Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 830
Filter
1.
Yakugaku Zasshi ; 144(6): 633-641, 2024.
Article in Japanese | MEDLINE | ID: mdl-38825472

ABSTRACT

Iron is necessary for all living organisms, and bacteria that cause infections in human hosts also need ferrous ions for their growth and proliferation. In the human body, most ferric ions (Fe3+) are tightly bound to iron-binding proteins such as hemoglobin, transferrin, lactoferrin, and ferritin. Pathogenic bacteria express highly specific iron uptake systems, including siderophores and specific receptors. Most bacteria secrete siderophores, which are low-molecular weight metal-chelating agents, to capture Fe3+ outside cell. Siderophores are mainly classified as either catecholate or hydroxamate. Vibrio vulnificus, a Gram-negative pathogenic bacterium, is responsible for serious infections in humans and requires iron for growth. A clinical isolate, V. vulnificus M2799, secretes a catecholate siderophore, vulnibactin, that captures ferric ions from the environment. In our study, we generated deletion mutants of the genes encoding proteins involved in the vulnibactin mediated iron-utilization system, such as ferric-vulnibactin receptor protein (VuuA), periplasmic ferric-vulnibactin binding protein (FatB), ferric-vulnibactin reductase (VuuB), and isochorismate synthase (ICS). ICS and VuuA are required under low-iron conditions for ferric-utilization in M2799, but the alternative proteins FatB and VuuB can function as a periplasmic binding protein and a ferric-chelate reductase, respectively. VatD, which functions as ferric-hydroxamate siderophores periplasmic binding protein, was shown to participate in the ferric-vulnibactin uptake system in the absence of FatB. Furthermore, the ferric-hydroxamate siderophore reductase IutB was observed to participate in ferric-vulnibactin reduction in the absence of VuuB. We propose that ferric-siderophore periplasmic binding proteins and ferric-chelate reductases represent potential targets for drug discovery in the context of infectious diseases.


Subject(s)
Drug Discovery , Iron , Siderophores , Iron/metabolism , Siderophores/metabolism , Humans , Bacterial Infections/drug therapy , Bacterial Infections/metabolism , Molecular Targeted Therapy , Hydroxamic Acids/metabolism , Iron-Binding Proteins/metabolism
2.
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
3.
Protein Expr Purif ; 219: 106487, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38657915

ABSTRACT

The bacterial Efe system functions as an importer of free Fe2+ into cells independently of iron-chelating compounds such as siderophores and consisted of iron-binding protein EfeO, peroxidase EfeB, and transmembrane permease EfeU. While we and other researchers reported crystal structures of EfeO and EfeB, that of EfeU remains undetermined. In this study, we constructed expression system of EfeU derived from Escherichia coli, selected E. coli Rosetta-gami 2 (DE3) as an expression host, and succeeded in purification of the proteins which were indicated to form an oligomer by blue native PAGE. We obtained preliminary data of the X-ray crystallography, suggesting that expression and purification methods we established in this study enable structural analysis of the bacterial Efe system.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Iron , Escherichia coli/genetics , Escherichia coli/metabolism , Crystallography, X-Ray , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/isolation & purification , Iron/metabolism , Iron/chemistry , Gene Expression , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/biosynthesis , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/genetics , Iron-Binding Proteins/isolation & purification , Iron-Binding Proteins/metabolism
4.
Sci Rep ; 14(1): 8391, 2024 04 10.
Article in English | MEDLINE | ID: mdl-38600238

ABSTRACT

Friedreich's ataxia is a degenerative and progressive multisystem disorder caused by mutations in the highly conserved frataxin (FXN) gene that results in FXN protein deficiency and mitochondrial dysfunction. While gene therapy approaches are promising, consistent induction of therapeutic FXN protein expression that is sub-toxic has proven challenging, and numerous therapeutic approaches are being tested in animal models. FXN (hFXN in humans, mFXN in mice) is proteolytically modified in mitochondria to produce mature FXN. However, unlike endogenous hFXN, endogenous mFXN is further processed into N-terminally truncated, extra-mitochondrial mFXN forms of unknown function. This study assessed mature exogenous hFXN expression levels in the heart and liver of C57Bl/6 mice 7-10 months after intravenous administration of a recombinant adeno-associated virus encoding hFXN (AAVrh.10hFXN) and examined the potential for hFXN truncation in mice. AAVrh.10hFXN induced dose-dependent expression of hFXN in the heart and liver. Interestingly, hFXN was processed into truncated forms, but found at lower levels than mature hFXN. However, the truncations were at different positions than mFXN. AAVrh.10hFXN induced mature hFXN expression in mouse heart and liver at levels that approximated endogenous mFXN levels. These results suggest that AAVrh.10hFXN can likely induce expression of therapeutic levels of mature hFXN in mice.


Subject(s)
Frataxin , Friedreich Ataxia , Humans , Animals , Mice , Heart , Protein Processing, Post-Translational , Liver/metabolism , Genetic Therapy , Iron-Binding Proteins/metabolism , Friedreich Ataxia/therapy , Friedreich Ataxia/drug therapy
5.
Nat Commun ; 15(1): 3269, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627381

ABSTRACT

Maturation of iron-sulfur proteins in eukaryotes is initiated in mitochondria by the core iron-sulfur cluster assembly (ISC) complex, consisting of the cysteine desulfurase sub-complex NFS1-ISD11-ACP1, the scaffold protein ISCU2, the electron donor ferredoxin FDX2, and frataxin, a protein dysfunctional in Friedreich's ataxia. The core ISC complex synthesizes [2Fe-2S] clusters de novo from Fe and a persulfide (SSH) bound at conserved cluster assembly site residues. Here, we elucidate the poorly understood Fe-dependent mechanism of persulfide transfer from cysteine desulfurase NFS1 to ISCU2. High-resolution cryo-EM structures obtained from anaerobically prepared samples provide snapshots that both visualize different stages of persulfide transfer from Cys381NFS1 to Cys138ISCU2 and clarify the molecular role of frataxin in optimally positioning assembly site residues for fast sulfur transfer. Biochemical analyses assign ISCU2 residues essential for sulfur transfer, and reveal that Cys138ISCU2 rapidly receives the persulfide without a detectable intermediate. Mössbauer spectroscopy assessing the Fe coordination of various sulfur transfer intermediates shows a dynamic equilibrium between pre- and post-sulfur-transfer states shifted by frataxin. Collectively, our study defines crucial mechanistic stages of physiological [2Fe-2S] cluster assembly and clarifies frataxin's molecular role in this fundamental process.


Subject(s)
Frataxin , Iron-Sulfur Proteins , Iron-Sulfur Proteins/metabolism , Sulfides/metabolism , Sulfur/metabolism , Carbon-Sulfur Lyases/metabolism , Iron-Binding Proteins/metabolism
6.
Thyroid ; 34(5): 659-667, 2024 May.
Article in English | MEDLINE | ID: mdl-38482822

ABSTRACT

Background: Congenital hypothyroidism (CH) is caused by mutations in cysteine residues, including Cys655 and Cys825 that form disulfide bonds in thyroid peroxidase (TPO). It is highly likely that these disulfide bonds could play an important role in TPO activity. However, to date, no study has comprehensively analyzed cysteine mutations that form disulfide bonds in TPO. In this study, we induced mutations in cysteine residues involved in disulfide bonds formation and analyzed their effect on subcellular localization, degradation, and enzyme activities to evaluate the importance of disulfide bonds in TPO activity. Methods: Vector plasmid TPO mutants, C655F and C825R, known to occur in CH, were transfected into HEK293 cells. TPO activity and protein expression levels were measured by the Amplex red assay and Western blotting. The same procedure was performed in the presence of MG132 proteasome inhibitor. Subcellular localization was determined using immunocytochemistry and flow cytometry. The locations of all disulfide bonds within TPO were predicted using in silico analysis. All TPO mutations associated with disulfide bonds were induced. TPO activity and protein expression levels were also measured in all TPO mutants associated with disulfide bonds using the Amplex red assay and Western blotting. Results: C655F and C825R showed significantly decreased activity and protein expression compared with the wild type (WT) (p < 0.05). In the presence of the MG132 proteasome inhibitor, the protein expression level of TPO increased to a level comparable with that of the WT without increases in its activity. The degree of subcellular distribution of TPO to the cell surface in the mutants was lower compared with the WT TPO. Twenty-four cysteine residues were involved in the formation of 12 disulfide bonds in TPO. All TPO mutants harboring an amino acid substitution in each cysteine showed significantly reduced TPO activity and protein expression levels. Furthermore, the differences in TPO activity depended on the position of the disulfide bond. Conclusions: All 12 disulfide bonds play an important role in the activity of TPO. Furthermore, the mutations lead to misfolding, degradation, and membrane insertion.


Subject(s)
Disulfides , Iodide Peroxidase , Proteasome Endopeptidase Complex , Humans , Iodide Peroxidase/metabolism , Iodide Peroxidase/genetics , Iodide Peroxidase/chemistry , HEK293 Cells , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/genetics , Disulfides/metabolism , Disulfides/chemistry , Mutation , Congenital Hypothyroidism/genetics , Congenital Hypothyroidism/metabolism , Cysteine/metabolism , Proteolysis , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Autoantigens
7.
Proc Natl Acad Sci U S A ; 121(12): e2308478121, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38489389

ABSTRACT

The marine cyanobacterium Prochlorococcus is a main contributor to global photosynthesis, whilst being limited by iron availability. Cyanobacterial genomes generally encode two different types of FutA iron-binding proteins: periplasmic FutA2 ABC transporter subunits bind Fe(III), while cytosolic FutA1 binds Fe(II). Owing to their small size and their economized genome Prochlorococcus ecotypes typically possess a single futA gene. How the encoded FutA protein might bind different Fe oxidation states was previously unknown. Here, we use structural biology techniques at room temperature to probe the dynamic behavior of FutA. Neutron diffraction confirmed four negatively charged tyrosinates, that together with a neutral water molecule coordinate iron in trigonal bipyramidal geometry. Positioning of the positively charged Arg103 side chain in the second coordination shell yields an overall charge-neutral Fe(III) binding state in structures determined by neutron diffraction and serial femtosecond crystallography. Conventional rotation X-ray crystallography using a home source revealed X-ray-induced photoreduction of the iron center with observation of the Fe(II) binding state; here, an additional positioning of the Arg203 side chain in the second coordination shell maintained an overall charge neutral Fe(II) binding site. Dose series using serial synchrotron crystallography and an XFEL X-ray pump-probe approach capture the transition between Fe(III) and Fe(II) states, revealing how Arg203 operates as a switch to accommodate the different iron oxidation states. This switching ability of the Prochlorococcus FutA protein may reflect ecological adaptation by genome streamlining and loss of specialized FutA proteins.


Subject(s)
Ferric Compounds , Prochlorococcus , Ferric Compounds/chemistry , Iron-Binding Proteins/metabolism , Prochlorococcus/metabolism , Iron/metabolism , Oxidation-Reduction , Transferrin/metabolism , Water/chemistry , Ferrous Compounds/chemistry , Crystallography, X-Ray
8.
Int J Mol Sci ; 25(3)2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38339189

ABSTRACT

Nqo15 is a subunit of respiratory complex I of the bacterium Thermus thermophilus, with strong structural similarity to human frataxin (FXN), a protein involved in the mitochondrial disease Friedreich's ataxia (FRDA). Recently, we showed that the expression of recombinant Nqo15 can ameliorate the respiratory phenotype of FRDA patients' cells, and this prompted us to further characterize both the Nqo15 solution's behavior and its potential functional overlap with FXN, using a combination of in silico and in vitro techniques. We studied the analogy of Nqo15 and FXN by performing extensive database searches based on sequence and structure. Nqo15's folding and flexibility were investigated by combining nuclear magnetic resonance (NMR), circular dichroism, and coarse-grained molecular dynamics simulations. Nqo15's iron-binding properties were studied using NMR, fluorescence, and specific assays and its desulfurase activation by biochemical assays. We found that the recombinant Nqo15 isolated from complex I is monomeric, stable, folded in solution, and highly dynamic. Nqo15 does not share the iron-binding properties of FXN or its desulfurase activation function.


Subject(s)
Frataxin , Friedreich Ataxia , Humans , Electron Transport Complex I/metabolism , Thermus thermophilus/metabolism , Molecular Dynamics Simulation , Iron/metabolism , Iron-Binding Proteins/metabolism , Friedreich Ataxia/metabolism
9.
J Neurol ; 271(4): 1844-1849, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38063871

ABSTRACT

BACKGROUND: Friedreich ataxia is a progressive multisystem disorder caused by deficiency of the protein frataxin; a small mitochondrial protein involved in iron sulfur cluster synthesis. Two types of frataxin exist: FXN-M, found in most cells, and FXN-E, found almost exclusively in red blood cells. Treatments in clinical trials include frataxin restoration by gene therapy, protein replacement, and epigenetic therapies, all of which necessitate sensitive assays for assessing frataxin levels. METHODS: In the present study, we have used a triple quadrupole mass spectrometry-based assay to examine the features of both types of frataxin levels in blood in a large heterogenous cohort of 106 patients with FRDA. RESULTS: Frataxin levels (FXN-E and FXN M) were predicted by GAA repeat length in regression models (R2 values = 0.51 and 0.27, respectively), and conversely frataxin levels predicted clinical status as determined by modified Friedreich Ataxia Rating scale scores and by disability status (R2 values = 0.13-0.16). There was no significant change in frataxin levels in individual subjects over time, and apart from start codon mutations, FXN-E and FXN-M levels were roughly equal. Accounting for hemoglobin levels in a smaller sub-cohort improved prediction of both FXN-E and FXN-M levels from R2 values of (0.3-0.38 to 0.20-0.51). CONCLUSION: The present data show that assay of FXN-M and FXN-E levels in blood provides an appropriate biofluid for assessing their repletion in particular clinical contexts.


Subject(s)
Frataxin , Friedreich Ataxia , Humans , Friedreich Ataxia/genetics , Mitochondrial Proteins/genetics , Mass Spectrometry , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism
10.
Am J Physiol Heart Circ Physiol ; 326(2): H357-H369, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38038720

ABSTRACT

Friedreich's ataxia (FA) is an autosomal recessive disorder caused by a deficiency in frataxin (FXN), a mitochondrial protein that plays a critical role in the synthesis of iron-sulfur clusters (Fe-S), vital inorganic cofactors necessary for numerous cellular processes. FA is characterized by progressive ataxia and hypertrophic cardiomyopathy, with cardiac dysfunction as the most common cause of mortality in patients. Commonly used cardiac-specific mouse models of FA use the muscle creatine kinase (MCK) promoter to express Cre recombinase in cardiomyocytes and striated muscle cells in mice with one conditional Fxn allele and one floxed-out/null allele. These mice quickly develop cardiomyopathy that becomes fatal by 9-11 wk of age. Here, we generated a cardiac-specific model with floxed Fxn allele homozygosity (MCK-Fxnflox/flox). MCK-Fxnflox/flox mice were phenotypically normal at 9 wk of age, despite no detectable FXN protein expression. Between 13 and 15 wk of age, these mice began to display progressive cardiomyopathy, including decreased ejection fraction and fractional shortening and increased left ventricular mass. MCK-Fxnflox/flox mice began to lose weight around 16 wk of age, characteristically associated with heart failure in other cardiac-specific FA models. By 18 wk of age, MCK-Fxnflox/flox mice displayed elevated markers of Fe-S deficiency, cardiac stress and injury, and cardiac fibrosis. This modified model reproduced important pathophysiological and biochemical features of FA over a longer timescale than previous cardiac-specific mouse models, offering a larger window for studying potential therapeutics.NEW & NOTEWORTHY Previous cardiac-specific frataxin knockout models exhibit rapid and fatal cardiomyopathy by 9 wk of age. This severe phenotype poses challenges for the design and execution of intervention studies. We introduce an alternative cardiac-specific model, MCK-Fxnflox/flox, with increased longevity and delayed onset of all major phenotypes. These phenotypes develop to the same severity as previous models. Thus, this new model provides the same cardiomyopathy-associated mortality with a larger window for potential studies.


Subject(s)
Cardiomyopathies , Friedreich Ataxia , Humans , Mice , Animals , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Alleles , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Disease Models, Animal , Frataxin , Myocytes, Cardiac/metabolism
11.
Cell Death Dis ; 14(12): 805, 2023 12 08.
Article in English | MEDLINE | ID: mdl-38062036

ABSTRACT

Friedreich ataxia (FRDA) is a rare, inherited neurodegenerative disease caused by an expanded GAA repeat in the first intron of the FXN gene, leading to transcriptional silencing and reduced expression of frataxin. Frataxin participates in the mitochondrial assembly of FeS clusters, redox cofactors of the respiratory complexes I, II and III. To date it is still unclear how frataxin deficiency culminates in the decrease of bioenergetics efficiency in FRDA patients' cells. We previously demonstrated that in healthy cells frataxin is closely attached to the mitochondrial cristae, which contain both the FeS cluster assembly machinery and the respiratory chain complexes, whereas in FRDA patients' cells with impaired respiration the residual frataxin is largely displaced in the matrix. To gain novel insights into the function of frataxin in the mitochondrial pathophysiology, and in the upstream metabolic defects leading to FRDA disease onset and progression, here we explored the potential interaction of frataxin with the FeS cluster-containing respiratory complexes I, II and III. Using healthy cells and different FRDA cellular models we found that frataxin interacts with these three respiratory complexes. Furthermore, by EPR spectroscopy, we observed that in mitochondria from FRDA patients' cells the decreased level of frataxin specifically affects the FeS cluster content of complex I. Remarkably, we also found that the frataxin-like protein Nqo15 from T. thermophilus complex I ameliorates the mitochondrial respiratory phenotype when expressed in FRDA patient's cells. Our data point to a structural and functional interaction of frataxin with complex I and open a perspective to explore therapeutic rationales for FRDA targeted to this respiratory complex.


Subject(s)
Friedreich Ataxia , Neurodegenerative Diseases , Humans , Electron Transport , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Mitochondrial Membranes/metabolism , Neurodegenerative Diseases/metabolism
12.
Emerg Top Life Sci ; 7(3): 313-323, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-37698160

ABSTRACT

Friedreich ataxia (FRDA) is an inherited disease that is typically caused by GAA repeat expansion within the first intron of the FXN gene coding for frataxin. This results in the frataxin deficiency that affects mostly muscle, nervous, and cardiovascular systems with progressive worsening of the symptoms over the years. This review summarizes recent progress that was achieved in understanding of molecular mechanism of the disease over the last few years and latest treatment strategies focused on overcoming the frataxin deficiency.


Subject(s)
Friedreich Ataxia , Humans , Friedreich Ataxia/genetics , Friedreich Ataxia/therapy , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Trinucleotide Repeat Expansion/genetics , Introns
13.
Dis Model Mech ; 16(10)2023 10 01.
Article in English | MEDLINE | ID: mdl-37691621

ABSTRACT

Cardiomyopathy is often fatal in Friedreich ataxia (FA). However, FA hearts maintain adequate function until advanced disease stages, suggesting initial adaptation to the loss of frataxin (FXN). Conditional cardiac knockout mouse models of FXN show transcriptional and metabolic profiles of the mitochondrial integrated stress response (ISRmt), which could play an adaptive role. However, the ISRmt has not been investigated in models with disease-relevant, partial decrease in FXN. We characterized the heart transcriptomes and metabolomes of three mouse models with varying degrees of FXN depletion: YG8-800, KIKO-700 and FXNG127V. Few metabolites were changed in YG8-800 mice, which did not provide a signature of cardiomyopathy or ISRmt; several metabolites were altered in FXNG127V and KIKO-700 hearts. Transcriptional changes were found in all models, but differentially expressed genes consistent with cardiomyopathy and ISRmt were only identified in FXNG127V hearts. However, these changes were surprisingly mild even at advanced age (18 months), despite a severe decrease in FXN levels to 1% of those of wild type. These findings indicate that the mouse heart has low reliance on FXN, highlighting the difficulty in modeling genetically relevant FA cardiomyopathy.


Subject(s)
Cardiomyopathies , Friedreich Ataxia , Mice , Animals , Multiomics , Heart , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Cardiomyopathies/genetics , Mice, Knockout , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Frataxin
14.
FEBS J ; 290(24): 5811-5834, 2023 12.
Article in English | MEDLINE | ID: mdl-37646174

ABSTRACT

Notch receptor activation is regulated by the intramembrane protease γ-secretase, which cleaves and liberates the Notch intracellular domain (Nicd) that regulates gene transcription. While γ-secretase cleavage is necessary, we demonstrate it is insufficient for Notch activation and requires vesicular trafficking. Here, we report Divalent metal transporter 1 (Dmt1, Slc11A2) as a novel and essential regulator of Notch signalling. Dmt1-deficient cells are defective in Notch signalling and have perturbed endolysosomal trafficking and function. Dmt1 encodes for two isoforms, with and without an iron response element (ire). We show that isoform-specific silencing of Dmt1-ire and Dmt1+ire has opposite consequences on Notch-dependent cell fates in cell lines and intestinal organoids. Loss of Dmt1-ire suppresses Notch activation and promotes differentiation, whereas loss of Dmt1+ire causes Notch activation and maintains stem-progenitor cell fates. Dmt1 isoform expression correlates with Notch and Wnt signalling in Apc-deficient intestinal organoids and human colorectal cancers. Consistently, Dmt1-ire silencing induces Notch-dependent differentiation in colorectal cancer cells. These data identify Dmt1 isoforms as binary switches controlling Notch cell fate decisions in normal and tumour cells.


Subject(s)
Amyloid Precursor Protein Secretases , Cation Transport Proteins , Iron , Humans , Amyloid Precursor Protein Secretases/metabolism , Cell Line , Iron/metabolism , Iron-Binding Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Cation Transport Proteins/genetics , Regulatory Sequences, Nucleic Acid
15.
J Proteome Res ; 22(9): 3081-3095, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37585105

ABSTRACT

In a currently 13-year-old girl of consanguineous Turkish parents, who developed unsteady gait and polyneuropathy at the ages of 3 and 6 years, respectively, we performed whole genome sequencing and identified a biallelic missense variant c.424C>T, p.R142W in glypican 1 (GPC1) as a putative disease-associated variant. Up to date, GPC1 has not been associated with a neuromuscular disorder, and we hypothesized that this variant, predicted as deleterious, may be causative for the disease. Using mass spectrometry-based proteomics, we investigated the interactome of GPC1 WT and the missense variant. We identified 198 proteins interacting with GPC1, of which 16 were altered for the missense variant. This included CANX as well as vacuolar ATPase (V-ATPase) and the mammalian target of rapamycin complex 1 (mTORC1) complex members, whose dysregulation could have a potential impact on disease severity in the patient. Importantly, these proteins are novel interaction partners of GPC1. At 10.5 years, the patient developed dilated cardiomyopathy and kyphoscoliosis, and Friedreich's ataxia (FRDA) was suspected. Given the unusually severe phenotype in a patient with FRDA carrying only 104 biallelic GAA repeat expansions in FXN, we currently speculate that disturbed GPC1 function may have exacerbated the disease phenotype. LC-MS/MS data are accessible in the ProteomeXchange Consortium (PXD040023).


Subject(s)
Friedreich Ataxia , Proteomics , Humans , Ataxia , Chromatography, Liquid , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Glypicans/metabolism , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Tandem Mass Spectrometry , Female , Adolescent
16.
J Pharmacol Exp Ther ; 386(1): 1-3, 2023 07.
Article in English | MEDLINE | ID: mdl-37321645

ABSTRACT

Divalent metal transporter 1 (DMT1) is the major importer of ferrous iron at the apical surface of enterocytes in the duodenum. Multiple groups have tried to design specific inhibitors for DMT1 both to study its contributions to iron (and metal ion) homeostasis and to provide a pharmacological means to treat iron overload disorders like hereditary hemochromatosis and thalassemias. This task faces challenges because many tissues express DMT1 and DMT1 transports other metals adding to standard risks in making specific inhibitors. Xenon Pharmaceuticals have published several papers on their efforts. Their latest paper in this issue of the journal culminates their efforts with compounds named XEN601 and XEN602 but implies that these very effective inhibitors have sufficient toxicity for them to halt development. This Viewpoint evaluates their efforts and briefly considers alternative routes to the goal. SIGNIFICANCE STATEMENT: This Viewpoint briefly reviews the paper on inhibitors of DMT1 that appears in this issue of the journal and commends the effort and research utility of those developed by Xenon. The inhibitors have proven to be valuable research tools for studying metal ion homeostasis particularly for iron. If Xenon is ceasing to try to develop them for treatment of iron overload disorders, then new alternatives need to come to the fore.


Subject(s)
Iron Overload , Humans , Iron Overload/drug therapy , Iron Overload/metabolism , Iron/metabolism , Biological Transport , Iron-Binding Proteins/metabolism , Enterocytes/metabolism
17.
Hum Mol Genet ; 32(13): 2241-2250, 2023 06 19.
Article in English | MEDLINE | ID: mdl-37027192

ABSTRACT

OBJECTIVE: In Friedreich's ataxia (FRDA), the most affected tissues are not accessible to sampling and available transcriptomic findings originate from blood-derived cells and animal models. Herein, we aimed at dissecting for the first time the pathophysiology of FRDA by means of RNA-sequencing in an affected tissue sampled in vivo. METHODS: Skeletal muscle biopsies were collected from seven FRDA patients before and after treatment with recombinant human Erythropoietin (rhuEPO) within a clinical trial. Total RNA extraction, 3'-mRNA library preparation and sequencing were performed according to standard procedures. We tested for differential gene expression with DESeq2 and performed gene set enrichment analysis with respect to control subjects. RESULTS: FRDA transcriptomes showed 1873 genes differentially expressed from controls. Two main signatures emerged: (1) a global downregulation of the mitochondrial transcriptome as well as of ribosome/translational machinery and (2) an upregulation of genes related to transcription and chromatin regulation, especially of repressor terms. Downregulation of the mitochondrial transcriptome was more profound than previously shown in other cellular systems. Furthermore, we observed in FRDA patients a marked upregulation of leptin, the master regulator of energy homeostasis. RhuEPO treatment further enhanced leptin expression. INTERPRETATION: Our findings reflect a double hit in the pathophysiology of FRDA: a transcriptional/translational issue and a profound mitochondrial failure downstream. Leptin upregulation in the skeletal muscle in FRDA may represent a compensatory mechanism of mitochondrial dysfunction, which is amenable to pharmacological boosting. Skeletal muscle transcriptomics is a valuable biomarker to monitor therapeutic interventions in FRDA.


Subject(s)
Erythropoietin , Friedreich Ataxia , Animals , Humans , Transcriptome/genetics , Leptin/genetics , Friedreich Ataxia/pathology , Erythropoietin/genetics , RNA , Muscle, Skeletal/metabolism , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism
18.
Toxicol Appl Pharmacol ; 465: 116440, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36870574

ABSTRACT

Ischemia/reperfusion- (I/R-) induced injury is unavoidable and a major risk factor for graft failure and acute rejection following kidney transplantation. However, few effective interventions are available to improve the outcome due to the complicated mechanisms and lack of appropriate therapeutic targets. Hence, this research aimed to explore the effect of the thiazolidinedione (TZD) compounds on I/R-induced kidney damage. One of the main causes of renal I/R injury is the ferroptosis of renal tubular cells. In this study, compared with the antidiabetic TZD pioglitazone (PGZ), we found its derivative mitoglitazone (MGZ) exerted significantly inhibitory effects on erastin-induced ferroptosis by suppressing mitochondrial membrane potential hyperpolarization and lipid ROS production in HEK293 cells. Moreover, MGZ pretreatment remarkably alleviated I/R-induced renal damages by inhibiting cell death and inflammation, upregulating the expression of glutathione peroxidase 4 (GPX4), and reducing iron-related lipid peroxidation in C57BL/6 N mice. Additionally, MGZ exhibited excellent protection against I/R-induced mitochondrial dysfunction by restoring ATP production, mitochondrial DNA copy numbers, and mitochondrial morphology in kidney tissues. Mechanistically, molecular docking and surface plasmon resonance experiments demonstrated that MGZ exhibited a high binding affinity with the mitochondrial outer membrane protein mitoNEET. Collectively, our findings indicated the renal protective effect of MGZ was closely linked to regulating the mitoNEET-mediated ferroptosis pathway, thus offering potential therapeutic strategies for ameliorating I/R injuries.


Subject(s)
Ferroptosis , Reperfusion Injury , Mice , Animals , Humans , HEK293 Cells , Molecular Docking Simulation , Mice, Inbred C57BL , Kidney/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Membrane Proteins/metabolism , Iron-Binding Proteins/metabolism , Iron-Binding Proteins/pharmacology
19.
Mov Disord ; 38(6): 970-977, 2023 06.
Article in English | MEDLINE | ID: mdl-36928898

ABSTRACT

BACKGROUND: Friedreich's ataxia (FRDA), most commonly caused by a GAA triplet repeat (GAA-TR) expansion in intron 1 of the FXN gene, is characterized by deficiency of frataxin protein and clinical features such as progressive ataxia, dysarthria, impaired proprioception and vibration, abolished deep tendon reflexes, Babinski sign, and vision loss in association with non-neurological features such as skeletal anomalies, hearing loss, cardiomyopathy, and diabetes. Pathogenic GAA-TRs range in size from 60 to 1500 triplets and negatively correlate with age of onset. Clinical severity is predicted by a combination of GAA-TR length and disease duration (DD) via multivariable regressions, which cannot typically be used for the small sample sizes in most studies on this rare disease. OBJECTIVE: We aimed to develop a single metric, which we call "disease burden" (DB), that encompasses both GAA-TR length and DD for predicting disease features of FRDA in small sample sizes. METHODS: Linear regression and multivariable regression analysis was used to determine correlation coefficients between different disease features of FRDA. RESULTS: Using large datasets for validation, we found that DB predicts measures of neurological dysfunction in FRDA better than GAA-TR length or DD. Analogous results were found using small datasets. CONCLUSIONS: FRDA DB is a novel metric of disease severity that has utility in small datasets to demonstrate correlations that would not otherwise be evident with either GAA-TR or DD alone. This is important for discovering new biomarkers, as well as improving the prediction of severity of disease features in FRDA. © 2023 International Parkinson and Movement Disorder Society.


Subject(s)
Friedreich Ataxia , Humans , Friedreich Ataxia/genetics , Trinucleotide Repeats , Trinucleotide Repeat Expansion/genetics , Introns , Severity of Illness Index , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism
20.
J Pharmacol Exp Ther ; 386(1): 4-14, 2023 07.
Article in English | MEDLINE | ID: mdl-36958846

ABSTRACT

Divalent metal transporter 1 (DMT1) cotransports ferrous iron and protons and is the primary mechanism for uptake of nonheme iron by enterocytes. Inhibitors are potentially useful as therapeutic agents to treat iron overload disorders such as hereditary hemochromatosis or ß-thalassemia intermedia, provided that inhibition can be restricted to the duodenum. We used a calcein quench assay to identify human DMT1 inhibitors. Dimeric compounds were made to generate more potent compounds with low systemic exposure. Direct block of DMT1 was confirmed by voltage clamp measurements. The lead compound, XEN602, strongly inhibits dietary nonheme iron uptake in both rats and pigs yet has negligible systemic exposure. Efficacy is maintained for >2 weeks in a rat subchronic dosing assay. Doses that lowered iron content in the spleen and liver by >50% had no effect on the tissue content of other divalent cations except for cobalt. XEN602 represents a powerful pharmacological tool for understanding the physiologic function of DMT1 in the gut. SIGNIFICANCE STATEMENT: This report introduces methodology to develop potent, gut-restricted inhibitors of divalent metal transporter 1 (DMT1) and identifies XEN602 as a suitable compound for in vivo studies. We also report novel animal models to quantify the inhibition of dietary uptake of iron in both rodents and pigs. This research shows that inhibition of DMT1 is a promising means to treat iron overload disorders.


Subject(s)
Iron Overload , Humans , Rats , Animals , Swine , Iron Overload/drug therapy , Iron/metabolism , Biological Transport , Iron-Binding Proteins/metabolism , Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL
...