Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.899
Filtrer
1.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(7): 790-796, 2024 Jul 10.
Article de Chinois | MEDLINE | ID: mdl-38946359

RÉSUMÉ

OBJECTIVE: To explore the clinical characteristics and genetic variants in three children with late-onset Multiple acyl-Coenzyme A dehydrogenase deficiency (MADD type Ⅲ). METHODS: Clinical data of three children diagnosed with late-onset MADD at the Children's Hospital Affiliated to Zhengzhou University between March 2020 and March 2022 were retrospectively analyzed. All children were subjected to whole exome sequencing (WES), and candidate variants were verified by Sanger sequencing. All children had received improved metabolic therapy and followed up for 1 ~ 3 years. RESULTS: The children had included 2 males and 1 female, and aged from 2 months to 11 years and 7 months. Child 1 had intermittent vomiting, child 2 had weakness in lower limbs, while child 3 had no symptom except abnormal neonatal screening. Tandem mass spectrometry of the three children showed elevation of multiple acylcarnitines with short, medium and long chains. Children 1 and 2 showed increased glutaric acid and multiple dicarboxylic acids by urine Gas chromatography-mass spectrometry (GC-MS) analysis. All children were found to harbor compound heterozygous variants of the ETFDH gene, including a paternal c.1211T>C (p.M404T) and a maternal c.488-22T>G variant in child 1, a paternal c.1717C>T (p.Q573X) and a maternal c.250G>A (p.A84T) variant in child 2, and a paternal c.1285+1G>A and maternal c.629A>G (p.S210N) variant in child 3. As for the treatment, high-dose vitamin B2, levocarnitine and coenzyme Q10 were given to improve the metabolism, in addition with a low fat, hypoproteinic and high carbohydrate diet. All children showed a stable condition with normal growth and development during the follow-up. CONCLUSION: The compound heterozygous variants of the ETFDH gene probably underlay the muscle weakness, remittent vomiting, elevated short, medium, and long chain acylcarnitine, as well as elevated glutaric acid and various dicarboxylic acids in the three children with type Ⅲ MADD.


Sujet(s)
Flavoprotéines de transfert d'électrons , Déficit multiple en acyl CoA déshydrogénase , Humains , Mâle , Femelle , Déficit multiple en acyl CoA déshydrogénase/génétique , Nourrisson , Enfant , Enfant d'âge préscolaire , Flavoprotéines de transfert d'électrons/génétique , Mutation , Études rétrospectives , Carnitine/analogues et dérivés , Carnitine/sang , Ferrosulfoprotéines/génétique , , Oxidoreductases acting on CH-NH group donors/génétique , Variation génétique
2.
J Clin Invest ; 134(12)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38950322

RÉSUMÉ

Cytoplasmic and nuclear iron-sulfur (Fe-S) enzymes that are essential for genome maintenance and replication depend on the cytoplasmic Fe-S assembly (CIA) machinery for cluster acquisition. The core of the CIA machinery consists of a complex of CIAO1, MMS19 and FAM96B. The physiological consequences of loss of function in the components of the CIA pathway have thus far remained uncharacterized. Our study revealed that patients with biallelic loss of function in CIAO1 developed proximal and axial muscle weakness, fluctuating creatine kinase elevation, and respiratory insufficiency. In addition, they presented with CNS symptoms including learning difficulties and neurobehavioral comorbidities, along with iron deposition in deep brain nuclei, mild normocytic to macrocytic anemia, and gastrointestinal symptoms. Mutational analysis revealed reduced stability of the variants compared with WT CIAO1. Functional assays demonstrated failure of the variants identified in patients to recruit Fe-S recipient proteins, resulting in compromised activities of DNA helicases, polymerases, and repair enzymes that rely on the CIA complex to acquire their Fe-S cofactors. Lentivirus-mediated restoration of CIAO1 expression reversed all patient-derived cellular abnormalities. Our study identifies CIAO1 as a human disease gene and provides insights into the broader implications of the cytosolic Fe-S assembly pathway in human health and disease.


Sujet(s)
Ferrosulfoprotéines , Humains , Ferrosulfoprotéines/génétique , Ferrosulfoprotéines/métabolisme , Mâle , Femelle , Maladies neuromusculaires/génétique , Maladies neuromusculaires/enzymologie , Maladies neuromusculaires/métabolisme , Maladies neuromusculaires/anatomopathologie , Enfant , Noyau de la cellule/métabolisme , Noyau de la cellule/enzymologie , Noyau de la cellule/génétique , Cytoplasme/métabolisme , Cytoplasme/enzymologie , Métallochaperons
3.
Sci Rep ; 14(1): 15175, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956251

RÉSUMÉ

In the current study, we aimed to investigate whether disulfiram (DSF) exerts a neuroprotective role in cerebral ischemiareperfusion (CI-RI) injury by modulating ferredoxin 1 (FDX1) to regulate copper ion (Cu) levels and inhibiting inflammatory responses. To simulate CI-RI, a transient middle cerebral artery occlusion (tMCAO) model in C57/BL6 mice was employed. Mice were administered with or without DSF before and after tMCAO. Changes in infarct volume after tMCAO were observed using TTC staining. Nissl staining and hematoxylin-eosin (he) staining were used to observe the morphological changes of nerve cells at the microscopic level. The inhibitory effect of DSF on initial inflammation was verified by TUNEL assay, apoptosis-related protein detection and iron concentration detection. FDX1 is the main regulatory protein of copper death, and the occurrence of copper death will lead to the increase of HSP70 stress and inflammatory response. Cuproptosis-related proteins and downstream inflammatory factors were detected by western blotting, immunofluorescence staining, and immunohistochemistry. The content of copper ions was detected using a specific kit, while electron microscopy was employed to examine mitochondrial changes. We found that DSF reduced the cerebral infarction volume, regulated the expression of cuproptosis-related proteins, and modulated copper content through down regulation of FDX1 expression. Moreover, DSF inhibited the HSP70/TLR-4/NLRP3 signaling pathway. Collectively, DSF could regulate Cu homeostasis by inhibiting FDX1, acting on the HSP70/TLR4/NLRP3 pathway to alleviate CI/RI. Accordingly, DSF could mitigate inflammatory responses and safeguard mitochondrial integrity, yielding novel therapeutic targets and mechanisms for the clinical management of ischemia-reperfusion injury.


Sujet(s)
Cuivre , Disulfirame , Homéostasie , Inflammation , Souris de lignée C57BL , Lésion d'ischémie-reperfusion , Animaux , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/anatomopathologie , Disulfirame/pharmacologie , Souris , Cuivre/métabolisme , Homéostasie/effets des médicaments et des substances chimiques , Mâle , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Régulation négative/effets des médicaments et des substances chimiques , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Modèles animaux de maladie humaine , Ferrosulfoprotéines/métabolisme , Encéphalopathie ischémique/métabolisme , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Récepteur de type Toll-4/métabolisme
4.
Mol Genet Genomic Med ; 12(7): e2489, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38967380

RÉSUMÉ

BACKGROUND: Glutaric aciduria type II (GA2) is a rare genetic disorder inherited in an autosomal recessive manner. Double dosage mutations in GA2 corresponding genes, ETFDH, ETFA, and ETFB, lead to defects in the catabolism of fatty acids, and amino acids lead to broad-spectrum phenotypes, including muscle weakness, developmental delay, and seizures. product of these three genes have crucial role in transferring electrons to the electron transport chain (ETC), but are not directly involve in ETC complexes. METHODS: Here, by using exome sequencing, the cause of periodic cryptic gastrointestinal complications in a 19-year-old girl was resolved after years of diagnostic odyssey. Protein modeling for the novel variant served as another line of validation for it. RESULTS: Exome Sequencing (ES) identified two variants in ETFDH: ETFDH:c.926T>G and ETFDH:c.1141G>C. These variants are likely contributing to the crisis in this case. To the best of our knowledge at the time of writing this manuscript, variant ETFDH:c.926T>G is reported here for the first time. Clinical manifestations of the case and pathological analysis are in consistent with molecular findings. Protein modeling provided another line of evidence proving the pathogenicity of the novel variant. ETFDH:c.926T>G is reported here for the first time in relation to the causation GA2. CONCLUSION: Given the milder symptoms in this case, a review of GA2 cases caused by compound heterozygous mutations was conducted, highlighting the range of symptoms observed in these patients, from mild fatigue to more severe outcomes. The results underscore the importance of comprehensive genetic analysis in elucidating the spectrum of clinical presentations in GA2 and guiding personalized treatment strategies.


Sujet(s)
Flavoprotéines de transfert d'électrons , Hétérozygote , Ferrosulfoprotéines , Oxidoreductases acting on CH-NH group donors , Humains , Femelle , Flavoprotéines de transfert d'électrons/génétique , Ferrosulfoprotéines/génétique , Oxidoreductases acting on CH-NH group donors/génétique , Jeune adulte , Encéphalopathies métaboliques/génétique , Encéphalopathies métaboliques/anatomopathologie , Encéphalopathies métaboliques/diagnostic , Mutation , Déficit multiple en acyl CoA déshydrogénase/génétique , Déficit multiple en acyl CoA déshydrogénase/anatomopathologie
5.
Cell ; 187(13): 3357-3372.e19, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38866018

RÉSUMÉ

Microbial hydrogen (H2) cycling underpins the diversity and functionality of diverse anoxic ecosystems. Among the three evolutionarily distinct hydrogenase superfamilies responsible, [FeFe] hydrogenases were thought to be restricted to bacteria and eukaryotes. Here, we show that anaerobic archaea encode diverse, active, and ancient lineages of [FeFe] hydrogenases through combining analysis of existing and new genomes with extensive biochemical experiments. [FeFe] hydrogenases are encoded by genomes of nine archaeal phyla and expressed by H2-producing Asgard archaeon cultures. We report an ultraminimal hydrogenase in DPANN archaea that binds the catalytic H-cluster and produces H2. Moreover, we identify and characterize remarkable hybrid complexes formed through the fusion of [FeFe] and [NiFe] hydrogenases in ten other archaeal orders. Phylogenetic analysis and structural modeling suggest a deep evolutionary history of hybrid hydrogenases. These findings reveal new metabolic adaptations of archaea, streamlined H2 catalysts for biotechnological development, and a surprisingly intertwined evolutionary history between the two major H2-metabolizing enzymes.


Sujet(s)
Archéobactéries , Hydrogène , Hydrogenase , Phylogenèse , Archéobactéries/génétique , Archéobactéries/enzymologie , Protéines d'archée/métabolisme , Protéines d'archée/composition chimique , Protéines d'archée/génétique , Génome d'archéobactérie , Hydrogène/métabolisme , Hydrogenase/métabolisme , Hydrogenase/génétique , Hydrogenase/composition chimique , Ferrosulfoprotéines/métabolisme , Ferrosulfoprotéines/génétique , Ferrosulfoprotéines/composition chimique , Modèles moléculaires , Structure tertiaire des protéines
6.
Spectrochim Acta A Mol Biomol Spectrosc ; 320: 124603, 2024 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-38878720

RÉSUMÉ

Iron-sulfur cluster conversion and nitrosyl modification are involved in regulating their functions and play critical roles in signaling for biological systems. Hereby, the photo-induced dynamic process of (Me4N)2[Fe2S2(NO)4] was monitored using time-resolved electron paramagnetic resonance (EPR) spectra, MS spectra and cellular imaging methods. Photo-irradiation and the solvent affect the reaction rates and products. Spectroscopic and kinetic studies have shown that the process involves at least three intermediates: spin-trapped NO free radical species with a gav at 2.040, and two other iron nitrosyl species, dinitrosyl iron units (DNICs) and mononitrosyl iron units (MNICs) with gav values at 2.031 and 2.024, respectively. Moreover, the [Fe2S2(NO)4]2- cluster could bind with ferritin and decompose gradually, and a binding state of dinitrosyl iron coordinated with Cys102 of the recombinant human heavy chain ferritin (rHuHF) was finally formed. This study provides insight into the photodynamic mechanism of nitrosyl iron - sulfur clusters to improve the understanding of physiological activity.


Sujet(s)
Fer , Humains , Spectroscopie de résonance de spin électronique , Fer/composition chimique , Fer/métabolisme , Oxydes d'azote/composition chimique , Oxydes d'azote/métabolisme , Liaison aux protéines , Cinétique , Ferrosulfoprotéines/métabolisme , Ferrosulfoprotéines/composition chimique , Soufre/composition chimique , Soufre/métabolisme , Ferritines/composition chimique , Ferritines/métabolisme , Lumière
7.
Gut Microbes ; 16(1): 2359665, 2024.
Article de Anglais | MEDLINE | ID: mdl-38831611

RÉSUMÉ

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.


Sujet(s)
Protéines bactériennes , Enterococcus faecium , Ferrosulfoprotéines , Enterococcus faecium/génétique , Enterococcus faecium/métabolisme , Enterococcus faecium/croissance et développement , Animaux , Souris , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Anaérobiose , Ferrosulfoprotéines/génétique , Ferrosulfoprotéines/métabolisme , Tube digestif/microbiologie , Microbiome gastro-intestinal , Infections bactériennes à Gram positif/microbiologie , Humains , Éléments transposables d'ADN , Métabolisme glucidique , Femelle , Acetyltransferases
8.
Nat Commun ; 15(1): 5303, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38906898

RÉSUMÉ

The methylerythritol phosphate (MEP) pathway is responsible for biosynthesis of the precursors of isoprenoid compounds in eubacteria and plastids. It is a metabolic alternative to the well-known mevalonate pathway for isoprenoid production found in archaea and eukaryotes. Recently, a role for the MEP pathway in oxidative stress detection, signalling, and response has been identified. This role is executed in part through the unusual cyclic intermediate, methylerythritol cyclodiphosphate (MEcDP). We postulate that this response is triggered through the oxygen sensitivity of the MEP pathway's terminal iron-sulfur (Fe-S) cluster enzymes. MEcDP is the substrate of IspG, the first Fe-S cluster enzyme in the pathway; it accumulates under oxidative stress conditions and acts as a signalling molecule. It may also act as an antioxidant. Furthermore, evidence is emerging for a broader and highly nuanced role of the MEP pathway in oxidative stress responses, implemented through a complex system of differential regulation and sensitivity at numerous nodes in the pathway. Here, we explore the evidence for such a role (including the contribution of the Fe-S cluster enzymes and different pathway metabolites, especially MEcDP), the evolutionary implications, and the many questions remaining about the behaviour of the MEP pathway in the presence of oxidative stress.


Sujet(s)
Érythritol , Stress oxydatif , Oses phosphates , Érythritol/métabolisme , Érythritol/analogues et dérivés , Oses phosphates/métabolisme , Ferrosulfoprotéines/métabolisme , Transduction du signal , Terpènes/métabolisme
9.
Zool Res ; 45(4): 821-830, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-38894524

RÉSUMÉ

Magnetic sense, or termed magnetoreception, has evolved in a broad range of taxa within the animal kingdom to facilitate orientation and navigation. MagRs, highly conserved A-type iron-sulfur proteins, are widely distributed across all phyla and play essential roles in both magnetoreception and iron-sulfur cluster biogenesis. However, the evolutionary origins and functional diversification of MagRs from their prokaryotic ancestor remain unclear. In this study, MagR sequences from 131 species, ranging from bacteria to humans, were selected for analysis, with 23 representative sequences covering species from prokaryotes to Mollusca, Arthropoda, Osteichthyes, Reptilia, Aves, and mammals chosen for protein expression and purification. Biochemical studies revealed a gradual increase in total iron content in MagRs during evolution. Three types of MagRs were identified, each with distinct iron and/or iron-sulfur cluster binding capacity and protein stability, indicating continuous expansion of the functional roles of MagRs during speciation and evolution. This evolutionary biochemical study provides valuable insights into how evolution shapes the physical and chemical properties of biological molecules such as MagRs and how these properties influence the evolutionary trajectories of MagRs.


Sujet(s)
Ferrosulfoprotéines , Animaux , Ferrosulfoprotéines/génétique , Ferrosulfoprotéines/métabolisme , Ferrosulfoprotéines/composition chimique , Évolution biologique , Évolution moléculaire , Phylogenèse , Fer/métabolisme
10.
Open Biol ; 14(6): 240033, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38919062

RÉSUMÉ

Aspergillus fumigatus is the predominant mould pathogen for humans. Adaption to host-imposed iron limitation has previously been demonstrated to be essential for its virulence. [2Fe-2S] clusters are crucial as cofactors of several metabolic pathways and mediate cytosolic/nuclear iron sensing in fungi including A. fumigatus. [2Fe-2S] cluster trafficking has been shown to involve BolA family proteins in both mitochondria and the cytosol/nucleus. Interestingly, both A. fumigatus homologues, termed Bol1 and Bol3, possess mitochondrial targeting sequences, suggesting the lack of cytosolic/nuclear versions. Here, we show by the combination of mutational, proteomic and fluorescence microscopic analyses that expression of the Bol3 encoding gene leads to dual localization of gene products to mitochondria and the cytosol/nucleus via alternative translation initiation downstream of the mitochondrial targeting sequence, which appears to be highly conserved in various Aspergillus species. Lack of either mitochondrial Bol1 or Bol3 was phenotypically inconspicuous while lack of cytosolic/nuclear Bol3 impaired growth during iron limitation but not iron sensing which indicates a particular importance of [2Fe-2S] cluster trafficking during iron limitation. Remarkably, cytosolic/nuclear Bol3 differs from the mitochondrial version only by N-terminal acetylation, a finding that was only possible by mutational hypothesis testing.


Sujet(s)
Aspergillus fumigatus , Cytosol , Protéines fongiques , Fer , Mitochondries , Aspergillus fumigatus/métabolisme , Aspergillus fumigatus/génétique , Protéines fongiques/métabolisme , Protéines fongiques/génétique , Cytosol/métabolisme , Mitochondries/métabolisme , Fer/métabolisme , Adaptation physiologique , Noyau de la cellule/métabolisme , Transport des protéines , Protéomique/méthodes , Ferrosulfoprotéines/métabolisme , Ferrosulfoprotéines/génétique , Régulation de l'expression des gènes fongiques , Acétylation
11.
Biophys J ; 123(12): 1648-1653, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38733082

RÉSUMÉ

DNA primase is an iron sulfur enzyme in DNA replication responsible for synthesizing short RNA primers that serve as starting points for DNA synthesis. The role of the [4Fe-4S] cluster is not well determined. Here, we calculate the redox potential of the [4Fe-4S] with and without DNA/RNA using continuum electrostatics. In addition, we identify the structural changes coupled to the oxidation/reduction. Our calculations show that the DNA/RNA primer lowers the redox potential by 110 and 50 mV for the [4Fe-4S]+ and [4Fe-4S]2+ states, respectively. The oxidation of the cluster is coupled to structural changes that significantly reduce the binding energies between the DNA and the nearby residues. The negative charges accumulated by the DNA and the RNA primers induce the oxidation of the [4Fe-4S] cluster. This in turn stimulates structural changes on the DNA-protein interface that significantly reduce the binding energies.


Sujet(s)
DNA primase , Ferrosulfoprotéines , Oxydoréduction , Liaison aux protéines , ARN , DNA primase/métabolisme , DNA primase/composition chimique , ARN/métabolisme , ARN/composition chimique , Ferrosulfoprotéines/composition chimique , Ferrosulfoprotéines/métabolisme , ADN/métabolisme , ADN/composition chimique , Thermodynamique , Modèles moléculaires
12.
Microbiol Spectr ; 12(7): e0425623, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38785428

RÉSUMÉ

Isoprenoids are a diverse family of compounds that are synthesized from two isomeric compounds, isopentenyl diphosphate and dimethylallyl diphosphate. In most bacteria, isoprenoids are produced from the essential methylerythritol phosphate (MEP) pathway. The terminal enzymes of the MEP pathway IspG and IspH are [4Fe-4S] cluster proteins, and in Zymomonas mobilis, the substrates of IspG and IspH accumulate in cells in response to O2, suggesting possible lability of their [4Fe-4S] clusters. Here, we show using complementation assays in Escherichia coli that even under anaerobic conditions, Z. mobilis IspG and IspH are not as functional as their E. coli counterparts, requiring higher levels of expression to rescue viability. A deficit of the sulfur utilization factor (SUF) Fe-S cluster biogenesis pathway did not explain the reduced function of Z. mobilis IspG and IspH since no improvement in viability was observed in E. coli expressing the Z. mobilis SUF pathway or having increased expression of the E. coli SUF pathway. Complementation of single and double mutants with various combinations of Z. mobilis and E. coli IspG and IspH indicated that optimal growth required the pairing of IspG and IspH from the same species. Furthermore, Z. mobilis IspH conferred an O2-sensitive growth defect to E. coli that could be partially rescued by co-expression of Z. mobilis IspG. In vitro analysis showed O2 sensitivity of the [4Fe-4S] cluster of both Z. mobilis IspG and IspH. Altogether, our data indicate an important role of the cognate protein IspG in Z. mobilis IspH function under both aerobic and anaerobic conditions. IMPORTANCE: Isoprenoids are one of the largest classes of natural products, exhibiting diversity in structure and function. They also include compounds that are essential for cellular life across the biological world. In bacteria, isoprenoids are derived from two precursors, isopentenyl diphosphate and dimethylallyl diphosphate, synthesized primarily by the methylerythritol phosphate pathway. The aerotolerant Z. mobilis has the potential for methylerythritol phosphate pathway engineering by diverting some of the glucose that is typically efficiently converted into ethanol to produce isoprenoid precursors to make bioproducts and biofuels. Our data revealed the surprising finding that Z. mobilis IspG and IspH need to be co-optimized to improve flux via the methyl erythritol phosphate pathway in part to evade the oxygen sensitivity of IspH.


Sujet(s)
Protéines bactériennes , Érythritol , Escherichia coli , Zymomonas , Zymomonas/métabolisme , Zymomonas/enzymologie , Zymomonas/génétique , Érythritol/métabolisme , Érythritol/analogues et dérivés , Escherichia coli/génétique , Escherichia coli/métabolisme , Escherichia coli/enzymologie , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Protéines Escherichia coli/métabolisme , Protéines Escherichia coli/génétique , Ferrosulfoprotéines/métabolisme , Ferrosulfoprotéines/génétique , Terpènes/métabolisme , Oxidoreductases
13.
Nat Commun ; 15(1): 4655, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38821922

RÉSUMÉ

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.


Sujet(s)
ATPases associated with diverse cellular activities , Complexe III de la chaîne respiratoire , Humains , ADP/métabolisme , Adénosine triphosphate/métabolisme , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/composition chimique , Cryomicroscopie électronique , Complexe III de la chaîne respiratoire/métabolisme , Complexe III de la chaîne respiratoire/composition chimique , Hydrolyse , Ferrosulfoprotéines/métabolisme , Ferrosulfoprotéines/composition chimique , Modèles moléculaires , Conformation des protéines , Pliage des protéines , Transport des protéines
14.
Proc Natl Acad Sci U S A ; 121(21): e2400740121, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38743629

RÉSUMÉ

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.


Sujet(s)
Cytosol , Glutarédoxines , Glutathion , Ferrosulfoprotéines , Mitochondries , Protéines de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Cytosol/métabolisme , Ferrosulfoprotéines/métabolisme , Humains , Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , Glutathion/métabolisme , Mitochondries/métabolisme , Glutarédoxines/métabolisme , Glutarédoxines/génétique , Transporteurs ABC/métabolisme , Protéines mitochondriales/métabolisme
15.
Adv Microb Physiol ; 84: 243-307, 2024.
Article de Anglais | MEDLINE | ID: mdl-38821633

RÉSUMÉ

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.


Sujet(s)
Organites , Organites/métabolisme , Mitochondries/métabolisme , Eucaryotes/métabolisme , Ferrosulfoprotéines/métabolisme , Ferrosulfoprotéines/génétique
16.
J Am Chem Soc ; 146(23): 15771-15778, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38819401

RÉSUMÉ

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.


Sujet(s)
Hydrogenase , Ferrosulfoprotéines , Hydrogenase/composition chimique , Hydrogenase/métabolisme , Ferrosulfoprotéines/composition chimique , Ferrosulfoprotéines/métabolisme , Sites de fixation , Protéines bactériennes/composition chimique , Protéines bactériennes/métabolisme , Fer/composition chimique , Fer/métabolisme , Modèles moléculaires
17.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119746, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38719030

RÉSUMÉ

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.


Sujet(s)
Protéines bactériennes , Ferrosulfoprotéines , Ferrosulfoprotéines/métabolisme , Ferrosulfoprotéines/génétique , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Fer/métabolisme , Soufre/métabolisme , Bactéries/génétique , Bactéries/métabolisme
18.
J Mol Cell Cardiol ; 192: 36-47, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38734062

RÉSUMÉ

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.


Sujet(s)
Aconitate hydratase , Ferroptose , Souris knockout , Myocytes cardiaques , Phénylènediamines , Sirtuine-3 , Animaux , Sirtuine-3/métabolisme , Sirtuine-3/génétique , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Aconitate hydratase/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Souris , Acétylation , Phénylènediamines/pharmacologie , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Ferrosulfoprotéines/métabolisme , Ferrosulfoprotéines/génétique , Fer/métabolisme , , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/génétique , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/effets des médicaments et des substances chimiques , Protéines de liaison au fer/métabolisme , Protéines de liaison au fer/génétique , Défaillance cardiaque/métabolisme , Défaillance cardiaque/génétique , Cytosol/métabolisme , Cyclohexylamines
19.
Biochim Biophys Acta Mol Cell Res ; 1871(6): 119750, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38762171

RÉSUMÉ

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.


Sujet(s)
Azotobacter vinelandii , Protéines bactériennes , Ferrosulfoprotéines , Fixation de l'azote , Azotobacter vinelandii/métabolisme , Azotobacter vinelandii/génétique , Ferrosulfoprotéines/métabolisme , Ferrosulfoprotéines/génétique , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique
20.
Biochim Biophys Acta Mol Cell Res ; 1871(6): 119749, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38763301

RÉSUMÉ

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.


Sujet(s)
Protéines Escherichia coli , Homéostasie , Ferrosulfoprotéines , Fer , Ferrosulfoprotéines/métabolisme , Ferrosulfoprotéines/génétique , Fer/métabolisme , Protéines Escherichia coli/métabolisme , Protéines Escherichia coli/génétique , Régulation de l'expression des gènes bactériens , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Humains , Escherichia coli/métabolisme , Escherichia coli/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...