Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Cell ; 145(5): 720-31, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21620137

ABSTRACT

Extracellular free heme can intercalate into membranes and promote damage to cellular macromolecules. Thus it is likely that specific intercellular pathways exist for the directed transport, trafficking, and delivery of heme to cellular destinations, although none have been found to date. Here we show that Caenorhabditis elegans HRG-3 is required for the delivery of maternal heme to developing embryos. HRG-3 binds heme and is exclusively secreted by maternal intestinal cells into the interstitial fluid for transport of heme to extraintestinal cells, including oocytes. HRG-3 deficiency results either in death during embryogenesis or in developmental arrest immediately post-hatching-phenotypes that are fully suppressed by maternal but not zygotic hrg-3 expression. Our results establish a role for HRG-3 as an intercellular heme-trafficking protein.


Subject(s)
Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Heme/metabolism , Hemeproteins/metabolism , Animals , Animals, Genetically Modified , Gene Expression Regulation, Developmental , Genes, Reporter , Heme/deficiency , Hemeproteins/chemistry , Hemeproteins/genetics , Intestinal Mucosa/metabolism , Mutation , Phenotype , Protein Transport , Secretory Pathway
2.
Blood ; 137(7): 945-958, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33254233

ABSTRACT

Isocitrate dehydrogenase (IDH) mutations are common genetic alterations in myeloid disorders, including acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Epigenetic changes, including abnormal histone and DNA methylation, have been implicated in the pathogenic build-up of hematopoietic progenitors, but it is still unclear whether and how IDH mutations themselves affect hematopoiesis. Here, we show that IDH1-mutant mice develop myeloid dysplasia in that these animals exhibit anemia, ineffective erythropoiesis, and increased immature progenitors and erythroblasts. In erythroid cells of these mice, D-2-hydroxyglutarate, an aberrant metabolite produced by the mutant IDH1 enzyme, inhibits oxoglutarate dehydrogenase activity and diminishes succinyl-coenzyme A (CoA) production. This succinyl-CoA deficiency attenuates heme biosynthesis in IDH1-mutant hematopoietic cells, thus blocking erythroid differentiation at the late erythroblast stage and the erythroid commitment of hematopoietic stem cells, while the exogenous succinyl-CoA or 5-ALA rescues erythropoiesis in IDH1-mutant erythroid cells. Heme deficiency also impairs heme oxygenase-1 expression, which reduces levels of important heme catabolites such as biliverdin and bilirubin. These deficits result in accumulation of excessive reactive oxygen species that induce the cell death of IDH1-mutant erythroid cells. Our results clearly show the essential role of IDH1 in normal erythropoiesis and describe how its mutation leads to myeloid disorders. These data thus have important implications for the devising of new treatments for IDH-mutant tumors.


Subject(s)
Erythropoiesis/genetics , Hematopoietic Stem Cells/metabolism , Heme/biosynthesis , Isocitrate Dehydrogenase/genetics , Mutation, Missense , Point Mutation , Preleukemia/genetics , Acyl Coenzyme A/biosynthesis , Acyl Coenzyme A/deficiency , Anemia/genetics , Animals , Bone Marrow/pathology , Erythroblasts/metabolism , Gene Knock-In Techniques , Glutarates/metabolism , Heme/deficiency , Heme Oxygenase-1/metabolism , Isocitrate Dehydrogenase/physiology , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/pathology , Myelopoiesis/genetics , Preleukemia/metabolism , Preleukemia/pathology , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Splenomegaly/etiology , Thrombocytopenia/genetics
3.
Ann Hematol ; 100(7): 1685-1693, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34050373

ABSTRACT

Givosiran is a novel approach to treat patients with acute intermittent porphyrias (AIP) by silencing of ∂-ALA-synthase 1, the first enzyme of heme biosynthesis in the liver. We included two patients in the Envision study who responded clinically well to this treatment. However, in both patients, therapy had to be discontinued because of severe adverse effects: One patient (A) developed local injection reactions which continued to spread all over her body with increasing number of injections and eventually caused a severe systemic allergic reaction. Patient B was hospitalized because of a fulminant pancreatitis. Searching for possible causes, we also measured the patients plasma homocysteine (Hcy) levels in fluoride-containing collection tubes: by LC-MS/MS unexpectedly, plasma Hcy levels were 100 and 200 in patient A and between 100 and 400Ā Āµmol/l in patient B. Searching for germline mutations in 10 genes that are relevant for homocysteine metabolism only revealed hetero- and homozygous polymorphisms in the MTHFR gene. Alternatively, an acquired inhibition of cystathionine-beta-synthase which is important for homocysteine metabolism could explain the plasma homocysteine increase. This enzyme is heme-dependent: when we gave heme arginate to our patients, Hcy levels rapidly dropped. Hence, we conclude that inhibition of ∂-ALA-synthase 1 by givosiran causes a drop of free heme in the hepatocyte and therefore the excessive increase of plasma homocysteine. Hyperhomocysteinemia may contribute to the adverse effects seen in givosiran-treated patients which may be due to protein-N-homocysteinylation.


Subject(s)
5-Aminolevulinate Synthetase/antagonists & inhibitors , Acetylgalactosamine/analogs & derivatives , Heme/deficiency , Hyperhomocysteinemia/etiology , Porphyria, Acute Intermittent/drug therapy , Pyrrolidines/therapeutic use , Acetylgalactosamine/adverse effects , Acetylgalactosamine/therapeutic use , Adult , Arginine/therapeutic use , Colitis/etiology , Colon, Sigmoid/pathology , Controlled Clinical Trials as Topic , Drug Hypersensitivity/etiology , Female , Fibrosis , Heme/analysis , Heme/therapeutic use , Hepatocytes/drug effects , Hepatocytes/metabolism , High-Throughput Nucleotide Sequencing , Homocysteine/metabolism , Humans , Hydroxymethylbilane Synthase/blood , Hydroxymethylbilane Synthase/genetics , Male , Models, Biological , Pancreatitis/etiology , Porphyria, Acute Intermittent/blood , Porphyria, Acute Intermittent/complications , Porphyria, Acute Intermittent/genetics , Pyrrolidines/adverse effects
4.
J Inherit Metab Dis ; 44(4): 961-971, 2021 07.
Article in English | MEDLINE | ID: mdl-33861472

ABSTRACT

Acute intermittent porphyria (AIP) is a rare metabolic disease caused by mutations within the hydroxymethylbilane synthase gene. Previous studies have reported increased levels of plasma total homocysteine (tHcy) in symptomatic AIP patients. In this study, we present long-term data for tHcy and related parameters for an AIP patient cohort (nĀ =Ā 37) in different clinical disease-states. In total, 25 patients (68%) presented with hyperhomocysteinemia (HHcy; tHcy > 15 Āµmol/L) during the observation period. HHcy was more frequent in AIP patients with recurrent disease receiving heme arginate, than in nonrecurrent (median tHcy: 21.6Ā Āµmol/L; range: 10-129 vs median tHcy: 14.5Ā Āµmol/L; range 6-77). Long-term serial analyses showed a high within-person tHcy variation, especially among the recurrent patients (coefficient of variation: 16.4%-78.8%). HHcy was frequently associated with low blood concentrations of pyridoxal-5'-phosphate and folate, while cobalamin concentration and the allele distribution of the methylene-tetrahydrofolate-reductase gene were normal. Strikingly, 6 out of the 9 recurrent patients who were later included in a regime of givosiran, a small-interfering RNA that effectively reduced recurrent attacks, showed further increased tHcy (median tHcy in 9 patients: 105 Āµmol/L; range 16-212). Screening of amino acids in plasma by liquid-chromatography showed co-increased levels of methionine (median 71 Āµmol/L; range 23-616; normal <40), suggestive of acquired deficiency of cystathionine-Ɵ-synthase. The kynunerine/tryptophan ratio in plasma was, however, normal, indicating a regular metabolism of tryptophan by heme-dependent enzymes. In conclusion, even if HHcy was observed in AIP patients receiving heme arginate, givosiran induced an aggravation of the dysregulation, causing a co-increase of tHcy and methionine resembling classic homocystinuria.


Subject(s)
Acetylgalactosamine/analogs & derivatives , Arginine/deficiency , Heme/deficiency , Hyperhomocysteinemia/etiology , Porphyria, Acute Intermittent/drug therapy , Pyrrolidines/therapeutic use , Acetylgalactosamine/adverse effects , Acetylgalactosamine/therapeutic use , Adult , Arginine/therapeutic use , Cystathionine beta-Synthase/genetics , Female , Folic Acid/blood , Heme/therapeutic use , Homeostasis , Homocysteine/metabolism , Homocystinuria/complications , Humans , Hydroxymethylbilane Synthase/blood , Hydroxymethylbilane Synthase/genetics , Male , Methionine/blood , Middle Aged , Porphyria, Acute Intermittent/blood , Porphyria, Acute Intermittent/complications , Porphyria, Acute Intermittent/genetics , Pyridoxal Phosphate/blood , Pyrrolidines/adverse effects , Young Adult
5.
BMC Genomics ; 21(1): 604, 2020 Aug 31.
Article in English | MEDLINE | ID: mdl-32867680

ABSTRACT

BACKGROUND: Aedes aegypti is the principle vector of many arboviruses, including dengue virus and Zika virus, which are transmitted when an infected female mosquito takes a blood meal in order to initiate vitellogenesis. During blood digestion, ~ 10 mM heme-iron is ingested into the midgut lumen. While heme acts as both a nutrient and signaling molecule during blood digestion, it can also be highly toxic if left unchaperoned. Both signaling by, and degradation of, heme are intracellular processes, occurring in the nucleus and cytoplasm, respectively. However, the precise mechanism of heme uptake into the midgut epithelium is not currently known. RESULTS: We used next generation RNA sequencing with the goal to identify genes that code for membrane bound heme import protein(s) responsible for heme uptake into the midgut epithelium. Heme deprivation increased uptake of a heme fluorescent analog in cultured cells, while treatment of midguts with an excess of heme decreased uptake, confirming physiological changes were occurring in these heme-sensitive cells/tissues prior to sequencing. A list of candidate genes was assembled for each of the experimental sample sets, which included Aag2 and A20 cultured cells as well as midgut tissue, based on the results of a differential expression analysis, soft cluster analysis and number of predicted transmembrane domains. Lastly, the functions related to heme transport were examined through RNAi knockdown. CONCLUSIONS: Despite a large number of transmembrane domain containing genes differentially expressed in response to heme, very few were highly differentially expressed in any of the datasets examined. RNAi knockdown of a subset of candidates resulted in subtle changes in heme uptake, but minimal overall disruption to blood digestion/egg production. These results could indicate that heme import in Ae. aegypti may be controlled by a redundant system of multiple distinct transport proteins. Alternatively, heme membrane bound transport in Ae. aegypti could be regulated post-translationally.


Subject(s)
Aedes/genetics , Heme/metabolism , Insect Proteins/genetics , Intestinal Mucosa/metabolism , Membrane Transport Proteins/genetics , Transcriptome , Aedes/metabolism , Animals , Cells, Cultured , Heme/deficiency , Insect Proteins/metabolism , Membrane Transport Proteins/metabolism
6.
PLoS Pathog ; 14(10): e1007355, 2018 10.
Article in English | MEDLINE | ID: mdl-30332468

ABSTRACT

Bacterial pathogens must sense, respond and adapt to a myriad of dynamic microenvironmental stressors to survive. Adaptation is key for colonization and long-term ability to endure fluctuations in nutrient availability and inflammatory processes. We hypothesize that strains adapted to survive nutrient deprivation are more adept for colonization and establishment of chronic infection. In this study, we detected microevolution in response to transient nutrient limitation through mutation of icc. The mutation results in decreased 3',5'-cyclic adenosine monophosphate phosphodiesterase activity in nontypeable Haemophilus influenzae (NTHI). In a preclinical model of NTHI-induced otitis media (OM), we observed a significant decrease in the recovery of effusion from ears infected with the icc mutant strain. Clinically, resolution of OM coincides with the clearance of middle ear fluid. In contrast to this clinical paradigm, we observed that the icc mutant strain formed significantly more intracellular bacterial communities (IBCs) than the parental strain early during experimental OM. Although the number of IBCs formed by the parental strain was low at early stages of OM, we observed a significant increase at later stages that coincided with absence of recoverable effusion, suggesting the presence of a mucosal reservoir following resolution of clinical disease. These data provide the first insight into NTHI microevolution during nutritional limitation and provide the first demonstration of IBCs in a preclinical model of chronic OM.


Subject(s)
Haemophilus Infections/microbiology , Haemophilus influenzae/pathogenicity , Heme/deficiency , Iron Deficiencies , Otitis Media/microbiology , Virulence , Animals , Chinchilla , Disease Models, Animal , Ear, Middle/microbiology , Haemophilus Infections/metabolism , Haemophilus influenzae/genetics , Haemophilus influenzae/isolation & purification , Humans , Otitis Media with Effusion/microbiology , Phosphoric Diester Hydrolases/metabolism
7.
J Sex Med ; 14(2): 196-204, 2017 02.
Article in English | MEDLINE | ID: mdl-28161078

ABSTRACT

INTRODUCTION: The nitric oxide (NO), soluble guanylate cyclase (sGC), and cyclic guanosine monophosphate (cGMP) pathway is the leading pathway in penile erection. AIM: To assess erectile function in a mouse model in which sGC is deficient in heme (apo-sGC) and unresponsive to NO. METHODS: Mutant mice (sGCƟ1ki/ki) that express an sGC enzyme that retains basal activity but fails to respond to NO because of heme deficiency (apo-sGC) were used. Isolated corpora cavernosa from sGCƟ1ki/ki and wild-type mice were mounted inĀ vitro for isometric tension recordings in response to sGC-dependent and -independent vasorelaxant agents. In addition, the erectile effects of some of these agents were tested inĀ vivo at intracavernosal injection. MAIN OUTCOME MEASURES: InĀ vitro and inĀ vivo recordings of erectile responses in sGCƟ1ki/ki and wild-type mice after stimulation with sGC-dependent and -independent vasorelaxant agents. RESULTS: NO-induced responses were abolished in sGCƟ1ki/ki mice inĀ vitro and inĀ vivo. The ability of the heme-dependent, NO-independent sGC stimulator BAY 41-2272 to relax the corpora cavernosa was markedly attenuated in sGCƟ1ki/ki mice. In contrast, the relaxation response to the heme- and NO-independent sGC activator BAY 58-2667 was significantly enhanced in sGCƟ1ki/ki mice. The relaxing effect of sGC-independent vasorelaxant agents was similar in wild-type and sGCƟ1ki/ki mice, illustrating that the observed alterations in vasorelaxation are limited to NO-sGC-cGMP-mediated processes. CONCLUSION: Our results suggest that sGC is the sole target of NO in erectile physiology. Furthermore, thisĀ study provides indirect evidence that, in addition to sGCα1Ɵ1, sGCα2Ɵ1 is important for erectile function. In addition, the significant relaxation observed in sGCƟ1ki/ki mice with the cumulative addition of the sGCĀ activator BAY 58-2667 indicates that sGC activators might offer value in treating erectile dysfunction.


Subject(s)
Cyclic GMP/metabolism , Erectile Dysfunction/physiopathology , Heme/deficiency , Soluble Guanylyl Cyclase/metabolism , Animals , Disease Models, Animal , Guanylate Cyclase/metabolism , Humans , Male , Mice , Nitric Oxide/metabolism , Penile Erection/drug effects , Penis/physiopathology
8.
Proc Natl Acad Sci U S A ; 109(47): 19504-9, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23129650

ABSTRACT

Recent advances in metabolic engineering have demonstrated that microbial biosynthesis can provide a viable alternative to chemical synthesis for the production of bulk and fine chemicals. Introduction of a new biosynthetic pathway typically requires the expression of multiple heterologous enzymes in the production host, which can impose stress on the host cell and, thereby, limit performance of the pathway. Unfortunately, analysis and treatment of the host stress response can be difficult, because there are many sources of stress that may interact in complex ways. We use a systems biological approach to analyze the stress imposed by expressing different enzyme variants from a lineage of soluble P450 monooxygenases, previously evolved for heterologous activity in Saccharomyces cerevisiae. Our analysis identifies patterns of stress imposed on the host by heterologous enzyme overexpression that are consistent across the evolutionary lineage, ultimately implicating heme depletion as the major stress. We show that the monooxygenase evolution, starting from conditions of either high or low stress, caused the cellular stress to converge to a common level. Overexpression of rate-limiting enzymes in the endogenous heme biosynthetic pathway alleviates the stress imposed by expression of the P450 monooxygenases and increases the enzymatic activity of the final evolved P450 by an additional 2.3-fold. Heme overexpression also increases the total activity of an endogenous cytosolic heme-containing catalase but not a heterologous P450 that is membrane-associated. This work demonstrates the utility of combining systems and synthetic biology to analyze and optimize heterologous enzyme expression.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Evolution, Molecular , Heme/deficiency , Phylogeny , Saccharomyces cerevisiae/enzymology , Cytochrome P-450 Enzyme System/metabolism , Cytosol/metabolism , Gene Expression Regulation, Fungal , Heme/biosynthesis , Intracellular Space/metabolism , Metabolic Networks and Pathways , Oligonucleotide Array Sequence Analysis , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Stress, Physiological , Systems Biology , Theophylline/metabolism
9.
PLoS Pathog ; 8(7): e1002795, 2012.
Article in English | MEDLINE | ID: mdl-22807677

ABSTRACT

Trypanosomatid protozoan parasites lack a functional heme biosynthetic pathway, so must acquire heme from the environment to survive. However, the molecular pathway responsible for heme acquisition by these organisms is unknown. Here we show that L. amazonensis LHR1, a homolog of the C. elegans plasma membrane heme transporter HRG-4, functions in heme transport. Tagged LHR1 localized to the plasma membrane and to endocytic compartments, in both L. amazonensis and mammalian cells. Heme deprivation in L. amazonensis increased LHR1 transcript levels, promoted uptake of the fluorescent heme analog ZnMP, and increased the total intracellular heme content of promastigotes. Conversely, deletion of one LHR1 allele reduced ZnMP uptake and the intracellular heme pool by approximately 50%, indicating that LHR1 is a major heme importer in L. amazonensis. Viable parasites with correct replacement of both LHR1 alleles could not be obtained despite extensive attempts, suggesting that this gene is essential for the survival of promastigotes. Notably, LHR1 expression allowed Saccharomyces cerevisiae to import heme from the environment, and rescued growth of a strain deficient in heme biosynthesis. Syntenic genes with high sequence identity to LHR1 are present in the genomes of several species of Leishmania and also Trypanosoma cruzi and Trypanosoma brucei, indicating that therapeutic agents targeting this transporter could be effective against a broad group of trypanosomatid parasites that cause serious human disease.


Subject(s)
Heme/metabolism , Leishmania mexicana/metabolism , Membrane Transport Proteins/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , HeLa Cells , Heme/deficiency , Humans , Leishmania mexicana/pathogenicity , Macrophages/metabolism , Macrophages/parasitology , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Metalloporphyrins/metabolism , Mice , Molecular Sequence Data , Protein Structure, Tertiary , Protein Transport , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
10.
FEMS Yeast Res ; 14(2): 310-23, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24119181

ABSTRACT

Squalene is a valuable natural substance with several biotechnological applications. In the yeast Saccharomyces cerevisiae, it is produced in the isoprenoid pathway as the first precursor dedicated to ergosterol biosynthesis. The aim of this study was to explore the potential of squalene epoxidase encoded by the ERG1 gene as the target for manipulating squalene levels in yeast. Highest squalene levels (over 1000Ā Āµg squalene per 10(9) Ā cells) were induced by specific point mutations in ERG1 gene that reduced activity of squalene epoxidase and caused hypersensitivity to terbinafine. This accumulation of squalene in erg1 mutants did not significantly disturb their growth. Treatment with squalene epoxidase inhibitor terbinafine revealed a limit in squalene accumulation at 700Ā Āµg squalene per 10(9) Ā cells which was associated with pronounced growth defects. Inhibition of squalene epoxidase activity by anaerobiosis or heme deficiency resulted in relatively low squalene levels. These levels were significantly increased by ergosterol depletion in anaerobic cells which indicated feedback inhibition of squalene production by ergosterol. Accumulation of squalene in erg1 mutants and terbinafine-treated cells were associated with increased cellular content and aggregation of lipid droplets. Our results prove that targeted genetic manipulation of the ERG1 gene is a promising tool for increasing squalene production in yeast.


Subject(s)
Saccharomyces cerevisiae/metabolism , Squalene Monooxygenase/metabolism , Squalene/metabolism , Anaerobiosis , Antifungal Agents/pharmacology , Enzyme Activation/drug effects , Heme/deficiency , Mutation , Naphthalenes/pharmacology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Terbinafine
11.
Biochim Biophys Acta ; 1823(2): 484-92, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22101253

ABSTRACT

Ferredoxins are iron-sulfur proteins that have been studied for decades because of their role in facilitating the monooxygenase reactions catalyzed by p450 enzymes. More recently, studies in bacteria and yeast have demonstrated important roles for ferredoxin and ferredoxin reductase in iron-sulfur cluster assembly. The human genome contains two homologous ferredoxins, ferredoxin 1 (FDX1) and ferredoxin 2 (FDX2--formerly known as ferredoxin 1L). More recently, the roles of these two human ferredoxins in iron-sulfur cluster assembly were assessed, and it was concluded that FDX1 was important solely for its interaction with p450 enzymes to synthesize mitochondrial steroid precursors, whereas FDX2 was used for synthesis of iron-sulfur clusters, but not steroidogenesis. To further assess the role of the FDX-FDXR system in mammalian iron-sulfur cluster biogenesis, we performed siRNA studies on FDX1 and FDX2, on several human cell lines, using oligonucleotides identical to those previously used, along with new oligonucleotides that specifically targeted each gene. We concluded that both FDX1 and FDX2 were important in iron-sulfur cluster biogenesis. Loss of FDX1 activity disrupted activity of iron-sulfur cluster enzymes and cellular iron homeostasis, causing mitochondrial iron overload and cytosolic iron depletion. Moreover, knockdown of the sole human ferredoxin reductase, FDXR, diminished iron-sulfur cluster assembly and caused mitochondrial iron overload in conjunction with cytosolic depletion. Our studies suggest that interference with any of the three related genes, FDX1, FDX2 or FDXR, disrupts iron-sulfur cluster assembly and maintenance of normal cytosolic and mitochondrial iron homeostasis.


Subject(s)
Ferredoxins/metabolism , Iron-Sulfur Proteins/metabolism , Multigene Family , Oxidoreductases/metabolism , Amino Acid Sequence , Cell Line , Cytosol/enzymology , Electron Transport Complex I/metabolism , Ferredoxins/genetics , Gene Knockdown Techniques , Heme/deficiency , Humans , Iron/metabolism , Iron Regulatory Protein 1/metabolism , Iron Regulatory Protein 2/metabolism , Iron-Sulfur Proteins/genetics , Mitochondria/enzymology , Molecular Sequence Data , Oxidoreductases/genetics , RNA Interference , Sequence Alignment
12.
Proteomics ; 11(12): 2476-90, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21595038

ABSTRACT

Small-colony variants (SCVs) of Staphylococcus aureus represent a slow-growing subpopulation causing chronic and relapsing infections due to their physiological adaptation on an intracellular lifestyle. In this first proteomic study on physiological changes associated with a natural, clinically derived SCV, its proteomic profile was investigated in comparison to corresponding isogenic strains displaying normal (clinical wild-type strain, complemented hemB mutant and spontaneous revertant of the clinical SCV) and SCV phenotypes (hemB mutant and gentamicin-induced SCV). Applying an ultra-high resolution chromatography and high mass accuracy MS(E) -based label-free relative and absolute protein quantification approach, the whole cytoplasmic proteome of this strain sextet was investigated in a growth phase-controlled manner covering early-exponential, late-exponential and stationary phases. Of 1019 cytoplasmic proteins identified, 154 were found to be differently regulated between strains. All SCV phenotypes showed down-regulation of the tricarboxylic acid (TCA) cycle-related proteins and of a protein cluster involved in purine/pyrimidine and folate metabolism. In contrast to hemB mutant and gentamicin-induced SCVs, the clinically derived SCVs showed no prominent up-regulation of glycolytic proteins. The spontaneous switch into the normal phenotype resulted in up-regulation of TCA cycle-related parts, while oxidative stress-related proteins were down-regulated. However, the natural revertant from the clinical SCV retained also dominant protein features of the clinical SCV phenotype. In conclusion, physiological changes between normal and SCV S. aureus phenotypes are more complex than reflected by defined electron transport chain-interrupting mutants and their complemented counterparts.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Bacterial , Genetic Association Studies , Genome, Bacterial , Heme/deficiency , Staphylococcus aureus/genetics , Bacterial Proteins/metabolism , Chromatography , Citric Acid Cycle/drug effects , Colony Count, Microbial , Gene Deletion , Genotype , Gentamicins/pharmacology , Glycolysis/drug effects , Heme/genetics , Mass Spectrometry , Mutation/drug effects , Organisms, Genetically Modified/physiology , Phenotype , Polymerase Chain Reaction , Proteomics/methods , Staphylococcal Infections/microbiology , Staphylococcus aureus/metabolism
13.
Biochemistry ; 50(31): 6730-7, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21728357

ABSTRACT

Degradation of specific native proteins allows bacteria to rapidly adapt to changing environments when the activity of those proteins is no longer required. Although these processes are vital to bacterial survival, relatively little is known regarding how bacterial proteins are recognized and targeted for degradation. Staphylococcus aureus is an important human pathogen that requires iron for growth and pathogenesis. In the vertebrate host, S. aureus fulfills its iron requirement by obtaining heme iron from host hemoproteins via IsdG- and IsdI-mediated heme degradation. IsdG and IsdI are structurally and mechanistically analogous but are differentially regulated by iron and heme availability. Specifically, IsdG is targeted for degradation in the absence of heme. Therefore, we utilized the differential regulation of IsdG and IsdI to investigate the mechanism of regulated proteolysis. In contrast to canonical protease recognition sequences, we show that IsdG is targeted for degradation by internally coded sequences. Specifically, a flexible loop near the heme-binding pocket is required for IsdG degradation in the absence of heme.


Subject(s)
Heme/deficiency , Oxygenases/chemistry , Oxygenases/metabolism , Staphylococcus aureus/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Gene Targeting , Heme/genetics , Hemeproteins/metabolism , Humans , Iron/chemistry , Iron/metabolism , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Mutant Chimeric Proteins/chemistry , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Oxygenases/genetics , Point Mutation , Protein Conformation , Protein Denaturation , Protein Stability , Staphylococcus aureus/genetics
14.
Science ; 222(4627): 1031-3, 1983 Dec 02.
Article in English | MEDLINE | ID: mdl-6648517

ABSTRACT

Hepatic porphyrias are disorders of heme synthesis characterized by genetically determined lesions of one of the key enzymes of heme synthesis. In carriers of such lesions, several factors (drugs, environmental chemicals, or diet) precipitate acute and often fatal attacks of neurologic dysfunction, which are promptly relieved by intravenous infusion of heme. However, the mechanism of such heme-induced amelioration remains elusive. To probe this mechanism, the biochemical events triggered by acute hepatic heme deficiency were examined in an animal model of chemically induced porphyria. Acute hepatic heme depletion in porphyric rats was found to impair hepatic tryptophan pyrrolase activity which, in turn, elevated tryptophan and 5-hydroxytryptamine turnover in the brain. These alterations in porphyric rats were dramatically reversed by parenteral heme administration. These findings suggest that increased tryptophan and 5-hydroxytryptamine in the nervous system may be responsible for the neurologic dysfunctions observed in humans with acute attacks of hepatic porphyria.


Subject(s)
Brain/metabolism , Heme/deficiency , Liver Diseases/metabolism , Porphyrias/metabolism , Tryptophan/metabolism , Animals , Heme/pharmacology , Liver/enzymology , Liver Diseases/complications , Male , Nervous System Diseases/etiology , Porphyrias/complications , Rats , Rats, Inbred Strains , Serotonin/metabolism , Tryptophan Oxygenase/metabolism
15.
Mol Biol Cell ; 17(1): 90-103, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16251356

ABSTRACT

The mechanisms that govern intracellular transport of sterols in eukaryotic cells are not well understood. Saccharomyces cerevisiae is a facultative anaerobic organism that becomes auxotroph for sterols and unsaturated fatty acids in the absence of oxygen. To identify pathways that are required for uptake and transport of sterols, we performed a systematic screen of the yeast deletion mutant collection for genes that are required for growth under anaerobic conditions. Of the approximately 4800 nonessential genes represented in the deletion collection, 37 were essential for growth under anaerobic conditions. These affect a wide range of cellular functions, including biosynthetic pathways for certain amino acids and cofactors, reprogramming of transcription and translation, mitochondrial function and biogenesis, and membrane trafficking. Thirty-three of these mutants failed to grow on lipid-supplemented media when combined with a mutation in HEM1, which mimics anaerobic conditions in the presence of oxygen. Uptake assays with radio- and fluorescently labeled cholesterol revealed that 17 of the 33 mutants strongly affect uptake and/or esterification of exogenously supplied cholesterol. Examination of the subcellular distribution of sterols in these uptake mutants by cell fractionation and fluorescence microscopy indicates that some of the mutants block incorporation of cholesterol into the plasma membrane, a presumably early step in sterol uptake. Unexpectedly, the largest class of uptake mutants is affected in mitochondrial functions, and many of the uptake mutants show electron-dense mitochondrial inclusions. These results indicate that a hitherto uncharacterized mitochondrial function is required for sterol uptake and/or transport under anaerobic conditions and are discussed in light of the fact that mitochondrial import of cholesterol is required for steroidogenesis in vertebrate cells.


Subject(s)
Cholesterol/metabolism , Genome, Fungal/genetics , Mitochondria/genetics , Mitochondria/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Anaerobiosis , Cell Proliferation , Heme/deficiency , Heme/metabolism , Microscopy, Electron , Mutation/genetics , Oxygen/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
16.
J Neurosci ; 27(32): 8475-85, 2007 Aug 08.
Article in English | MEDLINE | ID: mdl-17687025

ABSTRACT

The early stages of many neurodegenerative diseases and age-related degeneration are characterized by neurite damage and compromised synaptic function that precede neuronal cell death. We investigated the signaling mechanisms underlying neurite degeneration using cortical neuron cultures. Inhibition of heme synthesis caused neurite damage, without neuronal death, and was mediated by reduced NMDA receptor (NMDAR) expression and phosphorylation. The signaling toward the degenerative phenotype involved suppression of the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway, and electrophysiological recording showed that the neurodegeneration is accompanied by reduced NMDAR current and Ca2+ influx, as well as reduced voltage-gated sodium currents, consistent with compromised neurite integrity. Rescue from the degenerative phenotype by heme replacement was dependent on restoration of NR2B subunit phosphorylation and expression of NMDAR currents with higher Ca2+ permeability, consistent with triggering prosurvival ERK1/2 signaling to maintain and extend neurites. This study demonstrated a new mechanism of neurodegeneration in which impaired heme synthesis led to NMDAR signaling dysfunction, suppression of the prosurvival ERK1/2 pathway, and progressive fragmentation of neuronal projections.


Subject(s)
Heme/deficiency , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Nerve Degeneration/enzymology , Neurites/metabolism , Neurites/pathology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Cell Survival/physiology , Cells, Cultured , Enzyme Activation/physiology , Heme/physiology , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Degeneration/pathology , Neurites/enzymology
17.
J Clin Invest ; 115(6): 1562-70, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15931390

ABSTRACT

Heme-regulated eIF2alpha kinase (HRI) controls protein synthesis by phosphorylating the alpha-subunit of eukaryotic translational initiation factor 2 (eIF2alpha). In heme deficiency, HRI is essential for translational regulation of alpha- and beta-globins and for the survival of erythroid progenitors. HRI is also activated by a number of cytoplasmic stresses other than heme deficiency, including oxidative stress and heat shock. However, to date, HRI has not been implicated in the pathogenesis of any known human disease or mouse phenotype. Here we report the essential role of HRI in 2 mouse models of human rbc disorders, namely erythropoietic protoporphyria (EPP) and beta-thalassemia. In both cases, lack of HRI adversely modifies the phenotype: HRI deficiency exacerbates EPP and renders beta-thalassemia embryonically lethal. This study establishes the protective function of HRI in inherited rbc diseases in mice and suggests that HRI may be a significant modifier of many rbc disorders in humans. Our findings also demonstrate that translational regulation could play a critical role in the clinical manifestation of rbc diseases.


Subject(s)
Globins/biosynthesis , Heme/metabolism , Protoporphyria, Erythropoietic/metabolism , beta-Thalassemia/metabolism , eIF-2 Kinase/metabolism , Animals , Disease Models, Animal , Erythrocytes/metabolism , Erythrocytes/pathology , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Globins/genetics , Heat Stress Disorders , Heme/deficiency , Humans , Mice , Mice, Knockout , Oxidative Stress , Phenotype , Protein Biosynthesis/genetics , Protoporphyria, Erythropoietic/genetics , Protoporphyria, Erythropoietic/pathology , beta-Thalassemia/genetics , beta-Thalassemia/pathology , eIF-2 Kinase/genetics
18.
J Clin Invest ; 76(6): 2144-50, 1985 Dec.
Article in English | MEDLINE | ID: mdl-3001142

ABSTRACT

These studies were performed to determine whether the reticulocyte can synthesize its own transferrin receptor and, if so, whether synthesis is subject to translational control by intracellular heme. Reticulocytosis (20-35%) was produced by bleeding rabbits and the washed cells were incubated for 1-4 h at 37 degrees C in buffered nutritional medium containing L-[35S]methionine. After washing and detergent lysis in the presence of protease inhibitors, supernatant reticulocyte extracts were analyzed for transferrin receptors by immunoprecipitation with specific ovine receptor antibody raised against denatured rabbit transferrin receptor. Immunoprecipitates were analyzed by SDS-gel electrophoresis and fluorography. Antibody, but not preimmune sheep immunoglobin, consistently precipitated a 35S-labeled protein with an Mr of 90,000 (reduced), coincident with bona fide receptor subunits purified by ligand-affinity chromatography. Incorporation of radioactive methionine was exclusively associated with receptor in reticulocyte stroma, and nascent receptor was not detected on free polyribosomes. Incorporation of radioactivity in the receptor moiety accounted for 0.1-0.2% of total incorporation into TCA insoluble cell protein. Treatment of the cells with 40 micrograms/ml cycloheximide markedly inhibited amino acid incorporation into the receptor, thus indicating de novo synthesis of receptor protein. On treatment of reticulocytes with 4,6 dioxoheptanoate to induce heme deficiency by diminishing the formation of intracellular heme, synthesis of the receptor was inhibited by greater than 50%; synthesis was restored to control rates on addition of 50 microM exogenous hemin. These findings indicate that the reticulocyte retains receptor mRNA and that synthesis of the receptor in erythroid cells is subject to translational regulation by intracellular heme.


Subject(s)
Heme/physiology , Receptors, Cell Surface/biosynthesis , Reticulocytes/metabolism , Transferrin/metabolism , Animals , Heme/deficiency , Hemin/pharmacology , Molecular Weight , Protein Biosynthesis , Rabbits , Receptors, Transferrin
19.
J Pharmacol Exp Ther ; 323(3): 979-89, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17761498

ABSTRACT

Tryptophan 2,3-dioxygenase (TDO), a liver-specific cytosolic hemoprotein, is the rate-limiting enzyme in L-tryptophan catabolism and thus a key serotonergic determinant. Glucocorticoids transcriptionally activate the TDO gene with marked enzyme induction. TDO is also regulated by heme, its prosthetic moiety, as its expression and function are significantly reduced after acute hepatic heme depletion. Here we show in primary rat hepatocytes that this impairment is not due to faulty transcriptional activation of the TDO gene but rather due to its posttranscriptional regulation by heme. Accordingly, in acutely heme-depleted hepatocytes, the de novo synthesis of TDO protein is markedly decreased (>90%) along with that of other hepatic proteins. This global suppression of de novo hepatic protein syntheses in these heme-depleted cells is associated with a significantly enhanced phosphorylation of the alpha-subunit of the eukaryotic initiation factor eIF2 (eIF2alpha), as monitored by the phosphorylated eIF2alpha/total eIF2alpha ratio. Heme supplementation reversed these effects, indicating that heme regulates TDO induction by functional control of an eIF2alpha kinase. A cDNA was cloned from heme-depleted rat hepatocytes, and DNA sequencing verified its identity to the previously cloned rat brain heme-regulated inhibitor (HRI). Proteomic, biochemical, and/or immunoblotting analyses of the purified recombinant protein and the immunoaffinity-captured hepatic protein confirmed its identity as a rat heme-sensitive eIF2alpha kinase. These findings not only document that a hepatic HRI exists and is physiologically relevant but also implicate its translational shut-off of key proteins in the pathogenesis and symptomatology of the acute hepatic heme-deficient conditions clinically known as the hepatic porphyrias.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Heme/deficiency , Hepatocytes/drug effects , Tryptophan Oxygenase , eIF-2 Kinase/physiology , Animals , Cells, Cultured , Chromatography, Liquid , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Enzyme Induction , Escherichia coli/genetics , Heme/metabolism , Hepatocytes/enzymology , Male , Protein Biosynthesis , Protoporphyrins/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Tandem Mass Spectrometry , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan Oxygenase/biosynthesis , eIF-2 Kinase/genetics
20.
Mitochondrion ; 7(5): 297-310, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17625988

ABSTRACT

Several studies have demonstrated aberrations in the Electron Transport Complexes (ETC) and Krebs (TCA) cycle in Alzheimer's disease (AD) brain. Optimal activity of these key metabolic pathways depends on several redox active centers and metabolites including heme, coenzyme Q, iron-sulfur, vitamins, minerals, and micronutrients. Disturbed heme metabolism leads to increased aberrations in the ETC (loss of complex IV), dimerization of APP, free radical production, markers of oxidative damage, and ultimately cell death all of which represent key cytopathologies in AD. The mechanism of mitochondrial dysfunction in AD is controversial. The observations that Abeta is found both in the cells and in the mitochondria and that Abeta binds with heme may provide clues to this mechanism. Mitochondrial Abeta may interfere with key metabolites or metabolic pathways in a manner that overwhelms the mitochondrial mechanisms of repair. Identifying the molecular mechanism for how Abeta interferes with mitochondria and that explains the established key cytopathologies in AD may also suggest molecular targets for therapeutic interventions. Below we review recent studies describing the possible role of Abeta in altered energy production through heme metabolism. We further discuss how protecting mitochondria could confer resistance to oxidative and environmental insults. Therapies targeted at protecting mitochondria may improve the clinical outcome of AD patients.


Subject(s)
Alzheimer Disease/physiopathology , Energy Metabolism , Mitochondria/physiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Apolipoproteins E/metabolism , Brain/physiopathology , Cells, Cultured , Citric Acid Cycle/physiology , Electron Transport Chain Complex Proteins/metabolism , Free Radicals/metabolism , Heme/deficiency , Heme/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL