Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.121
Filter
1.
Methods Mol Biol ; 2839: 195-211, 2024.
Article in English | MEDLINE | ID: mdl-39008254

ABSTRACT

Recombinant expression and biogenesis of cytochrome c species is a simple and efficient method for the production of holocytochrome c species, thus presenting an avenue for the study of cytochrome c or the cytochrome c biogenesis pathways responsible for heme attachment. Here, we describe a method for recombinant E. coli production of holocytochrome c utilizing the System I (CcmABCDEFGH) bacterial cytochrome c biogenesis pathway, followed by analysis of cytochrome c species by cell lysis and heme stain.


Subject(s)
Cytochromes c , Escherichia coli , Heme , Recombinant Proteins , Cytochromes c/genetics , Cytochromes c/metabolism , Cytochromes c/biosynthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Heme/metabolism , Heme/biosynthesis
2.
Methods Mol Biol ; 2839: 213-223, 2024.
Article in English | MEDLINE | ID: mdl-39008255

ABSTRACT

The utilization of ultra-performance liquid chromatography (UPLC) to analyze the various intermediates in the heme biosynthetic pathway is presented. The first product, ALA, was derivatized to a highly fluorescent pyrrolizine; PBG, the second intermediate, was enzymatically converted to uroporphyrinogen, and all the porphyrinogen intermediates were oxidized in acid to form fluorescent porphyrins. Heme was measured as hemin. The stable porphyrin forms of the intermediates, are then resolved and quantified by UPLC. Further details about the various methods are discussed to promote successful UPLC analyses. Method variations that may be preferable in certain situations are also presented.


Subject(s)
Heme , Heme/biosynthesis , Heme/metabolism , Chromatography, High Pressure Liquid/methods , Aminolevulinic Acid/metabolism , Hemin/metabolism , Hemin/chemistry
3.
Commun Biol ; 7(1): 797, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956406

ABSTRACT

The nonconventional yeast Kluyveromyces marxianus has potential for industrial production, but the lack of advanced synthetic biology tools for precise engineering hinders its rapid development. Here, we introduce a CRISPR-Cas9-mediated multilocus integration method for assembling multiple exogenous genes. Using SlugCas9-HF, a high-fidelity Cas9 nuclease, we enhance gene editing precision. Specific genomic loci predisposed to efficient integration and expression of heterologous genes are identified and combined with a set of paired CRISPR-Cas9 expression plasmids and donor plasmids to establish a CRISPR-based biosynthesis toolkit. This toolkit enables genome integration of large gene modules over 12 kb and achieves simultaneous quadruple-locus integration in a single step with 20% efficiency. As a proof-of-concept, we apply the toolkit to screen for gene combinations that promote heme production, revealing the importance of HEM4Km and HEM12Sc. This CRISPR-based toolkit simplifies the reconstruction of complex pathways in K. marxianus, broadening its application in synthetic biology.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Kluyveromyces , Kluyveromyces/genetics , Gene Editing/methods , Plasmids/genetics , Synthetic Biology/methods , Heme/metabolism , Heme/genetics , Heme/biosynthesis
4.
Metab Eng ; 84: 59-68, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38839038

ABSTRACT

The development of a heme-responsive biosensor for dynamic pathway regulation in eukaryotes has never been reported, posing a challenge for achieving the efficient synthesis of multifunctional hemoproteins and maintaining intracellular heme homeostasis. Herein, a biosensor containing a newly identified heme-responsive promoter, CRISPR/dCas9, and a degradation tag N-degron was designed and optimized to fine-tune heme biosynthesis in the efficient heme-supplying Pichia pastoris P1H9 chassis. After identifying literature-reported promoters insensitive to heme, the endogenous heme-responsive promoters were mined by transcriptomics, and an optimal biosensor was screened from different combinations of regulatory elements. The dynamic regulation pattern of the biosensor was validated by the transcriptional fluctuations of the HEM2 gene involved in heme biosynthesis and the subsequent responsive changes in intracellular heme titers. We demonstrate the efficiency of this regulatory system by improving the production of high-active porcine myoglobin and soy hemoglobin, which can be used to develop artificial meat and artificial metalloenzymes. Moreover, these findings can offer valuable strategies for the synthesis of other hemoproteins.


Subject(s)
Biosensing Techniques , Heme , Hemeproteins , Heme/biosynthesis , Heme/genetics , Heme/metabolism , Hemeproteins/genetics , Hemeproteins/metabolism , Hemeproteins/biosynthesis , Transcriptome/genetics , Saccharomycetales/genetics , Saccharomycetales/metabolism , Animals , CRISPR-Cas Systems , Metabolic Engineering , Promoter Regions, Genetic
5.
FEBS J ; 291(16): 3737-3748, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38865576

ABSTRACT

Hemoglobins, with heme as a cofactor, are functional proteins that have extensive applications in the fields of artificial oxygen carriers and foods. Although Saccharomyces cerevisiae is an ideal host for hemoglobin synthesis, it lacks a suitable transport system to utilize additional heme for active expression of hemoglobins, resulting in the cellular aggregation and degradation of the latter. Here, an effective heme importer, heme-responsive gene 4 (Hrg-4), was selected from six candidates through the comparison of effects on the growth rates of Δhem1 S. cerevisiae strain and the activities of various hemoglobins when supplemented with 5 mg·L-1 exogenous heme. Additionally, to counter the instability of plasmid-based expression and the metabolic burden introduced from overexpressing Hrg-4, a series of hrg-4 integrated strains were constructed and the best engineered strain with five copies of hrg-4 was chosen. We found that this engineered strain was associated with an increased binding rate of heme in monomeric leghemoglobin and multimeric human hemoglobin (76.3% and 16.5%, respectively), as well as an enhanced expression of both hemoglobins (52.8% and 17.0%, respectively). Thus, the engineered strain with improved heme uptake can be used to efficiently synthesize other heme-binding proteins and enzymes in S. cerevisiae.


Subject(s)
Heme , Hemoglobins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Heme/metabolism , Heme/biosynthesis , Hemoglobins/genetics , Hemoglobins/metabolism , Humans , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Leghemoglobin/metabolism , Leghemoglobin/genetics , Biological Transport
6.
Int J Mol Sci ; 25(10)2024 May 12.
Article in English | MEDLINE | ID: mdl-38791308

ABSTRACT

Heme biosynthesis is a highly conserved pathway from bacteria to higher animals. Heme, which serves as a prosthetic group for various enzymes involved in multiple biochemical processes, is essential in almost all species, making heme homeostasis vital for life. However, studies on the biological functions of heme in filamentous fungi are scarce. In this study, we investigated the role of heme in Fusarium graminearum. A mutant lacking the rate-limiting enzymes in heme synthesis, coproporphyrinogen III oxidase (Cpo) or ferrochelatase (Fc), was constructed using a homologous recombination strategy. The results showed that the absence of these enzymes was lethal to F. graminearum, but the growth defect could be rescued by the addition of hemin, so we carried out further studies with the help of hemin. The results demonstrated that heme was required for the activity of FgCyp51, and its absence increased the sensitivity to tebuconazole and led to the upregulation of FgCYP51 in F. graminearum. Additionally, heme plays an indispensable role in the life cycle of F. graminearum, which is essential for vegetative growth, conidiation, external stress response (especially oxidative stress), lipid accumulation, fatty acid ß-oxidation, autophagy, and virulence.


Subject(s)
Fusarium , Heme , Fusarium/drug effects , Fusarium/metabolism , Fusarium/growth & development , Fusarium/genetics , Heme/biosynthesis , Heme/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Stress, Physiological , Oxidative Stress/drug effects , Triazoles/pharmacology , Gene Expression Regulation, Fungal/drug effects , Fungicides, Industrial/pharmacology , Ferrochelatase/metabolism , Ferrochelatase/genetics
7.
Mol Biol (Mosk) ; 57(6): 1085-1097, 2023.
Article in Russian | MEDLINE | ID: mdl-38062963

ABSTRACT

δ-Aminolevulinic acid dehydratase (ALAD) is a key enzyme of the cytoplasmic heme biosynthesis pathway. The primary structure of the ALAD gene, the multimeric structure of the ALAD/hemB protein, and ALAD expression during the annual reproductive cycle were studied in the cold-water marine sponge Halisarca dujardinii. The results implicated the GATA-1 transcription factor and DNA methylation in regulating ALAD expression. Re-aggregation of sponge cells was accompanied by a decrease in ALAD expression and a change in the cell content of an active ALAD/hemB form. Further study of heme biosynthesis and the role of ALAD/hemB in morphogenesis of basal animals may provide new opportunities for treating pathologies in higher animals.


Subject(s)
Porifera , Animals , Heme/biosynthesis , Heme/metabolism , Porifera/enzymology , Porifera/metabolism , Porphobilinogen Synthase/genetics , Porphobilinogen Synthase/metabolism
8.
Proc Natl Acad Sci U S A ; 119(30): e2108245119, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35858410

ABSTRACT

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.


Subject(s)
Heme , Metabolic Engineering , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Computer Simulation , Heme/biosynthesis , Heme/genetics , Hemeproteins/biosynthesis , Hemeproteins/genetics , Metabolic Engineering/methods , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
9.
Sci Rep ; 12(1): 1472, 2022 01 27.
Article in English | MEDLINE | ID: mdl-35087136

ABSTRACT

Obstructive sleep apnea (OSA) is a common disorder associated with increased risk of cardiovascular disease and mortality. Iron and heme metabolism, implicated in ventilatory control and OSA comorbidities, was associated with OSA phenotypes in recent admixture mapping and gene enrichment analyses. However, its causal contribution was unclear. In this study, we performed pathway-level transcriptional Mendelian randomization (MR) analysis to investigate the causal relationships between iron and heme related pathways and OSA. In primary analysis, we examined the expression level of four iron/heme Reactome pathways as exposures and four OSA traits as outcomes using cross-tissue cis-eQTLs from the Genotype-Tissue Expression portal and published genome-wide summary statistics of OSA. We identify a significant putative causal association between up-regulated heme biosynthesis pathway with higher sleep time percentage of hypoxemia (p = 6.14 × 10-3). This association is supported by consistency of point estimates in one-sample MR in the Multi-Ethnic Study of Atherosclerosis using high coverage DNA and RNA sequencing data generated by the Trans-Omics for Precision Medicine project. Secondary analysis for 37 additional iron/heme Gene Ontology pathways did not reveal any significant causal associations. This study suggests a causal association between increased heme biosynthesis and OSA severity.


Subject(s)
Heme/biosynthesis , Metabolic Networks and Pathways/genetics , Sleep Apnea, Obstructive/epidemiology , Aged , Datasets as Topic , Female , Genetic Predisposition to Disease , Humans , Iron/metabolism , Male , Mendelian Randomization Analysis , Middle Aged , Polysomnography , Quantitative Trait Loci , Severity of Illness Index , Sleep Apnea, Obstructive/blood , Sleep Apnea, Obstructive/diagnosis , Sleep Apnea, Obstructive/genetics , Up-Regulation
10.
Am J Pathol ; 192(1): 4-17, 2022 01.
Article in English | MEDLINE | ID: mdl-34924168

ABSTRACT

Metabolic heterogeneity or functional zonation is a key characteristic of the liver that allows different metabolic pathways to be spatially regulated within the hepatic system and together contribute to whole body homeostasis. These metabolic pathways are segregated along the portocentral axis of the liver lobule into three hepatic zones: periportal, intermediate or midzonal, and perivenous. The liver performs complementary or opposing metabolic functions within different hepatic zones while synergistic functions are regulated by overlapping zones, thereby maintaining the overall physiological stability. The Wnt/ß-catenin signaling pathway is well known for its role in liver growth, development, and regeneration. In addition, the Wnt/ß-catenin pathway plays a fundamental and dominant role in hepatic zonation and signals to orchestrate various functions of liver metabolism and pathophysiology. The ß-catenin protein is the central player in the Wnt/ß-catenin signaling cascade, and its activation is crucial for metabolic patterning of the liver. However, dysregulation of Wnt/ß-catenin signaling is also implicated in different liver pathologies, including those associated with metabolic syndrome. ß-Catenin is preferentially localized in the central region of the hepatic lobule surrounding the central vein and regulates multiple functions of this region. This review outlines the role of Wnt/ß-catenin signaling pathway in controlling the different metabolic processes surrounding the central vein and its relation to liver homeostasis and dysfunction.


Subject(s)
Homeostasis , Liver/metabolism , Wnt Signaling Pathway , Animals , Gene Expression Regulation , Heme/biosynthesis , Humans , Wnt Signaling Pathway/genetics , Xenobiotics/metabolism
11.
Biomolecules ; 11(12)2021 12 20.
Article in English | MEDLINE | ID: mdl-34944556

ABSTRACT

This study hypothesizes that bacteria inhabiting shale rock affect the content of the sedimentary cobalt protoporphyrin present in it and can use it as a precursor for heme synthesis. To verify this hypothesis, we conducted qualitative and quantitative comparative analyses of cobalt protoporphyrin as well as heme, and heme iron in shale rock that were (i) inhabited by bacteria in the field, (ii) treated with bacteria in the laboratory, and with (iii) bacterial culture on synthetic cobalt protoporphyrin. Additionally, we examined the above-mentioned samples for the presence of enzymes involved in the heme biosynthesis and uptake as well as hemoproteins. We found depletion of cobalt protoporphyrin and a much higher heme concentration in the shale rock inhabited by bacteria in the field as well as the shale rock treated with bacteria in the laboratory. Similarly, we observed the accumulation of protoporphyrin in bacterial cells grown on synthetic cobalt protoporphyrin. We detected numerous hemoproteins in metaproteome of bacteria inhabited shale rock in the field and in proteomes of bacteria inhabited shale rock and synthetic cobalt protoporhyrin in the laboratory, but none of them had all the enzymes involved in the heme biosynthesis. However, proteins responsible for heme uptake, ferrochelatase and sirohydrochlorin cobaltochelatase/sirohydrochlorin cobalt-lyase were detected in all studied samples.


Subject(s)
Bacteria/growth & development , Fossils/microbiology , Geologic Sediments/microbiology , Heme/analysis , Protoporphyrins/analysis , Bacteria/metabolism , Bacterial Proteins/metabolism , Bacteriological Techniques , Culture Media/chemistry , Ferrochelatase/metabolism , Gene Expression Regulation, Bacterial , Geologic Sediments/chemistry , Heme/biosynthesis , Lyases/metabolism , Proteomics , Protoporphyrins/biosynthesis
12.
Microbiology (Reading) ; 167(10)2021 10.
Article in English | MEDLINE | ID: mdl-34661520

ABSTRACT

Uroporphyrinogen III, the universal progenitor of macrocyclic, modified tetrapyrroles, is produced from aminolaevulinic acid (ALA) by a conserved pathway involving three enzymes: porphobilinogen synthase (PBGS), hydroxymethylbilane synthase (HmbS) and uroporphyrinogen III synthase (UroS). The gene encoding uroporphyrinogen III synthase has not yet been identified in Plasmodium falciparum, but it has been suggested that this activity is housed inside a bifunctional hybroxymethylbilane synthase (HmbS). Additionally, an unknown protein encoded by PF3D7_1247600 has also been predicted to possess UroS activity. In this study it is demonstrated that neither of these proteins possess UroS activity and the real UroS remains to be identified. This was demonstrated by the failure of codon-optimized genes to complement a defined Escherichia coli hemD- mutant (SASZ31) deficient in UroS activity. Furthermore, HPLC analysis of the oxidized reaction product from recombinant, purified P. falciparum HmbS showed that only uroporphyrin I could be detected (corresponding to hydroxymethylbilane production). No uroporphyrin III was detected, showing that P. falciparum HmbS does not have UroS activity and can only catalyze the formation of hydroxymethylbilane from porphobilinogen.


Subject(s)
Heme/biosynthesis , Hydroxymethylbilane Synthase/metabolism , Plasmodium falciparum/enzymology , Biosynthetic Pathways , Escherichia coli/genetics , Genetic Complementation Test , Hydroxymethylbilane Synthase/genetics , Mutation , Plasmodium falciparum/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Uroporphyrinogen III Synthetase/genetics , Uroporphyrinogen III Synthetase/metabolism , Uroporphyrinogens/metabolism
13.
Biomolecules ; 11(9)2021 09 21.
Article in English | MEDLINE | ID: mdl-34572607

ABSTRACT

In the traditional fermentative model yeast Saccharomyces cerevisiae, ScIxr1 is an HMGB (High Mobility Group box B) protein that has been considered as an important regulator of gene transcription in response to external changes like oxygen, carbon source, or nutrient availability. Kluyveromyces lactis is also a useful eukaryotic model, more similar to many human cells due to its respiratory metabolism. We cloned and functionally characterized by different methodologies KlIXR1, which encodes a protein with only 34.4% amino acid sequence similarity to ScIxr1. Our data indicate that both proteins share common functions, including their involvement in the response to hypoxia or oxidative stress induced by hydrogen peroxide or metal treatments, as well as in the control of key regulators for maintenance of the dNTP (deoxyribonucleotide triphosphate) pool and ribosome synthesis. KlIxr1 is able to bind specific regulatory DNA sequences in the promoter of its target genes, which are well conserved between S. cerevisiae and K. lactis. Oppositely, we found important differences between ScIrx1 and KlIxr1 affecting cellular responses to cisplatin or cycloheximide in these yeasts, which could be dependent on specific and non-conserved domains present in these two proteins.


Subject(s)
Deoxyribonucleotides/metabolism , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , HMGB Proteins/metabolism , Kluyveromyces/growth & development , Kluyveromyces/genetics , Base Sequence , Cadmium/toxicity , Carbon/pharmacology , Cell Cycle/drug effects , Cisplatin/pharmacology , Drug Resistance/drug effects , Fungal Proteins/chemistry , Gene Deletion , Gene Expression Regulation, Fungal/drug effects , HMGB Proteins/chemistry , Heme/biosynthesis , Hydrogen Peroxide/toxicity , Kluyveromyces/drug effects , Mutation/genetics , Oxidation-Reduction/drug effects , Phenotype , Promoter Regions, Genetic , Protein Binding/drug effects , RNA Processing, Post-Transcriptional/drug effects , RNA, Ribosomal/genetics , Ribosomes/drug effects , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism
14.
Crit Rev Biochem Mol Biol ; 56(6): 640-668, 2021 12.
Article in English | MEDLINE | ID: mdl-34428995

ABSTRACT

Aerobic respiration is a key energy-producing pathway in many prokaryotes and virtually all eukaryotes. The final step of aerobic respiration is most commonly catalyzed by heme-copper oxidases embedded in the cytoplasmic or mitochondrial membrane. The majority of these terminal oxidases contain a prenylated heme (typically heme a or occasionally heme o) in the active site. In addition, many heme-copper oxidases, including mitochondrial cytochrome c oxidases, possess a second heme a cofactor. Despite the critical role of heme a in the electron transport chain, the details of the mechanism by which heme b, the prototypical cellular heme, is converted to heme o and then to heme a remain poorly understood. Recent structural investigations, however, have helped clarify some elements of heme a biosynthesis. In this review, we discuss the insight gained from these advances. In particular, we present a new structural model of heme o synthase (HOS) based on distance restraints from inferred coevolutionary relationships and refined by molecular dynamics simulations that are in good agreement with the experimentally determined structures of HOS homologs. We also analyze the two structures of heme a synthase (HAS) that have recently been solved by other groups. For both HOS and HAS, we discuss the proposed catalytic mechanisms and highlight how new insights into the heme-binding site locations shed light on previously obtained biochemical data. Finally, we explore the implications of the new structural data in the broader context of heme trafficking in the heme a biosynthetic pathway and heme-copper oxidase assembly.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Bacterial Proteins/metabolism , Heme/analogs & derivatives , Animals , Archaea/metabolism , Bacteria/metabolism , Electron Transport Complex IV/metabolism , Eukaryota/metabolism , Heme/biosynthesis , Heme/metabolism , Humans , Molecular Dynamics Simulation , Protein Conformation , Protein Transport
15.
Biochem J ; 478(17): 3239-3252, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34402499

ABSTRACT

Ferrochelatase catalyzes the insertion of ferrous iron into a porphyrin macrocycle to produce the essential cofactor, heme. In humans this enzyme not only catalyzes the terminal step, but also serves a regulatory step in the heme synthesis pathway. Over a dozen crystal structures of human ferrochelatase have been solved and many variants have been characterized kinetically. In addition, hydrogen deuterium exchange, resonance Raman, molecular dynamics, and high level quantum mechanic studies have added to our understanding of the catalytic cycle of the enzyme. However, an understanding of how the metal ion is delivered and the specific role that active site residues play in catalysis remain open questions. Data are consistent with metal binding and insertion occurring from the side opposite from where pyrrole proton abstraction takes place. To better understand iron delivery and binding as well as the role of conserved residues in the active site, we have constructed and characterized a series of enzyme variants. Crystallographic studies as well as rescue and kinetic analysis of variants were performed. Data from these studies are consistent with the M76 residue playing a role in active site metal binding and formation of a weak iron protein ligand being necessary for product release. Additionally, structural data support a role for E343 in proton abstraction and product release in coordination with a peptide loop composed of Q302, S303 and K304 that act a metal sensor.


Subject(s)
Catalytic Domain/physiology , Ferrochelatase/chemistry , Ferrochelatase/metabolism , Models, Molecular , Biocatalysis , Crystallization , Heme/biosynthesis , Histidine/metabolism , Humans , Iron/metabolism , Kinetics , Ligands , Protein Binding , Protons , Protoporphyrins/metabolism
16.
J Biol Chem ; 297(2): 100972, 2021 08.
Article in English | MEDLINE | ID: mdl-34280433

ABSTRACT

Heme plays a critical role in catalyzing life-essential redox reactions in all cells, and its synthesis must be tightly balanced with cellular requirements. Heme synthesis in eukaryotes is tightly regulated by the mitochondrial AAA+ unfoldase CLPX (caseinolytic mitochondrial matrix peptidase chaperone subunit X), which promotes heme synthesis by activation of δ-aminolevulinate synthase (ALAS/Hem1) in yeast and regulates turnover of ALAS1 in human cells. However, the specific mechanisms by which CLPX regulates heme synthesis are unclear. In this study, we interrogated the mechanisms by which CLPX regulates heme synthesis in erythroid cells. Quantitation of enzyme activity and protein degradation showed that ALAS2 stability and activity were both increased in the absence of CLPX, suggesting that CLPX primarily regulates ALAS2 by control of its turnover, rather than its activation. However, we also showed that CLPX is required for PPOX (protoporphyrinogen IX oxidase) activity and maintenance of FECH (ferrochelatase) levels, which are the terminal enzymes in heme synthesis, likely accounting for the heme deficiency and porphyrin accumulation observed in Clpx-/- cells. Lastly, CLPX is required for iron utilization for hemoglobin synthesis during erythroid differentiation. Collectively, our data show that the role of CLPX in yeast ALAS/Hem1 activation is not conserved in vertebrates as vertebrates rely on CLPX to regulate ALAS turnover as well as PPOX and FECH activity. Our studies reveal that CLPX mutations may cause anemia and porphyria via dysregulation of ALAS, FECH, and PPOX activities, as well as of iron metabolism.


Subject(s)
5-Aminolevulinate Synthetase/metabolism , Endopeptidase Clp/metabolism , Ferrochelatase/metabolism , Heme/biosynthesis , Iron/metabolism , Leukemia, Erythroblastic, Acute/pathology , Mitochondria/metabolism , Animals , Cell Line, Tumor , Endopeptidase Clp/genetics , Enzyme Activation , Gene Knockout Techniques/methods , Leukemia, Erythroblastic, Acute/enzymology , Leukemia, Erythroblastic, Acute/genetics , Mice , Models, Animal , Proteolysis , Zebrafish
17.
Genome Med ; 13(1): 108, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34225776

ABSTRACT

BACKGROUND: Candidemia is one of the most common nosocomial bloodstream infections in the United States, causing significant morbidity and mortality in hospitalized patients, but the breadth of the host response to Candida infections in human patients remains poorly defined. METHODS: In order to better define the host response to Candida infection at the transcriptional level, we performed RNA sequencing on serial peripheral blood samples from 48 hospitalized patients with blood cultures positive for Candida species and compared them to patients with other acute viral, bacterial, and non-infectious illnesses. Regularized multinomial regression was utilized to develop pathogen class-specific gene expression classifiers. RESULTS: Candidemia triggers a unique, robust, and conserved transcriptomic response in human hosts with 1641 genes differentially upregulated compared to healthy controls. Many of these genes corresponded to components of the immune response to fungal infection, heavily weighted toward neutrophil activation, heme biosynthesis, and T cell signaling. We developed pathogen class-specific classifiers from these unique signals capable of identifying and differentiating candidemia, viral, or bacterial infection across a variety of hosts with a high degree of accuracy (auROC 0.98 for candidemia, 0.99 for viral and bacterial infection). This classifier was validated on two separate human cohorts (auROC 0.88 for viral infection and 0.87 for bacterial infection in one cohort; auROC 0.97 in another cohort) and an in vitro model (auROC 0.94 for fungal infection, 0.96 for bacterial, and 0.90 for viral infection). CONCLUSIONS: Transcriptional analysis of circulating leukocytes in patients with acute Candida infections defines novel aspects of the breadth of the human immune response during candidemia and suggests promising diagnostic approaches for simultaneously differentiating multiple types of clinical illnesses in at-risk, acutely ill patients.


Subject(s)
Candidemia/etiology , Candidemia/metabolism , Disease Susceptibility , Heme/biosynthesis , Host-Pathogen Interactions/genetics , Neutrophil Activation/genetics , Transcriptome , Adult , Aged , Aged, 80 and over , Biomarkers , Candidemia/diagnosis , Candidemia/drug therapy , Case-Control Studies , Computational Biology/methods , Databases, Genetic , Female , Gene Expression Profiling , Host-Pathogen Interactions/immunology , Humans , Male , Middle Aged , Neutrophil Activation/immunology , Prognosis , ROC Curve , Reproducibility of Results , Risk Factors , Severity of Illness Index
18.
Cell Rep ; 35(11): 109252, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34133926

ABSTRACT

Heme is an iron-containing porphyrin of vital importance for cell energetic metabolism. High rates of heme synthesis are commonly observed in proliferating cells. Moreover, the cell-surface heme exporter feline leukemia virus subgroup C receptor 1a (FLVCR1a) is overexpressed in several tumor types. However, the reasons why heme synthesis and export are enhanced in highly proliferating cells remain unknown. Here, we illustrate a functional axis between heme synthesis and heme export: heme efflux through the plasma membrane sustains heme synthesis, and implementation of the two processes down-modulates the tricarboxylic acid (TCA) cycle flux and oxidative phosphorylation. Conversely, inhibition of heme export reduces heme synthesis and promotes the TCA cycle fueling and flux as well as oxidative phosphorylation. These data indicate that the heme synthesis-export system modulates the TCA cycle and oxidative metabolism and provide a mechanistic basis for the observation that both processes are enhanced in cells with high-energy demand.


Subject(s)
Citric Acid Cycle , Heme/biosynthesis , Oxidative Phosphorylation , Animals , Biological Transport , Caco-2 Cells , Heme/metabolism , Humans , Membrane Transport Proteins/metabolism , Mice, Inbred C57BL , Mice, SCID , Receptors, Virus/metabolism , Xenograft Model Antitumor Assays
19.
Int J Mol Sci ; 22(10)2021 May 11.
Article in English | MEDLINE | ID: mdl-34064887

ABSTRACT

Bacterial response to nitric oxide (NO) is of major importance for bacterial survival. NO stress is a main actor of the eukaryotic immune response and several pathogenic bacteria have developed means for detoxification and repair of the damages caused by NO. However, bacterial mechanisms of NO resistance by Gram-positive bacteria are poorly described. In the opportunistic foodborne pathogen Bacillus cereus, genome sequence analyses did not identify homologs to known NO reductases and transcriptional regulators, such as NsrR, which orchestrate the response to NO of other pathogenic or non-pathogenic bacteria. Using a transcriptomic approach, we investigated the adaptation of B. cereus to NO stress. A cluster of 6 genes was identified to be strongly up-regulated in the early phase of the response. This cluster contains an iron-sulfur cluster repair enzyme, a nitrite reductase and three enzymes involved in siroheme biosynthesis. The expression pattern and close genetic localization suggest a functional link between these genes, which may play a pivotal role in the resistance of B. cereus to NO stress during infection.


Subject(s)
Bacillus cereus/metabolism , Bacterial Proteins/metabolism , Heme/analogs & derivatives , Iron/metabolism , Nitric Oxide/toxicity , Nitrite Reductases/metabolism , Oxidative Stress , Bacillus cereus/drug effects , Bacillus cereus/genetics , Bacillus cereus/growth & development , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Heme/biosynthesis , Transcription, Genetic
20.
Sci Rep ; 11(1): 7457, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33811225

ABSTRACT

Isolation of bacterial small colony variants (SCVs) from clinical specimens is not uncommon and can fundamentally change the outcome of the associated infections. Bacterial SCVs often emerge with their normal colony phenotype (NCV) co-isolates in the same sample. The basis of SCV emergence in vivo is not well understood in Gram-negative bacteria. In this study, we interrogated the causal genetic lesions of SCV growth in three pairs of NCV and SCV co-isolates of Escherichia coli, Citrobacter freundii, and Enterobacter hormaechei. We confirmed SCV emergence was attributed to limited genomic mutations: 4 single nucleotide variants in the E. coli SCV, 5 in C. freundii, and 8 in E. hormaechei. In addition, a 10.2 kb chromosomal segment containing 11 genes was deleted in the E. hormaechei SCV isolate. Each SCV had at least one coding change in a gene associated with bacterial oxidative respiration and another involved in iron capture. Chemical and genetic rescue confirmed defects in heme biosynthesis for E. coli and C. freundii and lipoic acid biosynthesis in E. hormaachei were responsible for the SCV phenotype. Prototrophic growth in all 3 SCV Enterobacteriaceae species was unaffected under anaerobic culture conditions in vitro, illustrating how SCVs may persist in vivo.


Subject(s)
Enterobacteriaceae/genetics , Enterobacteriaceae/isolation & purification , Gene Silencing , Genes, Bacterial , Iron/metabolism , Aerobiosis/genetics , Anaerobiosis/genetics , Biosynthetic Pathways/genetics , Child , Colony Count, Microbial , Drug Resistance, Bacterial/genetics , Enterobacteriaceae/growth & development , Female , Genetic Variation , Heme/biosynthesis , Humans , Infant , Kinetics , Male , Microbial Sensitivity Tests , Phenotype , Thioctic Acid/biosynthesis , Whole Genome Sequencing
SELECTION OF CITATIONS
SEARCH DETAIL