Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.887
Filter
1.
Gut Microbes ; 16(1): 2359665, 2024.
Article in English | MEDLINE | ID: mdl-38831611

ABSTRACT

The facultative anaerobic Gram-positive bacterium Enterococcus faecium is a ubiquitous member of the human gut microbiota. However, it has gradually evolved into a pathogenic and multidrug resistant lineage that causes nosocomial infections. The establishment of high-level intestinal colonization by enterococci represents a critical step of infection. The majority of current research on Enterococcus has been conducted under aerobic conditions, while limited attention has been given to its physiological characteristics in anaerobic environments, which reflects its natural colonization niche in the gut. In this study, a high-density transposon mutant library containing 26,620 distinct insertion sites was constructed. Tn-seq analysis identified six genes that significantly contribute to growth under anaerobic conditions. Under anaerobic conditions, deletion of sufB (encoding Fe-S cluster assembly protein B) results in more extensive and significant impairments on carbohydrate metabolism compared to aerobic conditions. Consistently, the pathways involved in this utilization-restricted carbohydrates were mostly expressed at significantly lower levels in mutant compared to wild-type under anaerobic conditions. Moreover, deletion of sufB or pflA (encoding pyruvate formate lyase-activating protein A) led to failure of gastrointestinal colonization in mice. These findings contribute to our understanding of the mechanisms by which E. faecium maintains proliferation under anaerobic conditions and establishes colonization in the gut.


Subject(s)
Bacterial Proteins , Enterococcus faecium , Iron-Sulfur Proteins , Enterococcus faecium/genetics , Enterococcus faecium/metabolism , Enterococcus faecium/growth & development , Animals , Mice , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Anaerobiosis , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Gastrointestinal Tract/microbiology , Gastrointestinal Microbiome , Gram-Positive Bacterial Infections/microbiology , Humans , DNA Transposable Elements , Carbohydrate Metabolism , Female , Acetyltransferases
2.
Adv Microb Physiol ; 84: 243-307, 2024.
Article in English | MEDLINE | ID: mdl-38821633

ABSTRACT

Organelles are membrane bound structures that compartmentalize biochemical and molecular functions. With improved molecular, biochemical and microscopy tools the diversity and function of protistan organelles has increased in recent years, providing a complex panoply of structure/function relationships. This is particularly noticeable with the description of hydrogenosomes, and the diverse array of structures that followed, having hybrid hydrogenosome/mitochondria attributes. These diverse organelles have lost the major, at one time, definitive components of the mitochondrion (tricarboxylic cycle enzymes and cytochromes), however they all contain the machinery for the assembly of Fe-S clusters, which is the single unifying feature they share. The plasticity of organelles, like the mitochondrion, is therefore evident from its ability to lose its identity as an aerobic energy generating powerhouse while retaining key ancestral functions common to both aerobes and anaerobes. It is interesting to note that the apicoplast, a non-photosynthetic plastid that is present in all apicomplexan protozoa, apart from Cryptosporidium and possibly the gregarines, is also the site of Fe-S cluster assembly proteins. It turns out that in Cryptosporidium proteins involved in Fe-S cluster biosynthesis are localized in the mitochondrial remnant organelle termed the mitosome. Hence, different organisms have solved the same problem of packaging a life-requiring set of reactions in different ways, using different ancestral organelles, discarding what is not needed and keeping what is essential. Don't judge an organelle by its cover, more by the things it does, and always be prepared for surprises.


Subject(s)
Organelles , Organelles/metabolism , Mitochondria/metabolism , Eukaryota/metabolism , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics
3.
Nat Commun ; 15(1): 4655, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38821922

ABSTRACT

The human AAA-ATPase Bcs1L translocates the fully assembled Rieske iron-sulfur protein (ISP) precursor across the mitochondrial inner membrane, enabling respiratory Complex III assembly. Exactly how the folded substrate is bound to and released from Bcs1L has been unclear, and there has been ongoing debate as to whether subunits of Bcs1L act in sequence or in unison hydrolyzing ATP when moving the protein cargo. Here, we captured Bcs1L conformations by cryo-EM during active ATP hydrolysis in the presence or absence of ISP substrate. In contrast to the threading mechanism widely employed by AAA proteins in substrate translocation, subunits of Bcs1L alternate uniformly between ATP and ADP conformations without detectable intermediates that have different, co-existing nucleotide states, indicating that the subunits act in concert. We further show that the ISP can be trapped by Bcs1 when its subunits are all in the ADP-bound state, which we propose to be released in the apo form.


Subject(s)
ATPases Associated with Diverse Cellular Activities , Adenosine Diphosphate , Adenosine Triphosphate , Cryoelectron Microscopy , Electron Transport Complex III , Adenosine Triphosphate/metabolism , Hydrolysis , Electron Transport Complex III/metabolism , Electron Transport Complex III/chemistry , Humans , Adenosine Diphosphate/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , ATPases Associated with Diverse Cellular Activities/chemistry , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/chemistry , Protein Conformation , Protein Folding , Models, Molecular , Protein Transport
4.
Genes (Basel) ; 15(5)2024 05 07.
Article in English | MEDLINE | ID: mdl-38790219

ABSTRACT

Pa0665 in Pseudomonas aeruginosa shares homologous sequences with that of the essential A-type iron-sulfur (Fe-S) cluster insertion protein ErpA in Escherichia coli. However, its essentiality in P. aeruginosa and its complementation with E. coli erpA has not been experimentally examined. To fulfill this task, we constructed plasmid-based ts-mutant Δpa0665/pTS-pa0665 using a three-step protocol. The mutant displayed growth defects at 42 °C, which were complemented by expressing ec.erpA. Microscopic observations indicated a petite cell phenotype for Δpa0665/pTS-pa0665 at 42 °C, correlated with the downregulation of the oprG gene. RNA sequencing revealed significant transcriptional changes in genes associated with the oxidative phosphorylation (OXPHOS) system, aligning with reduced ATP levels in Δpa0665/pTS-pa0665 under 42 °C. Additionally, the ts-mutant showed heightened sensitivity to H2O2 at 42 °C. Overall, our study demonstrates the essential role of pa0665 for OXPHOS function and is complemented by ec.erpA. We propose that the plasmid-based ts-allele is useful for genetic analysis of essential genes of interest in P. aeruginosa.


Subject(s)
Bacterial Proteins , Oxidative Phosphorylation , Pseudomonas aeruginosa , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Mutation , Escherichia coli/genetics , Escherichia coli/metabolism , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Gene Expression Regulation, Bacterial
5.
J Am Chem Soc ; 146(23): 15771-15778, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38819401

ABSTRACT

The active site cofactor of [FeFe]-hydrogenases consists of a cubane [4Fe-4S]-cluster and a unique [2Fe-2S]-cluster, harboring unusual CO- and CN--ligands. The biosynthesis of the [2Fe-2S]-cluster requires three dedicated maturation enzymes called HydG, HydE and HydF. HydG and HydE are both involved in synthesizing a [2Fe-2S]-precursor, still lacking parts of the azadithiolate (adt) moiety that bridge the two iron atoms. This [2Fe-2S]-precursor is then finalized within the scaffold protein HydF, which binds and transfers the [2Fe-2S]-precursor to the hydrogenase. However, its exact binding mode within HydF is still elusive. Herein, we identified the binding location of the [2Fe-2S]-precursor by altering size and charge of a highly conserved protein pocket via site directed mutagenesis (SDM). Moreover, we identified two serine residues that are essential for binding and assembling the [2Fe-2S]-precursor. By combining SDM and molecular docking simulations, we provide a new model on how the [2Fe-2S]-cluster is bound to HydF and demonstrate the important role of one highly conserved aspartate residue, presumably during the bioassembly of the adt moiety.


Subject(s)
Hydrogenase , Iron-Sulfur Proteins , Hydrogenase/chemistry , Hydrogenase/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/metabolism , Binding Sites , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Iron/chemistry , Iron/metabolism , Models, Molecular
6.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119746, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38719030

ABSTRACT

Iron­sulfur (Fe-S) clusters are one of the most ancient and versatile inorganic cofactors present in the three domains of life. Fe-S clusters are essential cofactors for the activity of a large variety of metalloproteins that play crucial physiological roles. Fe-S protein biogenesis is a complex process that starts with the acquisition of the elements (iron and sulfur atoms) and their assembly into an Fe-S cluster that is subsequently inserted into the target proteins. The Fe-S protein biogenesis is ensured by multiproteic systems conserved across all domains of life. Here, we provide an overview on how bacterial genetics approaches have permitted to reveal and dissect the Fe-S protein biogenesis process in vivo.


Subject(s)
Bacterial Proteins , Iron-Sulfur Proteins , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Iron/metabolism , Sulfur/metabolism , Bacteria/genetics , Bacteria/metabolism
7.
J Mol Cell Cardiol ; 192: 36-47, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734062

ABSTRACT

AIMS: Ferroptosis is a form of iron-regulated cell death implicated in ischemic heart disease. Our previous study revealed that Sirtuin 3 (SIRT3) is associated with ferroptosis and cardiac fibrosis. In this study, we tested whether the knockout of SIRT3 in cardiomyocytes (SIRT3cKO) promotes mitochondrial ferroptosis and whether the blockade of ferroptosis would ameliorate mitochondrial dysfunction. METHODS AND RESULTS: Mitochondrial and cytosolic fractions were isolated from the ventricles of mice. Cytosolic and mitochondrial ferroptosis were analyzed by comparison to SIRT3loxp mice. An echocardiography study showed that SIRT3cKO mice developed heart failure as evidenced by a reduction of EF% and FS% compared to SIRT3loxp mice. Comparison of mitochondrial and cytosolic fractions of SIRT3cKO and SIRT3loxp mice revealed that, upon loss of SIRT3, mitochondrial, but not cytosolic, total lysine acetylation was significantly increased. Similarly, acetylated p53 was significantly upregulated only in the mitochondria. These data demonstrate that SIRT3 is the primary mitochondrial deacetylase. Most importantly, loss of SIRT3 resulted in significant reductions of frataxin, aconitase, and glutathione peroxidase 4 (GPX4) in the mitochondria. This was accompanied by a significant increase in levels of mitochondrial 4-hydroxynonenal. Treatment of SIRT3cKO mice with the ferroptosis inhibitor ferrostatin-1 (Fer-1) for 14 days significantly improved preexisting heart failure. Mechanistically, Fer-1 treatment significantly increased GPX4 and aconitase expression/activity, increased mitochondrial iron­sulfur clusters, and improved mitochondrial membrane potential and Complex IV activity. CONCLUSIONS: Inhibition of ferroptosis ameliorated cardiac dysfunction by specifically targeting mitochondrial aconitase and iron­sulfur clusters. Blockade of mitochondrial ferroptosis may be a novel therapeutic target for mitochondrial cardiomyopathies.


Subject(s)
Aconitate Hydratase , Ferroptosis , Mice, Knockout , Myocytes, Cardiac , Phenylenediamines , Sirtuin 3 , Animals , Sirtuin 3/metabolism , Sirtuin 3/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Aconitate Hydratase/metabolism , Ferroptosis/drug effects , Mice , Acetylation , Phenylenediamines/pharmacology , Mitochondria/metabolism , Mitochondria/drug effects , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Iron/metabolism , Frataxin , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Iron-Binding Proteins/metabolism , Iron-Binding Proteins/genetics , Heart Failure/metabolism , Heart Failure/genetics , Cytosol/metabolism , Cyclohexylamines
8.
Biochim Biophys Acta Mol Cell Res ; 1871(6): 119750, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38762171

ABSTRACT

Azotobacter vinelandii is a genetically tractable Gram-negative proteobacterium able to fix nitrogen (N2) under aerobic growth conditions. This narrative describes how biochemical-genetic approaches using A. vinelandii to study nitrogen fixation led to the formulation of the "scaffold hypothesis" for the assembly of both simple and complex [Fe-S] clusters associated with biological nitrogen fixation. These studies also led to the discovery of a parallel, but genetically distinct, pathway for maturation of [Fe-S] proteins that support central metabolic processes.


Subject(s)
Azotobacter vinelandii , Bacterial Proteins , Iron-Sulfur Proteins , Nitrogen Fixation , Azotobacter vinelandii/metabolism , Azotobacter vinelandii/genetics , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics
9.
Biochim Biophys Acta Mol Cell Res ; 1871(6): 119749, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38763301

ABSTRACT

The role of IscR in regulating the transcription of genes involved in Fe-S cluster homeostasis has been well established for the model organism Escherichia coli K12. In this bacterium, IscR coordinates expression of the Isc and Suf Fe-S cluster assembly pathways to meet cellular Fe-S cluster demands shaped by a variety of environmental cues. However, since its initial discovery nearly 25 years ago, there has been growing evidence that IscR function extends well beyond Fe-S cluster homeostasis, not only in E. coli, but in bacteria of diverse lifestyles. Notably, pathogenic bacteria have exploited the ability of IscR to respond to changes in oxygen tension, oxidative and nitrosative stress, and iron availability to navigate their trajectory in their respective hosts as changes in these cues are frequently encountered during host infection. In this review, we highlight these broader roles of IscR in different cellular processes and, in particular, discuss the importance of IscR as a virulence factor for many bacterial pathogens.


Subject(s)
Escherichia coli Proteins , Homeostasis , Iron-Sulfur Proteins , Iron , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Iron/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Transcription Factors/metabolism , Transcription Factors/genetics , Humans , Escherichia coli/metabolism , Escherichia coli/genetics
10.
Metallomics ; 16(5)2024 05 02.
Article in English | MEDLINE | ID: mdl-38744662

ABSTRACT

Iron-sulfur (Fe-S) clusters are an essential and ubiquitous class of protein-bound prosthetic centers that are involved in a broad range of biological processes (e.g. respiration, photosynthesis, DNA replication and repair and gene regulation) performing a wide range of functions including electron transfer, enzyme catalysis, and sensing. In a general manner, Fe-S clusters can gain or lose electrons through redox reactions, and are highly sensitive to oxidation, notably by small molecules such as oxygen and nitric oxide. The [2Fe-2S] and [4Fe-4S] clusters, the most common Fe-S cofactors, are typically coordinated by four amino acid side chains from the protein, usually cysteine thiolates, but other residues (e.g. histidine, aspartic acid) can also be found. While diversity in cluster coordination ensures the functional variety of the Fe-S clusters, the lack of conserved motifs makes new Fe-S protein identification challenging especially when the Fe-S cluster is also shared between two proteins as observed in several dimeric transcriptional regulators and in the mitoribosome. Thanks to the recent development of in cellulo, in vitro, and in silico approaches, new Fe-S proteins are still regularly identified, highlighting the functional diversity of this class of proteins. In this review, we will present three main functions of the Fe-S clusters and explain the difficulties encountered to identify Fe-S proteins and methods that have been employed to overcome these issues.


Subject(s)
Iron-Sulfur Proteins , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Oxidation-Reduction
11.
Biochem Biophys Res Commun ; 722: 150167, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-38797154

ABSTRACT

Iron-sulfur (Fe-S) clusters are ubiquitous and are necessary to sustain basic life processes. The intracellular Fe-S clusters do not form spontaneously and many proteins are required for their biosynthesis and delivery. The bacterial P-loop NTPase family protein ApbC participates in Fe-S cluster assembly and transfers the cluster into apoproteins, with the Walker A motif and CxxC motif being essential for functionality of ApbC in Fe-S protein biogenesis. However, the structural basis underlying the ApbC activity and the motifs' role remains unclear. Here, we report the crystal structure of Escherichia coli ApbC at 2.8 Å resolution. The dimeric structure is in a W shape and the active site is located in the 2-fold center. The function of the motifs can be annotated by structural analyses. ApbC has an additional N-terminal domain that differs from other P-loop NTPases, possibly conferring its inherent specificity in vivo.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Iron-Sulfur Proteins , Models, Molecular , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli/metabolism , Escherichia coli/genetics , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Crystallography, X-Ray , Amino Acid Sequence , Protein Conformation , Catalytic Domain , Protein Multimerization
12.
Biochemistry ; 63(12): 1569-1577, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38813769

ABSTRACT

The Escherichia coli cysteine desulfurase SufS (EcSufS) is a dimeric, PLP-dependent enzyme responsible for sulfur mobilization in the SUF Fe-S cluster bioassembly pathway. The enzyme uses cysteine as a sulfur source and generates alanine and a covalent persulfide located on an active site of cysteine. Optimal in vitro activity of EcSufS requires the presence of the transpersulfurase protein, EcSufE, and a strong reductant. Here, presteady-state and single-turnover kinetics are used to investigate the mechanism of EcSufS activation by EcSufE. In the absence of EcSufE, EcSufS exhibits a presteady-state burst of product production with an amplitude of ∼0.4 active site equivalents, consistent with a half-sites reactivity. KinTek Explorer was used to isolate the first turnover of alanine formation and fit the data with a simplified kinetic mechanism with steps for alanine formation (k3) and a net rate constant for the downstream steps (k5). Using this treatment, microscopic rate constants of 2.3 ± 0.5 s-1 and 0.10 ± 0.01 s-1 were determined for k3 and k5, respectively. The inclusion of EcSufE in the reaction results in a similar rate constant for k3 but induces a 10-fold enhancement of k5 to 1.1 ± 0.2 s-1, such that both steps are partially rate-determining. The most likely downstream step where EcSufE could exert influence on EcSufS activity is the removal of the persulfide intermediate. Importantly, this step appears to serve as a limiting feature in the half-sites activity such that activating persulfide transfer allows for rapid shifting between active sites. Single-turnover assays show that the presence of EcSufE slightly slowed the rates of alanine-forming steps, suggesting it does not activate steps in the desulfurase half reaction.


Subject(s)
Carbon-Sulfur Lyases , Escherichia coli Proteins , Escherichia coli , Sulfides , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Sulfides/metabolism , Sulfides/chemistry , Escherichia coli/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Kinetics , Carbon-Sulfur Lyases/metabolism , Carbon-Sulfur Lyases/chemistry , Alanine/metabolism , Alanine/chemistry , Catalytic Domain , Cysteine/metabolism , Cysteine/chemistry , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/chemistry
13.
Biochemistry ; 63(12): 1588-1598, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38817151

ABSTRACT

Thioredoxin reductases (TrxR) activate thioredoxins (Trx) that regulate the activity of diverse target proteins essential to prokaryotic and eukaryotic life. However, very little is understood of TrxR/Trx systems and redox control in methanogenic microbes from the domain Archaea (methanogens), for which genomes are abundant with annotations for ferredoxin:thioredoxin reductases [Fdx/thioredoxin reductase (FTR)] from group 4 of the widespread FTR-like family. Only two from the FTR-like family are characterized: the plant-type FTR from group 1 and FDR from group 6. Herein, the group 4 archetype (AFTR) from Methanosarcina acetivorans was characterized to advance understanding of the family and TrxR/Trx systems in methanogens. The modeled structure of AFTR, together with EPR and Mössbauer spectroscopies, supports a catalytic mechanism similar to plant-type FTR and FDR, albeit with important exceptions. EPR spectroscopy of reduced AFTR identified a transient [4Fe-4S]1+ cluster exhibiting a mixture of S = 7/2 and typical S = 1/2 signals, although rare for proteins containing [4Fe-4S] clusters, it is most likely the on-pathway intermediate in the disulfide reduction. Furthermore, an active site histidine equivalent to residues essential for the activity of plant-type FTR and FDR was found dispensable for AFTR. Finally, a unique thioredoxin system was reconstituted from AFTR, ferredoxin, and Trx2 from M. acetivorans, for which specialized target proteins were identified that are essential for growth and other diverse metabolisms.


Subject(s)
Iron-Sulfur Proteins , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Methanosarcina/enzymology , Methanosarcina/genetics , Ferredoxins/metabolism , Ferredoxins/chemistry , Ferredoxins/genetics , Oxidation-Reduction , Models, Molecular , Thioredoxins/metabolism , Thioredoxins/chemistry , Thioredoxins/genetics , Oxidoreductases/metabolism , Oxidoreductases/chemistry , Oxidoreductases/genetics , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxin-Disulfide Reductase/chemistry , Thioredoxin-Disulfide Reductase/genetics , Archaeal Proteins/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Electron Spin Resonance Spectroscopy
14.
J Invertebr Pathol ; 205: 108141, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38788920

ABSTRACT

Electron-transferring flavoprotein (Etf) and its dehydrogenase (Etfdh) are integral components of the electron transport chain in mitochondria. In this study, we characterize two putative etf genes (Bbetfa and Bbetfb) and their dehydrogenase gene Bbetfdh in the entomopathogenic fungus Beauveria bassiana. Individual deletion of these genes caused a significant reduction in vegetative growth, conidiation, and delayed conidial germination. Lack of these genes also led to abnormal metabolism of fatty acid and increasing lipid body accumulation. Furthermore, the virulence of Bbetfs and Bbetfdh deletion mutants was severely impaired due to decreasing infection structure formation. Additionally, all deletion strains showed reduced ATP synthesis compared to the wild-type strain. Taken together, Bbetfa and Bbetfb, along with Bbetfdh, play principal roles in fungal vegetative growth, conidiation, conidial germination, and pathogenicity of B. bassiana due to their essential functions in fatty acid metabolism.


Subject(s)
Beauveria , Electron-Transferring Flavoproteins , Beauveria/pathogenicity , Beauveria/genetics , Beauveria/enzymology , Electron-Transferring Flavoproteins/genetics , Electron-Transferring Flavoproteins/metabolism , Virulence , Fungal Proteins/genetics , Fungal Proteins/metabolism , Spores, Fungal/growth & development , Oxidoreductases/metabolism , Oxidoreductases/genetics , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Oxidoreductases Acting on CH-NH Group Donors
15.
Proc Natl Acad Sci U S A ; 121(22): e2405123121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38781208

ABSTRACT

Mitochondria play a central role in muscle metabolism and function. A unique family of iron-sulfur proteins, termed CDGSH Iron Sulfur Domain-containing (CISD/NEET) proteins, support mitochondrial function in skeletal muscles. The abundance of these proteins declines during aging leading to muscle degeneration. Although the function of the outer mitochondrial CISD/NEET proteins, CISD1/mitoNEET and CISD2/NAF-1, has been defined in skeletal muscle cells, the role of the inner mitochondrial CISD protein, CISD3/MiNT, is currently unknown. Here, we show that CISD3 deficiency in mice results in muscle atrophy that shares proteomic features with Duchenne muscular dystrophy. We further reveal that CISD3 deficiency impairs the function and structure of skeletal muscles, as well as their mitochondria, and that CISD3 interacts with, and donates its [2Fe-2S] clusters to, complex I respiratory chain subunit NADH Ubiquinone Oxidoreductase Core Subunit V2 (NDUFV2). Using coevolutionary and structural computational tools, we model a CISD3-NDUFV2 complex with proximal coevolving residue interactions conducive of [2Fe-2S] cluster transfer reactions, placing the clusters of the two proteins 10 to 16 Å apart. Taken together, our findings reveal that CISD3/MiNT is important for supporting the biogenesis and function of complex I, essential for muscle maintenance and function. Interventions that target CISD3 could therefore impact different muscle degeneration syndromes, aging, and related conditions.


Subject(s)
Electron Transport Complex I , Mitochondrial Proteins , Muscle, Skeletal , Animals , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mice , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondria/metabolism , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Mice, Knockout , Mitochondria, Muscle/metabolism , Humans , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Atrophy/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/genetics
16.
Nat Commun ; 15(1): 4244, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762605

ABSTRACT

Cysteine metabolism occurs across cellular compartments to support diverse biological functions and prevent the induction of ferroptosis. Though the disruption of cytosolic cysteine metabolism is implicated in this form of cell death, it is unknown whether the substantial cysteine metabolism resident within the mitochondria is similarly pertinent to ferroptosis. Here, we show that despite the rapid depletion of intracellular cysteine upon loss of extracellular cystine, cysteine-dependent synthesis of Fe-S clusters persists in the mitochondria of lung cancer cells. This promotes a retention of respiratory function and a maintenance of the mitochondrial redox state. Under these limiting conditions, we find that glutathione catabolism by CHAC1 supports the mitochondrial cysteine pool to sustain the function of the Fe-S proteins critical to oxidative metabolism. We find that disrupting Fe-S cluster synthesis under cysteine restriction protects against the induction of ferroptosis, suggesting that the preservation of mitochondrial function is antagonistic to survival under starved conditions. Overall, our findings implicate mitochondrial cysteine metabolism in the induction of ferroptosis and reveal a mechanism of mitochondrial resilience in response to nutrient stress.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cysteine , Ferroptosis , Glutathione , Lung Neoplasms , Mitochondria , Humans , Cysteine/metabolism , Mitochondria/metabolism , Glutathione/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Cell Line, Tumor , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Iron-Sulfur Proteins/metabolism , Oxidation-Reduction , Mice
17.
Proc Natl Acad Sci U S A ; 121(21): e2400740121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38743629

ABSTRACT

The biogenesis of iron-sulfur (Fe/S) proteins entails the synthesis and trafficking of Fe/S clusters, followed by their insertion into target apoproteins. In eukaryotes, the multiple steps of biogenesis are accomplished by complex protein machineries in both mitochondria and cytosol. The underlying biochemical pathways have been elucidated over the past decades, yet the mechanisms of cytosolic [2Fe-2S] protein assembly have remained ill-defined. Similarly, the precise site of glutathione (GSH) requirement in cytosolic and nuclear Fe/S protein biogenesis is unclear, as is the molecular role of the GSH-dependent cytosolic monothiol glutaredoxins (cGrxs). Here, we investigated these questions in human and yeast cells by various in vivo approaches. [2Fe-2S] cluster assembly of cytosolic target apoproteins required the mitochondrial ISC machinery, the mitochondrial transporter Atm1/ABCB7 and GSH, yet occurred independently of both the CIA system and cGrxs. This mechanism was strikingly different from the ISC-, Atm1/ABCB7-, GSH-, and CIA-dependent assembly of cytosolic-nuclear [4Fe-4S] proteins. One notable exception to this cytosolic [2Fe-2S] protein maturation pathway defined here was yeast Apd1 which used the CIA system via binding to the CIA targeting complex through its C-terminal tryptophan. cGrxs, although attributed as [2Fe-2S] cluster chaperones or trafficking proteins, were not essential in vivo for delivering [2Fe-2S] clusters to either CIA components or target apoproteins. Finally, the most critical GSH requirement was assigned to Atm1-dependent export, i.e. a step before GSH-dependent cGrxs function. Our findings extend the general model of eukaryotic Fe/S protein biogenesis by adding the molecular requirements for cytosolic [2Fe-2S] protein maturation.


Subject(s)
Cytosol , Glutaredoxins , Glutathione , Iron-Sulfur Proteins , Mitochondria , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Cytosol/metabolism , Iron-Sulfur Proteins/metabolism , Humans , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Glutathione/metabolism , Mitochondria/metabolism , Glutaredoxins/metabolism , Glutaredoxins/genetics , ATP-Binding Cassette Transporters/metabolism , Mitochondrial Proteins/metabolism
18.
New Phytol ; 242(6): 2787-2802, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38693568

ABSTRACT

Root-knot nematodes (RKN; Meloidogyne species) are plant pathogens that introduce several effectors in their hosts to facilitate infection. The actual targets and functioning mechanism of these effectors largely remain unexplored. This study illuminates the role and interplay of the Meloidogyne javanica nematode effector ROS suppressor (Mj-NEROSs) within the host plant environment. Mj-NEROSs suppresses INF1-induced cell death as well as flg22-induced callose deposition and reactive oxygen species (ROS) production. A transcriptome analysis highlighted the downregulation of ROS-related genes upon Mj-NEROSs expression. NEROSs interacts with the plant Rieske's iron-sulfur protein (ISP) as shown by yeast-two-hybrid and bimolecular fluorescence complementation. Secreted from the subventral pharyngeal glands into giant cells, Mj-NEROSs localizes in the plastids where it interacts with ISP, subsequently altering electron transport rates and ROS production. Moreover, our results demonstrate that isp Arabidopsis thaliana mutants exhibit increased susceptibility to M. javanica, indicating ISP importance for plant immunity. The interaction of a nematode effector with a plastid protein highlights the possible role of root plastids in plant defense, prompting many questions on the details of this process.


Subject(s)
Arabidopsis , Electron Transport Complex III , Plant Immunity , Plastids , Reactive Oxygen Species , Tylenchoidea , Reactive Oxygen Species/metabolism , Arabidopsis/parasitology , Arabidopsis/immunology , Arabidopsis/genetics , Tylenchoidea/physiology , Tylenchoidea/pathogenicity , Animals , Plastids/metabolism , Electron Transport Complex III/metabolism , Plant Diseases/parasitology , Plant Diseases/immunology , Helminth Proteins/metabolism , Helminth Proteins/genetics , Gene Expression Regulation, Plant , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Protein Binding , Mutation/genetics , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics
19.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119723, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38599324

ABSTRACT

Viruses have evolved complex mechanisms to exploit host factors for replication and assembly. In response, host cells have developed strategies to block viruses, engaging in a continuous co-evolutionary battle. This dynamic interaction often revolves around the competition for essential resources necessary for both host cell and virus replication. Notably, iron, required for the biosynthesis of several cofactors, including iron­sulfur (FeS) clusters, represents a critical element in the ongoing competition for resources between infectious agents and host. Although several recent studies have identified FeS cofactors at the core of virus replication machineries, our understanding of their specific roles and the cellular processes responsible for their incorporation into viral proteins remains limited. This review aims to consolidate our current knowledge of viral components that have been characterized as FeS proteins and elucidate how viruses harness these versatile cofactors to their benefit. Its objective is also to propose that viruses may depend on incorporation of FeS cofactors more extensively than is currently known. This has the potential to revolutionize our understanding of viral replication, thereby carrying significant implications for the development of strategies to target infections.


Subject(s)
Iron-Sulfur Proteins , Viral Proteins , Virus Replication , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Humans , Viral Proteins/metabolism , Viral Proteins/genetics , Viruses/metabolism , Viruses/genetics , Virus Diseases/metabolism , Virus Diseases/virology , Iron/metabolism , Animals , Host-Pathogen Interactions
20.
Adv Biol (Weinh) ; 8(5): e2300545, 2024 May.
Article in English | MEDLINE | ID: mdl-38574244

ABSTRACT

HapX and SreA are transcription factors that regulate the response of the fungus Aspergillus fumigatus to the availability of iron. During iron starvation, HapX represses genes involved in iron consuming pathways and upon a shift to iron excess, HapX activates these same genes. SreA blocks the expression of genes needed for iron uptake during periods of iron availability. Both proteins possess cysteine-rich regions (CRR) that are hypothesized to be necessary for the sensing of iron levels. However, the contribution of each of these domains to the function of the protein has remained unclear. Here, the ability of peptide analogs of each CRR is determined to bind an iron-sulfur cluster in vitro. UV-vis and resonance Raman (RR) spectroscopies reveal that each CRR is capable of coordinating a [2Fe-2S] cluster with comparable affinities. The iron-sulfur cluster coordinated to the CRR-B domain of HapX displays particularly high stability. The data are consistent with HapX and SreA mediating responses to cellular iron levels through the direct coordination of [2Fe-2S] clusters. The high stability of the CRR-B peptide may also find use as a starting point for the development of new green catalysts.


Subject(s)
Cysteine , Fungal Proteins , Iron-Sulfur Proteins , Peptides , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Cysteine/metabolism , Cysteine/chemistry , Peptides/metabolism , Peptides/chemistry , Fungal Proteins/metabolism , Fungal Proteins/chemistry , Fungal Proteins/genetics , Aspergillus fumigatus/metabolism , Aspergillus fumigatus/chemistry , Aspergillus fumigatus/genetics , Iron/metabolism , Protein Binding , Spectrum Analysis, Raman , Transcription Factors/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...