instituto de Ciências Biológicas, Departamento de Biologia Celular, Bloco K, 1° andar, Universidade de Brasília, Brasília 70910-900, Brazil
| | - Janice L De Marco
- Instituto de Ciências Biológicas, Departamento de Biologia Celular, Bloco K, 1° andar, Universidade de Brasília, Brasília 70910-900, Brazil
| | - Hugo L A Vieira
- Instituto de Ciências Biológicas, Departamento de Biologia Celular, Bloco K, 1° andar, Universidade de Brasília, Brasília 70910-900, Brazil
| | - Cintia M Coelho
- Instituto de Ciências Biológicas, Departamento de Biologia Celular, Bloco K, 1° andar, Universidade de Brasília, Brasília 70910-900, Brazil
| | - Viviane C B Reis
- Instituto de Ciências Biológicas, Departamento de Biologia Celular, Bloco K, 1° andar, Universidade de Brasília, Brasília 70910-900, Brazil
| | - Fernando A G Torres
- Instituto de Ciências Biológicas, Departamento de Biologia Celular, Bloco K, 1° andar, Universidade de Brasília, Brasília 70910-900, Brazil.
| |
Collapse
|
16
|
Palermo GCDL, Coutouné N, Bueno JGR, Maciel LF, dos Santos LV. Exploring metal ion metabolisms to improve xylose fermentation in Saccharomyces cerevisiae. Microb Biotechnol 2021; 14:2101-2115. [PMID: 34313008 PMCID: PMC8449651 DOI: 10.1111/1751-7915.13887] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/25/2021] [Indexed: 01/22/2023] Open
Abstract
The development of high-performance xylose-fermenting yeast is essential to achieve feasible conversion of biomass-derived sugars in lignocellulose-based biorefineries. However, engineered C5-strains of Saccharomyces cerevisiae still present low xylose consumption rates under anaerobic conditions. Here, we explore alternative metabolisms involved in metal homeostasis, which positively affect C5 fermentation and analyse the non-obvious regulatory network connection of both metabolisms using time-course transcriptome analysis. Our results indicated the vacuolar Fe2+ /Mn2+ transporter CCC1, and the protein involved in heavy metal ion homeostasis BSD2, as promising new targets for rational metabolic engineering strategies, enhancing xylose consumption in nine and 2.3-fold compared with control. Notably, intracellular metal concentration levels were affected differently by mutations and the results were compared with positive controls isu1Δ, a Fe-S cluster scaffold protein, and ssk2Δ, a component of HOG pathway. Temporal expression profiles indicate a metabolic remodelling in response to xylose, demonstrating changes in the main sugar sensing signalling pathways.
Collapse
Affiliation(s)
- Gisele Cristina de Lima Palermo
- Brazilian Biorenewable National Laboratory (LNBR)Brazilian Center for Research in Energy and Materials (CNPEM)CampinasSão Paulo13083‐100Brazil
- Genetics and Molecular Biology Graduate ProgramInstitute of BiologyUniversity of Campinas (UNICAMP)CampinasSão PauloBrazil
| | - Natalia Coutouné
- Brazilian Biorenewable National Laboratory (LNBR)Brazilian Center for Research in Energy and Materials (CNPEM)CampinasSão Paulo13083‐100Brazil
- Genetics and Molecular Biology Graduate ProgramInstitute of BiologyUniversity of Campinas (UNICAMP)CampinasSão PauloBrazil
| | - João Gabriel Ribeiro Bueno
- Brazilian Biorenewable National Laboratory (LNBR)Brazilian Center for Research in Energy and Materials (CNPEM)CampinasSão Paulo13083‐100Brazil
- Genetics and Molecular Biology Graduate ProgramInstitute of BiologyUniversity of Campinas (UNICAMP)CampinasSão PauloBrazil
| | - Lucas Ferreira Maciel
- Brazilian Biorenewable National Laboratory (LNBR)Brazilian Center for Research in Energy and Materials (CNPEM)CampinasSão Paulo13083‐100Brazil
| | - Leandro Vieira dos Santos
- Brazilian Biorenewable National Laboratory (LNBR)Brazilian Center for Research in Energy and Materials (CNPEM)CampinasSão Paulo13083‐100Brazil
- Genetics and Molecular Biology Graduate ProgramInstitute of BiologyUniversity of Campinas (UNICAMP)CampinasSão PauloBrazil
| |
Collapse
|
17
|
Ishchuk OP, Frost AT, Muñiz-Paredes F, Matsumoto S, Laforge N, Eriksson NL, Martínez JL, Petranovic D. Improved production of human hemoglobin in yeast by engineering hemoglobin degradation. Metab Eng 2021; 66:259-267. [PMID: 33984513 DOI: 10.1016/j.ymben.2021.05.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/09/2021] [Accepted: 05/04/2021] [Indexed: 12/23/2022]
Abstract
With the increasing demand for blood transfusions, the production of human hemoglobin (Hb) from sustainable sources is increasingly studied. Microbial production is an attractive option, as it may provide a cheap, safe, and reliable source of this protein. To increase the production of human hemoglobin by the yeast Saccharomyces cerevisiae, the degradation of Hb was reduced through several approaches. The deletion of the genes HMX1 (encoding heme oxygenase), VPS10 (encoding receptor for vacuolar proteases), PEP4 (encoding vacuolar proteinase A), ROX1 (encoding heme-dependent repressor of hypoxic genes) and the overexpression of the HEM3 (encoding porphobilinogen deaminase) and the AHSP (encoding human alpha-hemoglobin-stabilizing protein) genes - these changes reduced heme and Hb degradation and improved heme and Hb production. The reduced hemoglobin degradation was validated by a bilirubin biosensor. During glucose fermentation, the engineered strains produced 18% of intracellular Hb relative to the total yeast protein, which is the highest production of human hemoglobin reported in yeast. This increased hemoglobin production was accompanied with an increased oxygen consumption rate and an increased glycerol yield, which (we speculate) is the yeast's response to rebalance its NADH levels under conditions of oxygen limitation and increased protein-production.
Collapse
Affiliation(s)
- Olena P Ishchuk
- Department of Biology and Biological Engineering, Systems and Synthetic Biology, Chalmers University of Technology, SE41296, Gothenburg, Sweden.
| | - August T Frost
- Department of Biotechnology and Biomedicine, Section for Synthetic Biology, Technical University of Denmark, DK-2800, Kgs. Lyngby, Denmark
| | - Facundo Muñiz-Paredes
- Department of Biology and Biological Engineering, Systems and Synthetic Biology, Chalmers University of Technology, SE41296, Gothenburg, Sweden
| | - Saki Matsumoto
- Department of Biology and Biological Engineering, Systems and Synthetic Biology, Chalmers University of Technology, SE41296, Gothenburg, Sweden
| | - Nathalie Laforge
- Department of Biology and Biological Engineering, Systems and Synthetic Biology, Chalmers University of Technology, SE41296, Gothenburg, Sweden
| | - Nélida Leiva Eriksson
- Department of Chemistry, Division of Biotechnology, Lund University, 221 00, Lund, Sweden
| | - José L Martínez
- Department of Biology and Biological Engineering, Systems and Synthetic Biology, Chalmers University of Technology, SE41296, Gothenburg, Sweden; Department of Biotechnology and Biomedicine, Section for Synthetic Biology, Technical University of Denmark, DK-2800, Kgs. Lyngby, Denmark
| | - Dina Petranovic
- Department of Biology and Biological Engineering, Systems and Synthetic Biology, Chalmers University of Technology, SE41296, Gothenburg, Sweden; Novo Nordisk Foundation Centre for Biosustainability, Chalmers University of Technology, SE41296, Gothenburg, Sweden.
| |
Collapse
|
18
|
Wang T, Wang Y, Chen C, Ren A, Yu H, Zhao M. Effect of the heme oxygenase gene on mycelial growth and polysaccharide synthesis in Ganoderma lucidum. J Basic Microbiol 2021; 61:253-264. [PMID: 33543807 DOI: 10.1002/jobm.202000622] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/23/2020] [Accepted: 01/24/2021] [Indexed: 11/06/2022]
Abstract
The heme oxygenase gene has antioxidant and cytoprotective effects in organisms, but no related research has been conducted in Ganoderma lucidum. For the first time, we cloned the HMX1 gene in G. lucidum. The CDS is 1092 bp in length and encodes 363 amino acids. The HMX1 protein was prokaryotically expressed and purified, and the enzyme activity of the purified protein was measured. The value of Km was 0.699 μM, and Vm was 81.9 nmol BV h-1 nmol-1 protein. By constructing the silencing vector pAN7-dual-HMX1i, the transformants HMX1i1 and HMX1i2 were obtained. Compared with the wild-type (WT), the average growth rate of HMX1i1 and HMX1i2 decreased by 31% and 23%, respectively, and the mycelium biomass decreased by 53% and 48%, respectively. Compared with the WT, the extracellular polysaccharide content of HMX1i1 and HMX1i2 increased by 59% and 51%, and the intracellular polysaccharide content increased by 24% and 22%, respectively. These results indicate that the HMX1 gene affects mycelial growth and polysaccharide synthesis in G. lucidum.
Collapse
Affiliation(s)
- Ting Wang
- Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Key Laboratory of Agricultural Environmental MicrobiologM, yinistry of Agriculture, Nanjing, Jiangsu, China
| | - Yihong Wang
- Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Key Laboratory of Agricultural Environmental MicrobiologM, yinistry of Agriculture, Nanjing, Jiangsu, China
| | - Chen Chen
- Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Key Laboratory of Agricultural Environmental MicrobiologM, yinistry of Agriculture, Nanjing, Jiangsu, China
| | - Ang Ren
- Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Key Laboratory of Agricultural Environmental MicrobiologM, yinistry of Agriculture, Nanjing, Jiangsu, China
| | - Hanshou Yu
- Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Key Laboratory of Agricultural Environmental MicrobiologM, yinistry of Agriculture, Nanjing, Jiangsu, China
| | - Mingwen Zhao
- Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Key Laboratory of Agricultural Environmental MicrobiologM, yinistry of Agriculture, Nanjing, Jiangsu, China
| |
Collapse
|
19
|
Hackett SR, Baltz EA, Coram M, Wranik BJ, Kim G, Baker A, Fan M, Hendrickson DG, Berndl M, McIsaac RS. Learning causal networks using inducible transcription factors and transcriptome-wide time series. Mol Syst Biol 2021; 16:e9174. [PMID: 32181581 PMCID: PMC7076914 DOI: 10.15252/msb.20199174] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 11/27/2022] Open
Abstract
We present IDEA (the Induction Dynamics gene Expression Atlas), a dataset constructed by independently inducing hundreds of transcription factors (TFs) and measuring timecourses of the resulting gene expression responses in budding yeast. Each experiment captures a regulatory cascade connecting a single induced regulator to the genes it causally regulates. We discuss the regulatory cascade of a single TF, Aft1, in detail; however, IDEA contains > 200 TF induction experiments with 20 million individual observations and 100,000 signal‐containing dynamic responses. As an application of IDEA, we integrate all timecourses into a whole‐cell transcriptional model, which is used to predict and validate multiple new and underappreciated transcriptional regulators. We also find that the magnitudes of coefficients in this model are predictive of genetic interaction profile similarities. In addition to being a resource for exploring regulatory connectivity between TFs and their target genes, our modeling approach shows that combining rapid perturbations of individual genes with genome‐scale time‐series measurements is an effective strategy for elucidating gene regulatory networks.
Collapse
Affiliation(s)
| | | | | | | | - Griffin Kim
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - Adam Baker
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | | | | | | | | |
Collapse
|
20
|
Weissman Z, Pinsky M, Donegan RK, Reddi AR, Kornitzer D. Using genetically encoded heme sensors to probe the mechanisms of heme uptake and homeostasis in Candida albicans. Cell Microbiol 2020; 23:e13282. [PMID: 33104284 DOI: 10.1111/cmi.13282] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/22/2020] [Accepted: 10/22/2020] [Indexed: 11/30/2022]
Abstract
Candida albicans is a major fungal pathogen that can utilise hemin and haemoglobin as iron sources in the iron-scarce host environment. While C. albicans is a heme prototroph, we show here that it can also efficiently utilise external heme as a cellular heme source. Using genetically encoded ratiometric fluorescent heme sensors, we show that heme extracted from haemoglobin and free hemin enter the cells with different kinetics. Heme supplied as haemoglobin is taken up via the Common in Fungal Extracellular Membrane (CFEM) hemophore cascade, and reaches the cytoplasm over several hours, whereas entry of free hemin via CFEM-dependent and independent pathways is much faster, less than an hour. To prevent an influx of extracellular heme from reaching toxic levels in the cytoplasm, the cells deploy Hmx1, a heme oxygenase. Hmx1 was previously suggested to be involved in utilisation of haemoglobin and hemin as iron sources, but we find that it is primarily required to prevent heme toxicity. Taken together, the combination of novel heme sensors with genetic analysis revealed new details of the fungal mechanisms of heme import and homeostasis, necessary to balance the uses of heme as essential cofactor and potential iron source against its toxicity.
Collapse
Affiliation(s)
- Ziva Weissman
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion-I.I.T., Haifa, Israel
| | - Mariel Pinsky
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion-I.I.T., Haifa, Israel
| | - Rebecca K Donegan
- School of Chemistry and Biochemistry and Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Amit R Reddi
- School of Chemistry and Biochemistry and Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Daniel Kornitzer
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion-I.I.T., Haifa, Israel
| |
Collapse
|
21
|
Hopper CP, De La Cruz LK, Lyles KV, Wareham LK, Gilbert JA, Eichenbaum Z, Magierowski M, Poole RK, Wollborn J, Wang B. Role of Carbon Monoxide in Host-Gut Microbiome Communication. Chem Rev 2020; 120:13273-13311. [PMID: 33089988 DOI: 10.1021/acs.chemrev.0c00586] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Nature is full of examples of symbiotic relationships. The critical symbiotic relation between host and mutualistic bacteria is attracting increasing attention to the degree that the gut microbiome is proposed by some as a new organ system. The microbiome exerts its systemic effect through a diverse range of metabolites, which include gaseous molecules such as H2, CO2, NH3, CH4, NO, H2S, and CO. In turn, the human host can influence the microbiome through these gaseous molecules as well in a reciprocal manner. Among these gaseous molecules, NO, H2S, and CO occupy a special place because of their widely known physiological functions in the host and their overlap and similarity in both targets and functions. The roles that NO and H2S play have been extensively examined by others. Herein, the roles of CO in host-gut microbiome communication are examined through a discussion of (1) host production and function of CO, (2) available CO donors as research tools, (3) CO production from diet and bacterial sources, (4) effect of CO on bacteria including CO sensing, and (5) gut microbiome production of CO. There is a large amount of literature suggesting the "messenger" role of CO in host-gut microbiome communication. However, much more work is needed to begin achieving a systematic understanding of this issue.
Collapse
Affiliation(s)
- Christopher P Hopper
- Institute for Experimental Biomedicine, University Hospital Wuerzburg, Wuerzburg, Bavaria DE 97080, Germany.,Department of Medicinal Chemistry, College of Pharmacy, The University of Florida, Gainesville, Florida 32611, United States
| | - Ladie Kimberly De La Cruz
- Department of Chemistry & Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| | - Kristin V Lyles
- Department of Biology, Georgia State University, Atlanta, Georgia 30303, United States
| | - Lauren K Wareham
- The Vanderbilt Eye Institute and Department of Ophthalmology & Visual Sciences, The Vanderbilt University Medical Center and School of Medicine, Nashville, Tennessee 37232, United States
| | - Jack A Gilbert
- Department of Pediatrics, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
| | - Zehava Eichenbaum
- Department of Biology, Georgia State University, Atlanta, Georgia 30303, United States
| | - Marcin Magierowski
- Cellular Engineering and Isotope Diagnostics Laboratory, Department of Physiology, Jagiellonian University Medical College, Cracow PL 31-531, Poland
| | - Robert K Poole
- Department of Molecular Biology and Biotechnology, The University of Sheffield, Firth Court, Sheffield S10 2TN, U.K
| | - Jakob Wollborn
- Department of Anesthesiology and Critical Care, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg DE 79085, Germany.,Department of Anesthesiology, Perioperative and Pain Management, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Binghe Wang
- Department of Chemistry & Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| |
Collapse
|
22
|
Kornitzer D, Roy U. Pathways of heme utilization in fungi. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118817. [PMID: 32777371 DOI: 10.1016/j.bbamcr.2020.118817] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/02/2020] [Accepted: 08/03/2020] [Indexed: 02/08/2023]
Abstract
Iron acquisition is challenging in most environments. As an alternative to elemental iron, organisms can take up iron-protoporphyrin IX, or heme. Heme can be found in decaying organic matter and is particularly prevalent in animal hosts. Fungi have evolved at least three distinct endocytosis-mediated heme uptake systems, which have been studied in detail in the organisms Candida albicans, Cryptococcus neoformans and Schizosaccharomyces pombe. Here we summarize the known molecular details of these three uptake systems that enable parasitic and saprophytic fungi to take advantage of external heme as either cellular iron or heme sources.
Collapse
Affiliation(s)
- Daniel Kornitzer
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| | - Udita Roy
- Department of Molecular Microbiology, B. Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| |
Collapse
|
23
|
Laguna-Teno F, Suarez-Diez M, Tamayo-Ramos JA. Commonalities and Differences in the Transcriptional Response of the Model Fungus Saccharomyces cerevisiae to Different Commercial Graphene Oxide Materials. Front Microbiol 2020; 11:1943. [PMID: 32849484 PMCID: PMC7431627 DOI: 10.3389/fmicb.2020.01943] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/23/2020] [Indexed: 12/31/2022] Open
Abstract
Graphene oxide has become a very appealing nanomaterial during the last years for many different applications, but its possible impact in different biological systems remains unclear. Here, an assessment to understand the toxicity of different commercial graphene oxide nanomaterials on the unicellular fungal model organism Saccharomyces cerevisiae was performed. For this task, an RNA purification protocol was optimized to avoid the high nucleic acid absorption capacity of graphene oxide. The developed protocol is based on a sorbitol gradient separation process for the isolation of adequate ribonucleic acid levels (in concentration and purity) from yeast cultures exposed to the carbon derived nanomaterial. To pinpoint potential toxicity mechanisms and pathways, the transcriptome of S. cerevisiae exposed to 160 mg L-1 of monolayer graphene oxide (GO) and graphene oxide nanocolloids (GOC) was studied and compared. Both graphene oxide products induced expression changes in a common group of genes (104), many of them related to iron homeostasis, starvation and stress response, amino acid metabolism and formate catabolism. Also, a high number of genes were only differentially expressed in either GO (236) or GOC (1077) exposures, indicating that different commercial products can induce specific changes in the physiological state of the fungus.
Collapse
Affiliation(s)
- Felix Laguna-Teno
- International Research Centre in Critical Raw Materials-ICCRAM, University of Burgos, Burgos, Spain
| | - Maria Suarez-Diez
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Wageningen, Netherlands
| | | |
Collapse
|
24
|
Martínez-Pastor MT, Puig S. Adaptation to iron deficiency in human pathogenic fungi. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118797. [PMID: 32663505 DOI: 10.1016/j.bbamcr.2020.118797] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/13/2020] [Accepted: 07/05/2020] [Indexed: 02/08/2023]
Abstract
Iron is an essential micronutrient for virtually all eukaryotic organisms and plays a central role during microbial infections. Invasive fungal diseases are associated with strikingly high rates of mortality, but their impact on human health is usually underestimated. Upon a fungal infection, hosts restrict iron availability in order to limit the growth and virulence of the pathogen. Here, we use two model yeasts, Saccharomyces cerevisiae and Schizosaccharomyces pombe, to delve into the response to iron deficiency of human fungal pathogens, such as Candida glabrata, Candida albicans, Aspergillus fumigatus and Cryptococcus neoformans. Fungi possess common and species-specific mechanisms to acquire iron and to control the response to iron limitation. Upon iron scarcity, fungi activate a wide range of elegant strategies to capture and import exogenous iron, mobilize iron from intracellular stores, and modulate their metabolism to economize and prioritize iron utilization. Hence, iron homeostasis genes represent remarkable virulence factors that can be used as targets for the development of novel antifungal treatments.
Collapse
Affiliation(s)
| | - Sergi Puig
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (IATA), Consejo Superior de Investigaciones Científicas (CSIC), Paterna, Valencia, Spain.
| |
Collapse
|
25
|
Heme-iron acquisition in fungi. Curr Opin Microbiol 2019; 52:77-83. [DOI: 10.1016/j.mib.2019.05.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 01/09/2023]
|
26
|
Bottino-Rojas V, Pereira LOR, Silva G, Talyuli OAC, Dunkov BC, Oliveira PL, Paiva-Silva GO. Non-canonical transcriptional regulation of heme oxygenase in Aedes aegypti. Sci Rep 2019; 9:13726. [PMID: 31551499 PMCID: PMC6760526 DOI: 10.1038/s41598-019-49396-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/22/2019] [Indexed: 12/19/2022] Open
Abstract
Heme oxygenase (HO) is a ubiquitous enzyme responsible for heme breakdown, which yields carbon monoxide (CO), biliverdin (BV) and ferrous ion. Here we show that the Aedes aegypti heme oxygenase gene (AeHO - AAEL008136) is expressed in different developmental stages and tissues. AeHO expression increases after a blood meal in the midgut, and its maximal transcription levels overlaps with the maximal rate of the further modified A. aegypti biglutaminyl-biliverdin (AeBV) pigment production. HO is a classical component of stress response in eukaryotic cells, being activated under oxidative stress or increased heme levels. Indeed, the final product of HO activity in the mosquito midgut, AeBV, exerts a protective antioxidant activity. AeHO, however, does not seem to be under a classical redox-sensitive transcriptional regulation, being unresponsive to heme itself, and even down regulated when insects face a pro-oxidant insult. In contrast, AeHO gene expression responds to nutrient sensing mechanisms, through the target of rapamycin (TOR) pathway. This unusual transcriptional control of AeHO, together with the antioxidant properties of AeBV, suggests that heme degradation by HO, in addition to its important role in protection of Aedes aegypti against heme exposure, also acts as a digestive feature, being an essential adaptation to blood feeding.
Collapse
Affiliation(s)
- Vanessa Bottino-Rojas
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Luiza O R Pereira
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, 21040-360, Brazil
| | - Gabriela Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Octavio A C Talyuli
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Boris C Dunkov
- Center for Insect Science, The University of Arizona, Tucson, AZ, 85721-0106, USA
| | - Pedro L Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Gabriela O Paiva-Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
| |
Collapse
|
27
|
Lindahl PA. A comprehensive mechanistic model of iron metabolism in Saccharomyces cerevisiae. Metallomics 2019; 11:1779-1799. [PMID: 31531508 DOI: 10.1039/c9mt00199a] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ironome of budding yeast (circa 2019) consists of approximately 139 proteins and 5 nonproteinaceous species. These proteins were grouped according to location in the cell, type of iron center(s), and cellular function. The resulting 27 groups were used, along with an additional 13 nonprotein components, to develop a mesoscale mechanistic model that describes the import, trafficking, metallation, and regulation of iron within growing yeast cells. The model was designed to be simultaneously mutually autocatalytic and mutually autoinhibitory - a property called autocatinhibitory that should be most realistic for simulating cellular biochemical processes. The model was assessed at the systems' level. General conclusions are presented, including a new perspective on understanding regulatory mechanisms in cellular systems. Some unsettled issues are described. This model, once fully developed, has the potential to mimic the phenotype (at a coarse-grain level) of all iron-related genetic mutations in this simple and well-studied eukaryote.
Collapse
Affiliation(s)
- Paul A Lindahl
- Departments of Chemistry and of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843-3255, USA.
| |
Collapse
|
28
|
Devaux F, Thiébaut A. The regulation of iron homeostasis in the fungal human pathogen Candida glabrata. MICROBIOLOGY-SGM 2019; 165:1041-1060. [PMID: 31050635 DOI: 10.1099/mic.0.000807] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Iron is an essential element to most microorganisms, yet an excess of iron is toxic. Hence, living cells have to maintain a tight balance between iron uptake and iron consumption and storage. The control of intracellular iron concentrations is particularly challenging for pathogens because mammalian organisms have evolved sophisticated high-affinity systems to sequester iron from microbes and because iron availability fluctuates among the different host niches. In this review, we present the current understanding of iron homeostasis and its regulation in the fungal pathogen Candida glabrata. This yeast is an emerging pathogen which has become the second leading cause of candidemia, a life-threatening invasive mycosis. C. glabrata is relatively poorly studied compared to the closely related model yeast Saccharomyces cerevisiae or to the pathogenic yeast Candida albicans. Still, several research groups have started to identify the actors of C. glabrata iron homeostasis and its transcriptional and post-transcriptional regulation. These studies have revealed interesting particularities of C. glabrata and have shed new light on the evolution of fungal iron homeostasis.
Collapse
Affiliation(s)
- Frédéric Devaux
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Laboratory of Computational and Quantitative Biology, F-75005, Paris, France
| | - Antonin Thiébaut
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Laboratory of Computational and Quantitative Biology, F-75005, Paris, France
| |
Collapse
|
29
|
Lu Y, Liu G, Jiang H, Chi Z, Chi Z. An insight into the iron acquisition and homeostasis in Aureobasidium melanogenum HN6.2 strain through genome mining and transcriptome analysis. Funct Integr Genomics 2018; 19:137-150. [PMID: 30251029 DOI: 10.1007/s10142-018-0633-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 08/07/2018] [Accepted: 08/23/2018] [Indexed: 11/26/2022]
Abstract
Aureobasidium melanogenum HN6.2 is a unique yeast strain who can produce the siderophore of fusigen under iron starvation to guarantee its survival. However, a comprehensive understanding of mechanisms involved in iron acquisition and homeostasis for it is still vacant. In this study, genome sequencing and mining revealed that A. melanogenum HN6.2 strain was the first yeast species that exclusively possessed all the four known mechanisms for the iron acquisition: (i) the siderophore-mediated iron uptake; (ii) reductive iron assimilation; (iii) low-affinity ferrous uptake; and (iv) heme utilization, which suggested its stronger adaptability than Aspergillus fumigatus and Saccharomyces cerevisiae. This HN6.2 strain also employed the vacuolar iron storage for immobilizing the excessive iron to avoid its cellular toxicity. Specially, genome mining indicated that A. melanogenum HN6.2 strain could also synthesize ferricrocin siderophore. Further HPLC and Q-Tof-MS analysis confirmed that the siderophores synthesized by this strain consisted of cyclic fusigen, linear fusigen, ferricrocin, and hydroxyferricrocin and they played parallel roles as both intracellular and extracellular siderophores. Also, the heme utilization for this strain was experimentally verified by the knock-out of heme oxygenase gene. For iron homeostasis, the transcriptome analysis revealed that this strain mainly employed two central regulators of SreA/HapX to tune iron uptake and storage at the transcriptional level. It was also noted that mitogen-activated protein kinase C gene (MpkC) exhibited a transcriptional up-regulation under iron sufficiency, suggesting that it may serve as another factor involved in the repression of siderophore biosynthesis. This is the first genetic blueprint of iron acquisition and homeostasis for A. melanogenum.
Collapse
Affiliation(s)
- Yi Lu
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, China
| | - Guanglei Liu
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, China
| | - Hong Jiang
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, China
| | - Zhenming Chi
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, China
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, Qingdao, 266003, China
| | - Zhe Chi
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
30
|
Martins TS, Costa V, Pereira C. Signaling pathways governing iron homeostasis in budding yeast. Mol Microbiol 2018; 109:422-432. [DOI: 10.1111/mmi.14009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Telma S. Martins
- I3S-Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Porto Portugal
- IBMC-Instituto de Biologia Molecular e Celular; Universidade do Porto; Porto Portugal
| | - Vítor Costa
- I3S-Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Porto Portugal
- IBMC-Instituto de Biologia Molecular e Celular; Universidade do Porto; Porto Portugal
- Departamento de Biologia Molecular; Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto; Porto Portugal
| | - Clara Pereira
- I3S-Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Porto Portugal
- IBMC-Instituto de Biologia Molecular e Celular; Universidade do Porto; Porto Portugal
- Departamento de Biologia Molecular; Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto; Porto Portugal
| |
Collapse
|
31
|
Hanna DA, Hu R, Kim H, Martinez-Guzman O, Torres MP, Reddi AR. Heme bioavailability and signaling in response to stress in yeast cells. J Biol Chem 2018; 293:12378-12393. [PMID: 29921585 DOI: 10.1074/jbc.ra118.002125] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 06/15/2018] [Indexed: 12/28/2022] Open
Abstract
Protoheme (hereafter referred to as heme) is an essential cellular cofactor and signaling molecule that is also potentially cytotoxic. To mitigate heme toxicity, heme synthesis and degradation are tightly coupled to heme utilization in order to limit the intracellular concentration of "free" heme. Such a model, however, would suggest that a readily accessible steady-state, bioavailable labile heme (LH) pool is not required for supporting heme-dependent processes. Using the yeast Saccharomyces cerevisiae as a model and fluorescent heme sensors, site-specific heme chelators, and molecular genetic approaches, we found here that 1) yeast cells preferentially use LH in heme-depleted conditions; 2) sequestration of cytosolic LH suppresses heme signaling; and 3) lead (Pb2+) stress contributes to a decrease in total heme, but an increase in LH, which correlates with increased heme signaling. We also observed that the proteasome is involved in the regulation of the LH pool and that loss of proteasomal activity sensitizes cells to Pb2+ effects on heme homeostasis. Overall, these findings suggest an important role for LH in supporting heme-dependent functions in yeast physiology.
Collapse
Affiliation(s)
| | - Rebecca Hu
- From the School of Chemistry and Biochemistry
| | - Hyojung Kim
- From the School of Chemistry and Biochemistry.,School of Biological Sciences, and
| | | | - Matthew P Torres
- School of Biological Sciences, and.,Parker Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Amit R Reddi
- From the School of Chemistry and Biochemistry, .,Parker Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332
| |
Collapse
|
32
|
Gerwien F, Safyan A, Wisgott S, Brunke S, Kasper L, Hube B. The Fungal Pathogen Candida glabrata Does Not Depend on Surface Ferric Reductases for Iron Acquisition. Front Microbiol 2017. [PMID: 28642757 PMCID: PMC5463049 DOI: 10.3389/fmicb.2017.01055] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Iron acquisition is a crucial virulence determinant for many bacteria and fungi, including the opportunistic fungal pathogens Candida albicans and C. glabrata. While the diverse strategies used by C. albicans for obtaining iron from the host are well-described, much less is known about the acquisition of this micronutrient from host sources by C. glabrata – a distant relative of C. albicans with closer evolutionary ties to Saccharomyces cerevisiae, which nonetheless causes severe clinical symptoms in humans. Here we show that C. glabrata is much more restricted than C. albicans in using host iron sources, lacking, for example, the ability to grow on transferrin and hemin/hemoglobin. Instead, C. glabrata is able to use ferritin and non-protein-bound iron (FeCl3) as iron sources in a pH-dependent manner. As in other fungal pathogens, iron-dependent growth requires the reductive high affinity (HA) iron uptake system. Typically highly conserved, this uptake mechanism normally relies on initial ferric reduction by cell-surface ferric reductases. The C. glabrata genome contains only three such putative ferric reductases, which were found to be dispensable for iron-dependent growth. In addition and in contrast to C. albicans and S. cerevisiae, we also detected no surface ferric reductase activity in C. glabrata. Instead, extracellular ferric reduction was found in this and the two other fungal species, which was largely dependent on an excreted low-molecular weight, non-protein ferric reductant. We therefore propose an iron acquisition strategy of C. glabrata which differs from other pathogenic fungi, such as C. albicans, in that it depends on a limited set of host iron sources and that it lacks the need for surface ferric reductases. Extracellular ferric reduction by a secreted molecule possibly compensates for the loss of surface ferric reductase activity in the HA iron uptake system.
Collapse
Affiliation(s)
- Franziska Gerwien
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell InstituteJena, Germany
| | - Abu Safyan
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell InstituteJena, Germany
| | - Stephanie Wisgott
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell InstituteJena, Germany
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell InstituteJena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell InstituteJena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell InstituteJena, Germany.,Department of Microbial Pathogenicity Mechanisms, Friedrich Schiller UniversityJena, Germany.,Center for Sepsis Control and Care, University HospitalJena, Germany
| |
Collapse
|
33
|
Abstract
Heme oxygenases are composed of two isozymes, Hmox1 and Hmox2, that catalyze the degradation of heme to carbon monoxide (CO), ferrous iron, and biliverdin, the latter of which is subsequently converted to bilirubin. While initially considered to be waste products, CO and biliverdin/bilirubin have been shown over the last 20 years to modulate key cellular processes, such as inflammation, cell proliferation, and apoptosis, as well as antioxidant defense. This shift in paradigm has led to the importance of heme oxygenases and their products in cell physiology now being well accepted. The identification of the two human cases thus far of heme oxygenase deficiency and the generation of mice deficient in Hmox1 or Hmox2 have reiterated a role for these enzymes in both normal cell function and disease pathogenesis, especially in the context of cardiovascular disease. This review covers the current knowledge on the function of both Hmox1 and Hmox2 at both a cellular and tissue level in the cardiovascular system. Initially, the roles of heme oxygenases in vascular health and the regulation of processes central to vascular diseases are outlined, followed by an evaluation of the role(s) of Hmox1 and Hmox2 in various diseases such as atherosclerosis, intimal hyperplasia, myocardial infarction, and angiogenesis. Finally, the therapeutic potential of heme oxygenases and their products are examined in a cardiovascular disease context, with a focus on how the knowledge we have gained on these enzymes may be capitalized in future clinical studies.
Collapse
Affiliation(s)
- Anita Ayer
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Abolfazl Zarjou
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Anupam Agarwal
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| |
Collapse
|
34
|
HAL2 overexpression induces iron acquisition in bdf1Δ cells and enhances their salt resistance. Curr Genet 2016; 63:229-239. [DOI: 10.1007/s00294-016-0628-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/26/2016] [Accepted: 06/27/2016] [Indexed: 11/26/2022]
|
35
|
Ghio AJ, Schreinemachers DM. Heme Oxygenase Activity Correlates with Serum Indices of Iron Homeostasis in Healthy Nonsmokers. Biomark Insights 2016; 11:49-54. [PMID: 27199547 PMCID: PMC4863832 DOI: 10.4137/bmi.s36226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 02/10/2016] [Accepted: 02/12/2016] [Indexed: 11/05/2022] Open
Abstract
Heme oxygenase (HO) catalyzes the breakdown of heme to carbon monoxide, iron, and biliverdin. While the use of genetically altered animal models in investigation has established distinct associations between HO activity and systemic iron availability, studies have not yet confirmed such participation of HO in iron homeostasis of humans. Carbon monoxide produced through HO activity will bind to hemoglobin in circulating erythrocytes, and therefore, blood carboxyhemoglobin (COHb) can be used as an index of HO activity. Using the second National Health and Nutrition Examination Survey, we tested the postulate that HO activity correlates with serum indices of iron homeostasis in healthy nonsmokers. The investigation included 844 lifetime nonsmokers (586 females) 18 years of age and older in the study population. Significant correlations were demonstrated between COHb and several indices of iron homeostasis including serum levels of both ferritin and iron and percentage iron saturation of transferrin. There was no significant association between COHb and hemoglobin, the largest repository of heme in the human body, which functions as the substrate for HO. We conclude that HO activity contributes to human iron homeostasis with significant correlations between COHb and serum ferritin and iron levels and percentage iron saturation of transferrin.
Collapse
Affiliation(s)
- Andrew J. Ghio
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Chapel Hill, NC, USA
| | - Dina M. Schreinemachers
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Chapel Hill, NC, USA
| |
Collapse
|
36
|
Bayeva M, Chang HC, Wu R, Ardehali H. When less is more: novel mechanisms of iron conservation. Trends Endocrinol Metab 2013; 24:569-577. [PMID: 23948590 PMCID: PMC4720524 DOI: 10.1016/j.tem.2013.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 07/15/2013] [Accepted: 07/16/2013] [Indexed: 01/24/2023]
Abstract
Disorders of iron homeostasis are very common, yet the molecular mechanisms of iron regulation remain understudied. Over 20 years have passed since the first characterization of iron-regulatory proteins (IRP) as mediators of cellular iron-deficiency response in mammals through iron acquisition. However, little is known about other mechanisms necessary for adaptation to low-iron states. In this review, we present recent evidence that establishes the existence of a new iron-regulatory pathway aimed at iron conservation and optimization of iron use through suppression of nonessential iron-consuming processes. Moreover, we discuss the possible links between iron homeostasis and energy metabolism uncovered by studies of iron-deficiency response.
Collapse
Affiliation(s)
- Marina Bayeva
- Feinberg Cardiovascular Research Institute, Northwestern University School of Medicine, Tarry 14-733, 303 E Chicago Ave, Chicago, IL
| | - Hsiang-Chun Chang
- Feinberg Cardiovascular Research Institute, Northwestern University School of Medicine, Tarry 14-733, 303 E Chicago Ave, Chicago, IL
| | - Rongxue Wu
- Feinberg Cardiovascular Research Institute, Northwestern University School of Medicine, Tarry 14-733, 303 E Chicago Ave, Chicago, IL
| | - Hossein Ardehali
- Feinberg Cardiovascular Research Institute, Northwestern University School of Medicine, Tarry 14-733, 303 E Chicago Ave, Chicago, IL
| |
Collapse
|
37
|
Abstract
All living organisms require nutrient minerals for growth and have developed mechanisms to acquire, utilize, and store nutrient minerals effectively. In the aqueous cellular environment, these elements exist as charged ions that, together with protons and hydroxide ions, facilitate biochemical reactions and establish the electrochemical gradients across membranes that drive cellular processes such as transport and ATP synthesis. Metal ions serve as essential enzyme cofactors and perform both structural and signaling roles within cells. However, because these ions can also be toxic, cells have developed sophisticated homeostatic mechanisms to regulate their levels and avoid toxicity. Studies in Saccharomyces cerevisiae have characterized many of the gene products and processes responsible for acquiring, utilizing, storing, and regulating levels of these ions. Findings in this model organism have often allowed the corresponding machinery in humans to be identified and have provided insights into diseases that result from defects in ion homeostasis. This review summarizes our current understanding of how cation balance is achieved and modulated in baker's yeast. Control of intracellular pH is discussed, as well as uptake, storage, and efflux mechanisms for the alkali metal cations, Na(+) and K(+), the divalent cations, Ca(2+) and Mg(2+), and the trace metal ions, Fe(2+), Zn(2+), Cu(2+), and Mn(2+). Signal transduction pathways that are regulated by pH and Ca(2+) are reviewed, as well as the mechanisms that allow cells to maintain appropriate intracellular cation concentrations when challenged by extreme conditions, i.e., either limited availability or toxic levels in the environment.
Collapse
|
38
|
|
39
|
Ahmed U, Latham PS, Oates PS. Interactions between hepatic iron and lipid metabolism with possible relevance to steatohepatitis. World J Gastroenterol 2012; 18:4651-8. [PMID: 23002334 PMCID: PMC3442203 DOI: 10.3748/wjg.v18.i34.4651] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 03/27/2012] [Accepted: 03/29/2012] [Indexed: 02/06/2023] Open
Abstract
The liver is an important site for iron and lipid metabolism and the main site for the interactions between these two metabolic pathways. Although conflicting results have been obtained, most studies support the hypothesis that iron plays a role in hepatic lipogenesis. Iron is an integral part of some enzymes and transporters involved in lipid metabolism and, as such, may exert a direct effect on hepatic lipid load, intrahepatic metabolic pathways and hepatic lipid secretion. On the other hand, iron in its ferrous form may indirectly affect lipid metabolism through its ability to induce oxidative stress and inflammation, a hypothesis which is currently the focus of much research in the field of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis (NAFLD/NASH). The present review will first discuss how iron might directly interact with the metabolism of hepatic lipids and then consider a new perspective on the way in which iron may have a role in the two hit hypothesis for the progression of NAFLD via ferroportin and the iron regulatory molecule hepcidin. The review concludes that iron has important interactions with lipid metabolism in the liver that can impact on the development of NAFLD/NASH. More defined studies are required to improve our understanding of these effects.
Collapse
|
40
|
Philpott CC, Leidgens S, Frey AG. Metabolic remodeling in iron-deficient fungi. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:1509-20. [PMID: 22306284 DOI: 10.1016/j.bbamcr.2012.01.012] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 01/13/2012] [Accepted: 01/18/2012] [Indexed: 01/12/2023]
Abstract
Eukaryotic cells contain dozens, perhaps hundreds, of iron-dependent proteins, which perform critical functions in nearly every major cellular process. Nutritional iron is frequently available to cells in only limited amounts; thus, unicellular and higher eukaryotes have evolved mechanisms to cope with iron scarcity. These mechanisms have been studied at the molecular level in the model eukaryotes Saccharomyces cerevisiae and Schizosaccharomyces pombe, as well as in some pathogenic fungi. Each of these fungal species exhibits metabolic adaptations to iron deficiency that serve to reduce the cell's reliance on iron. However, the regulatory mechanisms that accomplish these adaptations differ greatly between fungal species. This article is part of a Special Issue entitled: Cell Biology of Metals.
Collapse
Affiliation(s)
- Caroline C Philpott
- Genetics and Metabolism Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 10, Rm. 9B-16, 10 Center Drive, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
41
|
Identification of the molecular mechanisms underlying the cytotoxic action of a potent platinum metallointercalator. J Chem Biol 2011; 5:51-61. [PMID: 23226166 DOI: 10.1007/s12154-011-0070-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 11/22/2011] [Indexed: 10/15/2022] Open
Abstract
UNLABELLED Platinum-based DNA metallointercalators are structurally different from the covalent DNA binders such as cisplatin and its derivatives but have potent in vitro activity in cancer cell lines. However, limited understanding of their molecular mechanisms of cytotoxic action greatly hinders their further development as anticancer agents. In this study, a lead platinum-based metallointercalator, [(5,6-dimethyl-1,10-phenanthroline) (1S,2S-diaminocyclohexane)platinum(II)](2+) (56MESS) was found to be 163-fold more active than cisplatin in a cisplatin-resistant cancer cell line. By using transcriptomics in a eukaryotic model organism, yeast Saccharomyces cerevisiae, we identified 93 genes that changed their expressions significantly upon exposure of 56MESS in comparison to untreated controls (p ≤ 0.05). Bioinformatic analysis of these genes demonstrated that iron and copper metabolism, sulfur-containing amino acids and stress response were involved in the cytotoxicity of 56MESS. Follow-up experiments showed that the iron and copper concentrations were much lower in 56MESS-treated cells compared to controls as measured by inductively coupled plasma optical emission spectrometry. Deletion mutants of the key genes in the iron and copper metabolism pathway and glutathione synthesis were sensitive to 56MESS. Taken together, the study demonstrated that the cytotoxic action of 56MESS is mediated by its ability to disrupt iron and copper metabolism, suppress the biosynthesis of sulfur-containing amino acids and attenuate cellular defence capacity. As these mechanisms are in clear contrast to the DNA binding mechanism for cisplatin and its derivative, 56MESS may be able to overcome cisplatin-resistant cancers. These findings have provided basis to further develop the platinum-based metallointercalators as anticancer agents. ELECTRONIC SUPPLEMENTARY MATERIAL The online version of this article (doi:10.1007/s12154-011-0070-x) contains supplementary material, which is available to authorized users.
Collapse
|
42
|
Murray DB, Haynes K, Tomita M. Redox regulation in respiring Saccharomyces cerevisiae. Biochim Biophys Acta Gen Subj 2011; 1810:945-58. [PMID: 21549177 DOI: 10.1016/j.bbagen.2011.04.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 03/16/2011] [Accepted: 04/17/2011] [Indexed: 11/30/2022]
Abstract
BACKGROUND In biological systems, redox reactions are central to most cellular processes and the redox potential of the intracellular compartment dictates whether a particular reaction can or cannot occur. Indeed the widespread use of redox reactions in biological systems makes their detailed description outside the scope of one review. SCOPE OF THE REVIEW Here we will focus on how system-wide redox changes can alter the reaction and transcriptional landscape of Saccharomyces cerevisiae. To understand this we explore the major determinants of cellular redox potential, how these are sensed by the cell and the dynamic responses elicited. MAJOR CONCLUSIONS Redox regulation is a large and complex system that has the potential to rapidly and globally alter both the reaction and transcription landscapes. Although we have a basic understanding of many of the sub-systems and a partial understanding of the transcriptional control, we are far from understanding how these systems integrate to produce coherent responses. We argue that this non-linear system self-organises, and that the output in many cases is temperature-compensated oscillations that may temporally partition incompatible reactions in vivo. GENERAL SIGNIFICANCE Redox biochemistry impinges on most of cellular processes and has been shown to underpin ageing and many human diseases. Integrating the complexity of redox signalling and regulation is perhaps one of the most challenging areas of biology. This article is part of a Special Issue entitled Systems Biology of Microorganisms.
Collapse
Affiliation(s)
- Douglas B Murray
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan.
| | | | | |
Collapse
|
43
|
Collinson EJ, Wimmer-Kleikamp S, Gerega SK, Yang YH, Parish CR, Dawes IW, Stocker R. The yeast homolog of heme oxygenase-1 affords cellular antioxidant protection via the transcriptional regulation of known antioxidant genes. J Biol Chem 2010; 286:2205-14. [PMID: 21081499 DOI: 10.1074/jbc.m110.187062] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heme oxygenase-1 (HO-1) degrades heme and protects cells from oxidative challenge. This antioxidant activity is thought to result from the HO-1 enzymatic activity, manifested by a decrease in the concentration of the pro-oxidant substrate heme, and an increase in the antioxidant product bilirubin. Using a global transcriptional approach, and yeast as a model, we show that HO-1 affords cellular protection via up-regulation of transcripts encoding enzymes involved in cellular antioxidant defense, rather than via its oxygenase activity. Like mammalian cells, yeast responds to oxidative stress by expressing its HO-1 homolog and, compared with the wild type, heme oxygenase-null mutant cells have increased sensitivity toward oxidants that is rescued by overexpression of human HO-1 or its yeast homolog. Increased oxidant sensitivity of heme oxygenase-null mutant cells is explained by a decrease in the expression of the genes encoding γ-glutamylcysteine synthetase, glutathione peroxidase, catalase, and methionine sulfoxide reductase, because overexpression of any of these genes affords partial, and overexpression of all four genes provides complete, protection to the null mutant. Genes encoding antioxidant enzymes represent only a small portion of the 480 differentially expressed transcripts in heme oxygenase-null mutants. Transcriptional regulation may be explained by the nuclear localization of heme oxygenase observed in oxidant-challenged cells. Our results challenge the notion that HO-1 functions simply as a catabolic and antioxidant enzyme. They indicate much broader functions for HO-1, the unraveling of which may help explain the multiple biological responses reported in animals as a result of altered HO-1 expression.
Collapse
Affiliation(s)
- Emma J Collinson
- Centre for Vascular Research, School of Medical Sciences (Pathology) and Bosch Institute, Sydney Medical School, Sydney, New South Wales 2006, Australia
| | | | | | | | | | | | | |
Collapse
|
44
|
Shakoury-Elizeh M, Protchenko O, Berger A, Cox J, Gable K, Dunn TM, Prinz WA, Bard M, Philpott CC. Metabolic response to iron deficiency in Saccharomyces cerevisiae. J Biol Chem 2010; 285:14823-33. [PMID: 20231268 DOI: 10.1074/jbc.m109.091710] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Iron is an essential cofactor for enzymes involved in numerous cellular processes, yet little is known about the impact of iron deficiency on cellular metabolism or iron proteins. Previous studies have focused on changes in transcript and proteins levels in iron-deficient cells, yet these changes may not reflect changes in transport activity or flux through a metabolic pathway. We analyzed the metabolomes and transcriptomes of yeast grown in iron-rich and iron-poor media to determine which biosynthetic processes are altered when iron availability falls. Iron deficiency led to changes in glucose metabolism, amino acid biosynthesis, and lipid biosynthesis that were due to deficiencies in specific iron-dependent enzymes. Iron-sulfur proteins exhibited loss of iron cofactors, yet amino acid synthesis was maintained. Ergosterol and sphingolipid biosynthetic pathways had blocks at points where heme and diiron enzymes function, whereas Ole1, the essential fatty acid desaturase, was resistant to iron depletion. Iron-deficient cells exhibited depletion of most iron enzyme activities, but loss of activity during iron deficiency did not consistently disrupt metabolism. Amino acid homeostasis was robust, but iron deficiency impaired lipid synthesis, altering the properties and functions of cellular membranes.
Collapse
Affiliation(s)
- Minoo Shakoury-Elizeh
- Liver Diseases Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Iron regulation through the back door: iron-dependent metabolite levels contribute to transcriptional adaptation to iron deprivation in Saccharomyces cerevisiae. EUKARYOTIC CELL 2009; 9:460-71. [PMID: 20008079 DOI: 10.1128/ec.00213-09] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Budding yeast (Saccharomyces cerevisiae) responds to iron deprivation both by Aft1-Aft2-dependent transcriptional activation of genes involved in cellular iron uptake and by Cth1-Cth2-specific degradation of certain mRNAs coding for iron-dependent biosynthetic components. Here, we provide evidence for a novel principle of iron-responsive gene expression. This regulatory mechanism is based on the modulation of transcription through the iron-dependent variation of levels of regulatory metabolites. As an example, the LEU1 gene of branched-chain amino acid biosynthesis is downregulated under iron-limiting conditions through depletion of the metabolic intermediate alpha-isopropylmalate, which functions as a key transcriptional coactivator of the Leu3 transcription factor. Synthesis of alpha-isopropylmalate involves the iron-sulfur protein Ilv3, which is inactivated under iron deficiency. As another example, decreased mRNA levels of the cytochrome c-encoding CYC1 gene under iron-limiting conditions involve heme-dependent transcriptional regulation via the Hap1 transcription factor. Synthesis of the iron-containing heme is directly correlated with iron availability. Thus, the iron-responsive expression of genes that are downregulated under iron-limiting conditions is conferred by two independent regulatory mechanisms: transcriptional regulation through iron-responsive metabolites and posttranscriptional mRNA degradation. Only the combination of the two processes provides a quantitative description of the response to iron deprivation in yeast.
Collapse
|
46
|
Carbon monoxide in biology and microbiology: surprising roles for the "Detroit perfume". Adv Microb Physiol 2009; 56:85-167. [PMID: 20943125 DOI: 10.1016/s0065-2911(09)05603-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Carbon monoxide (CO) is a colorless, odorless gas with a reputation for being an anthropogenic poison; there is extensive documentation of the modes of human exposure, toxicokinetics, and health effects. However, CO is also generated endogenously by heme oxygenases (HOs) in mammals and microbes, and its extraordinary biological activities are now recognized and increasingly utilized in medicine and physiology. This review introduces recent advances in CO biology and chemistry and illustrates the exciting possibilities that exist for a deeper understanding of its biological consequences. However, the microbiological literature is scant and is currently restricted to: 1) CO-metabolizing bacteria, CO oxidation by CO dehydrogenase (CODH) and the CO-sensing mechanisms that enable CO oxidation; 2) the use of CO as a heme ligand in microbial biochemistry; and 3) very limited information on how microbes respond to CO toxicity. We demonstrate how our horizons in CO biology have been extended by intense research activity in recent years in mammalian and human physiology and biochemistry. CO is one of several "new" small gas molecules that are increasingly recognized for their profound and often beneficial biological activities, the others being nitric oxide (NO) and hydrogen sulfide (H2S). The chemistry of CO and other heme ligands (oxygen, NO, H2S and cyanide) and the implications for biological interactions are briefly presented. An important advance in recent years has been the development of CO-releasing molecules (CO-RMs) for aiding experimental administration of CO as an alternative to the use of CO gas. The chemical principles of CO-RM design and mechanisms of CO release from CO-RMs (dissociation, association, reduction and oxidation, photolysis, and acidification) are reviewed and we present a survey of the most commonly used CO-RMs. Amongst the most important new applications of CO in mammalian physiology and medicine are its vasoactive properties and the therapeutic potentials of CO-RMs in vascular disease, anti-inflammatory effects, CO-mediated cell signaling in apoptosis, applications in organ preservation, and the effects of CO on mitochondrial function. The very limited literature on microbial growth responses to CO and CO-RMs in vitro, and the transcriptomic and physiological consequences of microbial exposure to CO and CO-RMs are reviewed. There is current interest in CO and CO-RMs as antimicrobial agents, particularly in the control of bacterial infections. Future prospects are suggested and unanswered questions posed.
Collapse
|
47
|
Yasokawa D, Murata S, Iwahashi Y, Kitagawa E, Kishi K, Okumura Y, Iwahashi H. DNA microarray analysis suggests that zinc pyrithione causes iron starvation to the yeast Saccharomyces cerevisiae. J Biosci Bioeng 2009; 109:479-86. [PMID: 20347771 DOI: 10.1016/j.jbiosc.2009.10.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 10/30/2009] [Accepted: 10/30/2009] [Indexed: 10/20/2022]
Abstract
Zinc pyrithione has been used in anti-dandruff shampoos and in anti-fouling paint on ships. However, little is known of its mode of action. We characterized the effects of sub-lethal concentrations of zinc pyrithione (Zpt) on Saccharomyces cerevisiae using DNA microarrays. The majority of the strongly upregulated genes are related to iron transport, and many of the strongly downregulated genes are related to the biosynthesis of cytochrome (heme). These data suggest that Zpt induces severe iron starvation. To confirm the DNA microarray data, we supplemented cultures containing Zpt with iron, and the growth of the yeast was restored significantly. From these results, we propose that the principal toxicity of zinc pyrithione arises from iron starvation.
Collapse
Affiliation(s)
- Daisuke Yasokawa
- Hokkaido Food Processing Research Center, Department of Food Biotechnology, 589-4 Bunkyodai Midorimachi, Ebetsu, Hokkaido, 0690836, Japan.
| | | | | | | | | | | | | |
Collapse
|
48
|
Maghzal GJ, Leck MC, Collinson E, Li C, Stocker R. Limited role for the bilirubin-biliverdin redox amplification cycle in the cellular antioxidant protection by biliverdin reductase. J Biol Chem 2009; 284:29251-9. [PMID: 19690164 DOI: 10.1074/jbc.m109.037119] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In mammalian cells, heme is degraded by heme oxygenase to biliverdin, which is then reduced to bilirubin by biliverdin reductase (BVR). Both bile pigments have reducing properties, and bilirubin is now generally considered to be a potent antioxidant, yet it remains unclear how it protects cells against oxidative damage. A presently popular explanation for the antioxidant function of bilirubin is a redox cycle in which bilirubin is oxidized to biliverdin and then recycled by BVR. Here, we reexamined this putative BVR-mediated redox cycle. We observed that lipid peroxidation-mediated oxidation of bilirubin in chloroform, a model of cell membrane-bound bilirubin, did not yield biliverdin, a prerequisite for the putative redox cycle. Similarly, H(2)O(2) did not oxidize albumin-bound bilirubin to biliverdin, and in vitro oxidation of albumin or ligandin-bound bilirubin by peroxyl radicals gave modest yields of biliverdin. In addition, decreasing cellular BVR protein and activity in HeLa cells using RNA interference did not alter H(2)O(2)-mediated cell death, just as BVR overexpression failed to enhance protection of these cells against H(2)O(2)-mediated damage, irrespective of whether bilirubin or biliverdin were added to the cells as substrate for the putative redox cycle. Similarly, transformation of human BVR into hmx1 (heme oxygenase) mutant yeast did not provide protection against H(2)O(2) toxicity above that seen in hmx1 mutant yeast expressing human heme oxygenase-1. Together, these results argue against the BVR-mediated redox cycle playing a general or important role as cellular antioxidant defense mechanism.
Collapse
Affiliation(s)
- Ghassan J Maghzal
- Centre for Vascular Research, School of Medical Sciences (Pathology) and Bosch Institute, Sydney Medical School, University of Sydney, Medical Foundation Building K25, 92-94 Parramatta Road, Camperdown, New South Wales 2006, Australia
| | | | | | | | | |
Collapse
|
49
|
Evidence of carbon monoxide-mediated phase advancement of the yeast metabolic cycle. Proc Natl Acad Sci U S A 2009; 106:14293-6. [PMID: 19706514 DOI: 10.1073/pnas.0907786106] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Prototrophic strains of budding yeast exhibit robust metabolic cycles during continuous growth under nutrient-limiting conditions. Previous studies revealed periodic fluctuations of aminolevulinic acid, a precursor of heme, indicating that heme biosynthesis is temporally regulated during these metabolic cycles. The enzyme that catabolizes heme, heme oxygenase, was found to be expressed in a highly periodic manner at both the mRNA and protein level. Heme oxygenase generates the biological gas, carbon monoxide (CO), as a product of heme catabolism. It is shown that pulsed administration of CO induces a phase advancement into the oxidative, respiratory phase of the metabolic cycles. This CO-mediated phase advancement takes place only if the gas is administered during the temporal window when it is predicted to be generated. It is further shown that a yeast strain bearing a targeted deletion of the gene encoding heme oxygenase displays protracted metabolic cycles. These observations provide evidence that gaseous CO may function as a cellular signaling molecule that helps cue metabolic cycling.
Collapse
|
50
|
KlAft, the Kluyveromyces lactis ortholog of Aft1 and Aft2, mediates activation of iron-responsive transcription through the PuCACCC Aft-type sequence. Genetics 2009; 183:93-106. [PMID: 19581449 DOI: 10.1534/genetics.109.104364] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Iron homeostasis in fungi is regulated at the transcriptional level by two different mechanisms. It is mediated by a conserved GATA-type repressor in most fungi except in the yeast Saccharomyces cerevisiae, where it is controlled by the transcription activators Aft1 and Aft2. These activators are encoded by the paralogous genes AFT1 and AFT2, which result from the whole-genome duplication. Here, we explore regulation of iron homeostasis in the yeast Kluyveromyces lactis that diverged from S. cerevisiae before this event. We identify an ortholog of AFT1/AFT2, designated KlAFT, whose deletion leads to the inability to grow under iron limitation. We show with quantitative real-time PCR analysis that KlAft activates the transcription of all homologs of the Aft1-target genes involved in the iron transport at the cell surface in response to iron limitation. However, homologs of Aft2-specific target genes encoding intracellular iron transporters are regulated neither by KlAft nor by iron. Both bioinformatic and DNA binding and transcription analyses demonstrate that KlAft activates iron-responsive gene expression through the PuCACCC Aft-type sequence. Thus, K. lactis is the first documented species with a positive iron-transcriptional control mediated by only one copy of the Aft-type regulator. This indicates that this function was acquired before the whole-genome duplication and was then diversified into two regulators in S. cerevisiae.
Collapse
|