Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 67
3.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article En | MEDLINE | ID: mdl-38731873

The supply and control of iron is essential for all cells and vital for many physiological processes. All functions and activities of iron are expressed in conjunction with iron-binding molecules. For example, natural chelators such as transferrin and chelator-iron complexes such as haem play major roles in iron metabolism and human physiology. Similarly, the mainstay treatments of the most common diseases of iron metabolism, namely iron deficiency anaemia and iron overload, involve many iron-chelator complexes and the iron-chelating drugs deferiprone (L1), deferoxamine (DF) and deferasirox. Endogenous chelators such as citric acid and glutathione and exogenous chelators such as ascorbic acid also play important roles in iron metabolism and iron homeostasis. Recent advances in the treatment of iron deficiency anaemia with effective iron complexes such as the ferric iron tri-maltol complex (feraccru or accrufer) and the effective treatment of transfusional iron overload using L1 and L1/DF combinations have decreased associated mortality and morbidity and also improved the quality of life of millions of patients. Many other chelating drugs such as ciclopirox, dexrazoxane and EDTA are used daily by millions of patients in other diseases. Similarly, many other drugs or their metabolites with iron-chelation capacity such as hydroxyurea, tetracyclines, anthracyclines and aspirin, as well as dietary molecules such as gallic acid, caffeic acid, quercetin, ellagic acid, maltol and many other phytochelators, are known to interact with iron and affect iron metabolism and related diseases. Different interactions are also observed in the presence of essential, xenobiotic, diagnostic and theranostic metal ions competing with iron. Clinical trials using L1 in Parkinson's, Alzheimer's and other neurodegenerative diseases, as well as HIV and other infections, cancer, diabetic nephropathy and anaemia of inflammation, highlight the importance of chelation therapy in many other clinical conditions. The proposed use of iron chelators for modulating ferroptosis signifies a new era in the design of new therapeutic chelation strategies in many other diseases. The introduction of artificial intelligence guidance for optimal chelation therapeutic outcomes in personalised medicine is expected to increase further the impact of chelation in medicine, as well as the survival and quality of life of millions of patients with iron metabolic disorders and also other diseases.


Iron Chelating Agents , Iron Overload , Humans , Iron Overload/drug therapy , Iron Overload/metabolism , Iron Chelating Agents/therapeutic use , Iron Chelating Agents/pharmacology , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/metabolism , Iron/metabolism , Animals , Deferiprone/therapeutic use , Deferiprone/pharmacology
4.
Adv Sci (Weinh) ; 11(17): e2400862, 2024 May.
Article En | MEDLINE | ID: mdl-38408138

Hematoma, a risk factor of implant-associated infections (IAIs), creates a Fe-rich environment following implantation, which proliferates the growth of pathogenic bacteria. Fe metabolism is a major vulnerability for pathogens and is crucial for several fundamental physiological processes. Herein, a deferiprone (DFP)-loaded layered double hydroxide (LDH)-based nanomedicine (DFP@Ga-LDH) that targets the Fe-rich environments of IAIs is reported. In response to acidic changes at the infection site, DFP@Ga-LDH systematically interferes with bacterial Fe metabolism via the substitution of Ga3+ and Fe scavenging by DFP. DFP@Ga-LDH effectively reverses the Fe/Ga ratio in Pseudomonas aeruginosa, causing comprehensive interference in various Fe-associated targets, including transcription and substance metabolism. In addition to its favorable antibacterial properties, DFP@Ga-LDH functions as a nano-adjuvant capable of delaying the emergence of antibiotic resistance. Accordingly, DFP@Ga-LDH is loaded with a siderophore antibiotic (cefiderocol, Cefi) to achieve the antibacterial nanodrug DFP@Ga-LDH-Cefi. Antimicrobial and biosafety efficacies of DFP@Ga-LDH-Cefi are validated using ex vivo human skin and mouse IAI models. The pivotal role of the hematoma-created Fe-rich environment of IAIs is highlighted, and a nanoplatform that efficiently interferes with bacterial Fe metabolism is developed. The findings of the study provide promising guidance for future research on the exploration of nano-adjuvants as antibacterial agents.


Anti-Bacterial Agents , Biofilms , Iron , Prosthesis-Related Infections , Pseudomonas aeruginosa , Biofilms/drug effects , Mice , Iron/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Pseudomonas aeruginosa/drug effects , Prosthesis-Related Infections/drug therapy , Prosthesis-Related Infections/microbiology , Deferiprone/pharmacology , Disease Models, Animal , Cefiderocol , Pseudomonas Infections/drug therapy , Humans , Nanomedicine/methods
5.
Exp Neurol ; 374: 114703, 2024 Apr.
Article En | MEDLINE | ID: mdl-38281588

Germinal matrix hemorrhage (GMH) is a devasting neurological disease in premature newborns. After GMH, brain iron overload associated with hemoglobin degradation contributed to oxidative stress, causing disruption of the already vulnerable blood-brain barrier (BBB). Mitochondrial ferritin (FTMT), a novel mitochondrial outer membrane protein, is crucial in maintaining cellular iron homeostasis. We aimed to investigate the effect of FTMT upregulation on oxidative stress and BBB disruption associated with brain iron overload in rats. A total of 222 Sprague-Dawley neonatal rat pups (7 days old) were used to establish a collagenase-induced GMH model and an iron-overload model of intracerebral FeCl2 injection. Deferiprone was administered via gastric lavage 1 h after GMH and given daily until euthanasia. FTMT CRISPR Knockout and adenovirus (Ad)-FTMT were administered intracerebroventricularly 48 h before GMH and FeCl2 injection, respectively. Neurobehavioral tests, immunofluorescence, Western blot, Malondialdehyde measurement, and brain water content were performed to evaluate neurobehavior deficits, oxidative stress, and BBB disruption, respectively. The results demonstrated that brain expressions of iron exporter Ferroportin (FPN) and antioxidant glutathione peroxidase 4 (GPX4) as well as BBB tight junction proteins including Claudin-5 and Zona Occulta (ZO)-1 were found to be decreased at 72 h after GMH. FTMT agonist Deferiprone attenuated oxidative stress and preserved BBB tight junction proteins after GMH. These effects were partially reversed by FTMT CRISPR Knockout. Iron overload by FeCl2 injection resulted in oxidative stress and BBB disruption, which were improved by Ad-FTMT mediated FTMT overexpression. Collectively, FTMT upregulation is neuroprotective against brain injury associated with iron overload. Deferiprone reduced oxidative stress and BBB disruption by maintaining cellular iron homeostasis partially by the upregulating of FTMT after GMH. Deferiprone may be an effective treatment for patients with GMH.


Blood-Brain Barrier , Iron Overload , Humans , Infant, Newborn , Rats , Animals , Blood-Brain Barrier/metabolism , Animals, Newborn , Rats, Sprague-Dawley , Up-Regulation , Deferiprone/metabolism , Deferiprone/pharmacology , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/metabolism , Oxidative Stress , Iron/metabolism , Iron Overload/metabolism , Homeostasis , Ferritins/metabolism , Tight Junction Proteins/metabolism
6.
Lett Appl Microbiol ; 76(10)2023 Oct 04.
Article En | MEDLINE | ID: mdl-37791895

This study evaluated the antibiofilm activity of promethazine, deferiprone, and Manuka honey against Staphylococcus aureus and Pseudomonas aeruginosa in vitro and ex vivo in a wound model on porcine skin. The minimum inhibitory concentrations (MICs) and the effects of the compounds on biofilms were evaluated. Then, counting colony-forming units (CFUs) and confocal microscopy were performed on biofilms cultivated on porcine skin for evaluation of the compounds. For promethazine, MICs ranging from 97.66 to 781.25 µg/ml and minimum biofilm eradication concentration (MBEC) values ranging from 195.31 to 1562.5 µg/ml were found. In addition to reducing the biomass of both species' biofilms. As for deferiprone, the MICs were 512 and >1024 µg/ml, the MBECs were ≥1024 µg/ml, and it reduced the biomass of biofilms. Manuka honey had MICs of 10%-40%, MBECs of 20 to >40% and reduced the biomass of S. aureus biofilms only. Concerning the analyses in the ex vivo model, the compounds reduced (P < .05) CFU counts for both bacterial species, altering the biofilm architecture. The action of the compounds on biofilms in in vitro and ex vivo tests raises the possibility of using them against biofilm-associated wounds. However, further studies are needed to characterize the mechanisms of action and their effectiveness on biofilms in vivo.


Honey , Staphylococcus aureus , Animals , Swine , Promethazine/pharmacology , Deferiprone/pharmacology , Biofilms , Pseudomonas aeruginosa , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests
7.
Tuberculosis (Edinb) ; 142: 102390, 2023 09.
Article En | MEDLINE | ID: mdl-37506532

Non-Tuberculous Mycobacterial Pulmonary Disease (NTM-PD) caused by Mycobacterium abscessus is a frequent complication in patients with cystic fibrosis (CF) that worsens lung function over time. Currently, there is no cure for NTM-PD, hence new therapies are urgently required. Disrupting bacterial iron uptake pathways using gallium-protoporphyrin (IX) (GaPP), a heme analog, has been proposed as a novel antibacterial approach to tackle multi-drug resistant M. abscessus. However, the antibacterial activity of GaPP has been tested only in iron-deficient media, which cannot accurately mirror the potential activity in vivo. Herein, we investigated the potential synergistic activity between GaPP and the iron-chelating agent deferiprone (Def) in regular media against M. abscessus-infected macrophages. The safety of the treatment was assessed in vitro using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay in Nuli-1 and THP-1 cell lines. Def-GaPP had synergistic activity against M. abscessus-infected macrophages where 10 mM-12.5 mg/L of Def-GaPP reduced the viability by up to 0.9 log10. Furthermore, Def-GaPP showed no cytotoxicity to Nuli-1 and THP-1 cell lines at the effective antibacterial concentrations (10 mM-12.5 mg/L) of Def- GaPP. These data encourage future investigation of Def-GaPP as a novel antimicrobial against NTM-PD.


Anti-Bacterial Agents , Deferiprone , Gallium , Iron Chelating Agents , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Protoporphyrins , Deferiprone/pharmacology , Gallium/pharmacology , Protoporphyrins/pharmacology , Humans , Mycobacterium abscessus/drug effects , Mycobacterium Infections, Nontuberculous/microbiology , Anti-Bacterial Agents/pharmacology , Iron Chelating Agents/pharmacology , THP-1 Cells , Drug Synergism
8.
PeerJ ; 11: e15187, 2023.
Article En | MEDLINE | ID: mdl-37131988

Background: The spread of artemisinin (ART)-resistant Plasmodium falciparum threatens the control of malaria. Mutations in the propeller domains of P. falciparum Kelch13 (k13) are strongly associated with ART resistance. Ferredoxin (Fd), a component of the ferredoxin/NADP+ reductase (Fd/FNR) redox system, is essential for isoprenoid precursor synthesis in the plasmodial apicoplast, which is important for K13-dependent hemoglobin trafficking and ART activation. Therefore, Fd is an antimalarial drug target and fd mutations may modulate ART sensitivity. We hypothesized that loss of Fd/FNR function enhances the effect of k13 mutation on ART resistance. Methods: In this study, methoxyamino chalcone (C3), an antimalarial compound that has been reported to inhibit the interaction of recombinant Fd and FNR proteins, was used as a chemical inhibitor of the Fd/FNR redox system. We investigated the inhibitory effects of dihydroartemisinin (DHA), C3, and iron chelators including deferiprone (DFP), 1-(N-acetyl-6-aminohexyl)-3-hydroxy-2-methylpyridin-4-one (CM1) and deferiprone-resveratrol hybrid (DFP-RVT) against wild-type (WT), k13 mutant, fd mutant, and k13 fd double mutant P. falciparum parasites. Furthermore, we investigated the pharmacological interaction of C3 with DHA, in which the iron chelators were used as reference ART antagonists. Results: C3 showed antimalarial potency similar to that of the iron chelators. As expected, combining DHA with C3 or iron chelators exhibited a moderately antagonistic effect. No differences were observed among the mutant parasites with respect to their sensitivity to C3, iron chelators, or the interactions of these compounds with DHA. Discussion: The data suggest that inhibitors of the Fd/FNR redox system should be avoided as ART partner drugs in ART combination therapy for treating malaria.


Antimalarials , Chalcone , Malaria, Falciparum , Humans , Antimalarials/pharmacology , Plasmodium falciparum/genetics , Ferredoxins/chemistry , Chalcone/pharmacology , Deferiprone/pharmacology , Malaria, Falciparum/drug therapy , Ferredoxin-NADP Reductase , Iron Chelating Agents/pharmacology
9.
Biofouling ; 39(2): 135-144, 2023 02.
Article En | MEDLINE | ID: mdl-37013808

This study evaluated the effect of the iron chelator deferiprone (DFP) on antimicrobial susceptibility and biofilm formation and maintenance by Burkholderia pseudomallei. Planktonic susceptibility to DFP alone and in combination with antibiotics was evaluated by broth microdilution and biofilm metabolic activity was determined with resazurin. DFP minimum inhibitory concentration (MIC) range was 4-64 µg/mL and in combination reduced the MIC for amoxicillin/clavulanate and meropenem. DFP reduced the biomass of biofilms by 21 and 12% at MIC and MIC/2, respectively. As for mature biofilms, DFP reduced the biomass by 47%, 59%, 52% and 30% at 512, 256, 128 and 64 µg/mL, respectively, but did not affect B. pseudomallei biofilm viability nor increased biofilm susceptibility to amoxicillin/clavulanate, meropenem and doxycycline. DFP inhibits planktonic growth and potentiates the effect of ß-lactams against B. pseudomallei in the planktonic state and reduces biofilm formation and the biomass of B. pseudomallei biofilms.


Burkholderia pseudomallei , Meropenem/pharmacology , Deferiprone/pharmacology , Iron/pharmacology , Iron/metabolism , Biofilms , Anti-Bacterial Agents/pharmacology , Amoxicillin-Potassium Clavulanate Combination/pharmacology , Microbial Sensitivity Tests , Iron Chelating Agents/pharmacology
10.
Sci Rep ; 13(1): 6960, 2023 04 28.
Article En | MEDLINE | ID: mdl-37117329

Iron, supplemented as iron-loaded transferrin (holotransferrin), is an essential nutrient in mammalian cell cultures, particularly for erythroid cultures. The high cost of human transferrin represents a challenge for large scale production of red blood cells (RBCs) and for cell therapies in general. We evaluated the use of deferiprone, a cell membrane-permeable drug for iron chelation therapy, as an iron carrier for erythroid cultures. Iron-loaded deferiprone (Def3·Fe3+, at 52 µmol/L) could eliminate the need for holotransferrin supplementation during in vitro expansion and differentiation of erythroblast cultures to produce large numbers of enucleated RBC. Only the first stage, when hematopoietic stem cells committed to erythroblasts, required holotransferrin supplementation. RBCs cultured in presence of Def3·Fe3+ or holotransferrin (1000 µg/mL) were similar with respect to differentiation kinetics, expression of cell-surface markers CD235a and CD49d, hemoglobin content, and oxygen association/dissociation. Replacement of holotransferrin supplementation by Def3·Fe3+ was also successful in cultures of myeloid cell lines (MOLM13, NB4, EOL1, K562, HL60, ML2). Thus, iron-loaded deferiprone can partially replace holotransferrin as a supplement in chemically defined cell culture medium. This holds promise for a significant decrease in medium cost and improved economic perspectives of the large scale production of red blood cells for transfusion purposes.


Erythrocytes , Iron , Animals , Humans , Iron/metabolism , Deferiprone/pharmacology , Erythrocytes/metabolism , Iron Chelating Agents/therapeutic use , Hemoglobins/metabolism , Cells, Cultured , Mammals/metabolism
12.
Yeast ; 40(3-4): 143-151, 2023 03.
Article En | MEDLINE | ID: mdl-36755518

In the absence of YFH1, the yeast ortholog of the human FXN gene, budding yeast Saccharomyces cerevisiae experience similar problems to those of cells with Friedreich's ataxia (FRDA). The comparable phenotypic traits consist of impaired respiration, problems in iron homeostasis, decreased oxidative stress tolerance, and diminished iron-sulfur cluster synthesis, rendering yeast of potential use in FRDA modeling and drug trials. Deferiprone, an iron chelator, is one of the long-term studied potential drugs for FRDA, whereas metformin is a biguanide prescribed to treat type 2 diabetes. In the present study, the effects of deferiprone and metformin treatment on the yeast FRDA model are explored via RNA-sequencing analyses. The comparative inquiry of transcriptome data reveals new promising roles for metformin in FRDA treatment since deferiprone and metformin treatments produce overlapping transcriptional and phenotypic responses in YFH1Δ cells. The results revealed that both deferiprone and metformin treatment does not rescue aerobic respiration in YFH1Δ cells, but they alleviate the FRDA phenotype probably by triggering the retrograde mitochondria-to-nucleus signaling.


Diabetes Mellitus, Type 2 , Friedreich Ataxia , Metformin , Humans , Saccharomyces cerevisiae/genetics , Deferiprone/pharmacology , Deferiprone/therapeutic use , Friedreich Ataxia/drug therapy , Friedreich Ataxia/genetics , Metformin/pharmacology , Metformin/therapeutic use , Iron
13.
J Trace Elem Med Biol ; 76: 127113, 2023 Mar.
Article En | MEDLINE | ID: mdl-36516571

BACKGROUND: Aluminium (Al) overload has toxic effects on multiple organ systems, especially the nervous system. Al accumulation in the brain, especially the hippocampus, is an important factor contributing to Alzheimer's disease (AD). Deferiprone (DFP), a metal chelator, is used as a potential treatment for AD. In this study, we investigated the combined effect of taurine and DFP on Al chelation and hippocampal apoptosis in Al-exposed rats, as well as the underlying mechanisms of these effects to explore a possible therapy for AD. METHODS: Male Wistar rats were divided into seven groups: negative control group (administered saline), Al-exposure group (administered AlCl3 and saline), and five experimental groups (administered AlCl3 and taurine, varying doses of DFP, or taurine with varying doses of DFP). After 8 weeks of treatment, the rats were sacrificed, and the terminal deoxyribonucleotidyl transferase (TDT)-mediated dUTP-digoxigenin nick end labelling (TUNEL) assay was used to detect hippocampal apoptotic cells. Real-time quantitative PCR was used to assess the expression of the Bcl2 and Bax genes, and a western blotting assay was used to evaluate BCL2, BAX, and cleaved caspase-3 levels. RESULTS: Compared to the negative control group, the number of apoptotic cells in the hippocampus increased, Bcl2 expression significantly decreased, and BAX and cleaved caspase-3 levels increased in the Al-exposure group. The combination of taurine and DFP exerted a protective effect by inhibiting hippocampal cell apoptosis through the BCL2, BAX, and caspase-3 signalling pathways. Compared with the taurine-administered group, the group administered taurine with DFP showed a significantly increased Bcl2 and decreased Bax expression. CONCLUSION: The combination of taurine and DFP is a potential candidate for the treatment of AD induced by Al exposure.


Aluminum , Alzheimer Disease , Rats , Animals , Male , Deferiprone/pharmacology , Aluminum/metabolism , Caspase 3/metabolism , Rats, Wistar , bcl-2-Associated X Protein/metabolism , Taurine/pharmacology , Taurine/metabolism , Hippocampus , Apoptosis , Chelating Agents/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Alzheimer Disease/metabolism
14.
Cell Death Differ ; 30(1): 69-81, 2023 01.
Article En | MEDLINE | ID: mdl-35933500

Glaucoma can result in retinal ganglion cell (RGC) death and permanently damaged vision. Pathologically high intraocular pressure (ph-IOP) is the leading cause of damaged vision during glaucoma; however, controlling ph-IOP alone does not entirely prevent the loss of glaucomatous RGCs, and the underlying mechanism remains elusive. In this study, we reported an increase in ferric iron in patients with acute primary angle-closure glaucoma (the most typical glaucoma with ph-IOP damage) compared with the average population by analyzing free iron levels in peripheral serum. Thus, iron metabolism might be involved in regulating the injury of RGCs under ph-IOP. In vitro and in vivo studies confirmed that ph-IOP led to abnormal accumulation of ferrous iron in cells and retinas at 1-8 h post-injury and elevation of ferric iron in serum at 8 h post-injury. Nuclear receptor coactivator 4 (NCOA4)-mediated degradation of ferritin heavy polypeptide 1(FTH1) is essential to disrupt iron metabolism in the retina after ph-IOP injury. Furthermore, knockdown of Ncoa4 in vivo inhibited FTH1 degradation and reduced the retinal ferrous iron level. Elevated ferrous iron induced by ph-IOP led to a marked accumulation of pro-ferroptotic factors (lipid peroxidation and acyl CoA synthetase long-chain family member 4) and a depletion of anti-ferroptotic factors (glutathione, glutathione peroxidase 4, and nicotinamide adenine dinucleotide phosphate). These biochemical changes resulted in RGC ferroptosis. Deferiprone can pass through the blood-retinal barrier after oral administration and chelated abnormally elevated ferrous iron in the retina after ph-IOP injury, thus inhibiting RGC ferroptosis and protecting visual function. In conclusion, this study revealed the role of NCOA4-FTH1-mediated disturbance of iron metabolism and ferroptosis in RGCs during glaucoma. We demonstrate the protective effect of Deferiprone on RGCs via inhibition of ferroptosis, providing a research direction to understand and treat glaucoma via the iron homeostasis and ferroptosis pathways.


Ferroptosis , Glaucoma , Humans , Animals , Retinal Ganglion Cells/metabolism , Intraocular Pressure , Deferiprone/pharmacology , Deferiprone/metabolism , Glaucoma/metabolism , Homeostasis , Iron/metabolism , Disease Models, Animal
15.
Br J Pharmacol ; 180(2): 214-234, 2023 01.
Article En | MEDLINE | ID: mdl-36102035

BACKGROUND AND PURPOSE: Traumatic brain injury (TBI) remains a leading cause of mortality and morbidity in young adults. The role of iron in potentiating neurodegeneration following TBI has gained recent interest as iron deposition has been detected in the injured brain in the weeks to months post-TBI, in both the preclinical and clinical setting. A failure in iron homeostasis can lead to oxidative stress, inflammation and excitotoxicity; and whether this is a cause or consequence of the long-term effects of TBI remains unknown. EXPERIMENTAL APPROACH: We investigated the role of iron and the effect of therapeutic intervention using a brain-permeable iron chelator, deferiprone, in a controlled cortical impact mouse model of TBI. An extensive assessment of cognitive, motor and anxiety/depressive outcome measures were examined, and neuropathological and biochemical changes, over a 3-month period post-TBI. KEY RESULTS: Lesion volume was significantly reduced at 3 months, which was preceded by a reduction in astrogliosis, microglia/macrophages and preservation of neurons in the injured brain at 2 weeks and/or 1 month post-TBI in mice receiving oral deferiprone. Deferiprone treatment showed significant improvements in neurological severity scores, locomotor/gait performance and cognitive function, and attenuated anxiety-like symptoms post-TBI. Deferiprone reduced iron levels, lipid peroxidation/oxidative stress and altered expression of neurotrophins in the injured brain over this period. CONCLUSION AND IMPLICATIONS: Our findings support a detrimental role of iron in the injured brain and suggest that deferiprone (or similar iron chelators) may be promising therapeutic approaches to improve survival, functional outcomes and quality of life following TBI.


Brain Injuries, Traumatic , Quality of Life , Animals , Mice , Deferiprone/pharmacology , Deferiprone/therapeutic use , Mice, Inbred C57BL , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Iron
16.
N Engl J Med ; 387(22): 2045-2055, 2022 12 01.
Article En | MEDLINE | ID: mdl-36449420

BACKGROUND: Iron content is increased in the substantia nigra of persons with Parkinson's disease and may contribute to the pathophysiology of the disorder. Early research suggests that the iron chelator deferiprone can reduce nigrostriatal iron content in persons with Parkinson's disease, but its effects on disease progression are unclear. METHODS: We conducted a multicenter, phase 2, randomized, double-blind trial involving participants with newly diagnosed Parkinson's disease who had never received levodopa. Participants were assigned (in a 1:1 ratio) to receive oral deferiprone at a dose of 15 mg per kilogram of body weight twice daily or matched placebo for 36 weeks. Dopaminergic therapy was withheld unless deemed necessary for symptom control. The primary outcome was the change in the total score on the Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS; range, 0 to 260, with higher scores indicating more severe impairment) at 36 weeks. Secondary and exploratory clinical outcomes at up to 40 weeks included measures of motor and nonmotor disability. Brain iron content measured with the use of magnetic resonance imaging was also an exploratory outcome. RESULTS: A total of 372 participants were enrolled; 186 were assigned to receive deferiprone and 186 to receive placebo. Progression of symptoms led to the initiation of dopaminergic therapy in 22.0% of the participants in the deferiprone group and 2.7% of those in the placebo group. The mean MDS-UPDRS total score at baseline was 34.3 in the deferiprone group and 33.2 in the placebo group and increased (worsened) by 15.6 points and 6.3 points, respectively (difference, 9.3 points; 95% confidence interval, 6.3 to 12.2; P<0.001). Nigrostriatal iron content decreased more in the deferiprone group than in the placebo group. The main serious adverse events with deferiprone were agranulocytosis in 2 participants and neutropenia in 3 participants. CONCLUSIONS: In participants with early Parkinson's disease who had never received levodopa and in whom treatment with dopaminergic medications was not planned, deferiprone was associated with worse scores in measures of parkinsonism than those with placebo over a period of 36 weeks. (Funded by the European Union Horizon 2020 program; FAIRPARK-II ClinicalTrials.gov number, NCT02655315.).


Antiparkinson Agents , Deferiprone , Iron Chelating Agents , Iron , Parkinson Disease , Substantia Nigra , Humans , Deferiprone/administration & dosage , Deferiprone/adverse effects , Deferiprone/pharmacology , Deferiprone/therapeutic use , Iron/analysis , Iron/metabolism , Levodopa/therapeutic use , Neutropenia/chemically induced , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/adverse effects , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Substantia Nigra/chemistry , Substantia Nigra/diagnostic imaging , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Disease Progression , Double-Blind Method , Administration, Oral , Brain/diagnostic imaging , Brain Chemistry , Dopamine Agents/administration & dosage , Dopamine Agents/adverse effects , Dopamine Agents/pharmacology , Dopamine Agents/therapeutic use , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/adverse effects , Antiparkinson Agents/pharmacology , Antiparkinson Agents/therapeutic use
17.
Sci Rep ; 12(1): 19630, 2022 11 16.
Article En | MEDLINE | ID: mdl-36385152

Multiple sclerosis (MS) is a chronic inflammatory disease, which leads to focal demyelination in the brain and spinal cord. Studies showed that iron released during the course of myelin breakdown exacerbates tissue damage, which is in agreement with the features of iron-dependent cell death, ferroptosis. Here, we aimed to investigate the possible contribution of ferroptosis in the demyelinated optic nerve, and to explore the effectiveness of ferroptosis inhibitor, deferiprone (DFP), on the extent of demyelination, inflammation and axonal damage. For this purpose, focal demyelination was induced by injection of lysolecithin (LPC), into the optic nerve of male C57BL/6J mice. Afterward, optic nerves were harvested at different time points from as early as 6 h up to 7 days post-LPC injection. Next, to evaluate the effectiveness of DFP two groups of animals received daily intraperitoneal injection of DFP for 3 or 7 continuous days. Vehicle groups received saline. Iron deposition was observed at different time points post-LPC injection from 6 h to 7 days post injection. Examining ferroptosis markers showed a significant reduction in glutathione content along with increased level of malondialdehyde and upregulated ferroptosis marker genes at early time points after injection. Besides, DFP treatment during the inflammatory phase of the model resulted in decreased microgliosis and inflammation. Reduced demyelination, microgliosis and astrogliosis was shown in mice that received DFP for 7 days. Moreover, DFP protected against axonal damage and retinal ganglion cells loss. Our results suggest the possible contribution of ferroptosis pathway in the process of demyelination. The therapeutic strategies targeting iron deposition, e.g. DFP treatment might thus represent a promising therapeutic target for patients with MS.


Ferroptosis , Multiple Sclerosis , Animals , Male , Mice , Deferiprone/pharmacology , Inflammation , Iron/metabolism , Mice, Inbred C57BL , Myelin Sheath/metabolism , Neuroprotection , Optic Nerve/metabolism
18.
Life Sci ; 307: 120871, 2022 Oct 15.
Article En | MEDLINE | ID: mdl-35952729

AIMS: This study aimed to investigate the changes in gut microbiota in iron-overload thalassemia and the roles of an iron chelator on gut dysbiosis/inflammation, and metabolites, including short-chain fatty acids (SCFAs) and trimethylamine N-oxide (TMAO). MAIN METHODS: Adult male C57BL/6 mice both wild-type (WT: n = 15) and heterozygous ß-thalassemia (BKO: n = 15) were fed on either a normal (ND: n = 5/group) or a high­iron diet for four months (HFe: n = 10/group). HFe-treated WT and HFe-treated BKO groups were further subdivided into two subgroups and each subgroup given either vehicle (n = 5/subgroup) or deferiprone (n = 5/subgroup) during the last month. Gut microbiota profiles, gut barrier characteristics, levels of proinflammatory cytokines, and plasma SCFAs and TMAO were determined at the end of the study. KEY FINDINGS: HFe-fed WT mice showed distinct gut microbiota profiles from those of ND-fed WT mice, whereas HFe-fed BKO mice showed slightly different gut microbiota profiles from ND-fed BKO. Gut inflammation and barrier disruption were found only in HFe-fed BKO mice, however, an increase in plasma TMAO levels and decreased levels of SCFAs were observed in both WT and BKO mice with HFe-feeding. Treatment with deferiprone, gut dysbiosis and disturbance of metabolites were attenuated in HFe-fed WT mice, but not in HFe-fed BKO mice. Increased Verrucomicrobia and Ruminococcaceae were associated with the beneficial effects of deferiprone. SIGNIFICANCE: Iron-overload leads to gut dysbiosis/inflammation and disturbance of metabolites, and deferiprone alleviates those conditions more effectively in WT than in those that are thalassemic.


Gastrointestinal Microbiome , Iron Overload , Thalassemia , Animals , Cytokines/therapeutic use , Deferiprone/pharmacology , Diet , Dysbiosis/drug therapy , Inflammation/drug therapy , Iron/metabolism , Iron Chelating Agents/pharmacology , Iron Overload/complications , Male , Methylamines , Mice , Mice, Inbred C57BL
19.
J Trace Elem Med Biol ; 74: 127062, 2022 Dec.
Article En | MEDLINE | ID: mdl-35985070

INTRODUCTION: Cadmium (Cd) is а hazardous multi-organ toxin. In this study, we provide the first results about the effect of oral administration of deferiprone (DFP) on Cd accumulation and on the homeostasis of essential elements in the brain of Cd-exposed mice. METHODS: Adult Institute of Cancer Research (ICR) male mice were randomized into four experimental groups: untreated controls - administered distilled water for 28 days; Cd-exposed group - exposed to 18 mg/kg body weight (b.w.) Cd(II) acetate for 14 days followed by the administration of distilled water for two weeks; Cd + DFP (low dose) - Cd-intoxicated mice subsequently treated with 19 mg/kg b.w. DFP for two weeks; and Cd + DFP (high dose) - Cd-exposed mice administered high-dose DFP (135 mg/kg b.w.) for 14 days. Brains were subjected to inductively coupled plasma-mass spectrometry (ICP-MS) and histological analysis. RESULTS: The results revealed that exposure of mice to Cd for 14 days significantly increased Cd concentration and significantly decreased magnesium (Mg), phosphorus (P), and zinc (Zn) contents in the brain compared to untreated controls. This effect was accompanied by necrotic-degenerative changes in both the cerebrum and cerebellum. Oral administration of low-dose DFP to Cd-exposed mice decreased the concentration of the toxic metal in the brain by 16.37% and restored the concentration of the essential elements to normal control values. Histological analysis revealed substantially improved cerebral and cerebellar histoarchitectures. In contrast, oral administration of high-dose DFP increased Cd content and significantly decreased selenium (Se) concentration in the brain. Necrotic neurons and Purkinje cells were still observed in the cerebral and cerebellar cortices. CONCLUSION: The results demonstrated that oral administration of DFP at low doses has a better therapeutic potential for the treatment of Cd-induced brain damage compared to high doses.


Drinking Water , Selenium , Animals , Male , Mice , Acetates/pharmacology , Brain , Cadmium , Deferiprone/pharmacology , Homeostasis , Magnesium/pharmacology , Phosphorus , Selenium/pharmacology , Zinc/pharmacology
20.
Sci Rep ; 12(1): 9765, 2022 06 13.
Article En | MEDLINE | ID: mdl-35697900

Labile redox-active iron ions have been implicated in various neurodegenerative disorders, including the Parkinson's disease (PD). Iron chelation has been successfully used in clinical practice to manage iron overload in diseases such as thalassemia major; however, the use of conventional iron chelators in pathological states without systemic iron overload remains at the preclinical investigative level and is complicated by the risk of adverse outcomes due to systemic iron depletion. In this study, we examined three clinically-used chelators, namely, desferrioxamine, deferiprone and deferasirox and compared them with experimental agent salicylaldehyde isonicotinoyl hydrazone (SIH) and its boronate-masked prochelator BSIH for protection of differentiated PC12 cells against the toxicity of catecholamines 6-hydroxydopamine and dopamine and their oxidation products. All the assayed chelating agents were able to significantly reduce the catecholamine toxicity in a dose-dependent manner. Whereas hydrophilic chelator desferrioxamine exerted protection only at high and clinically unachievable concentrations, deferiprone and deferasirox significantly reduced the catecholamine neurotoxicity at concentrations that are within their plasma levels following standard dosage. SIH was the most effective iron chelator to protect the cells with the lowest own toxicity of all the assayed conventional chelators. This favorable feature was even more pronounced in prochelator BSIH that does not chelate iron unless its protective group is cleaved in disease-specific oxidative stress conditions. Hence, this study demonstrated that while iron chelation may have general neuroprotective potential against catecholamine auto-oxidation and toxicity, SIH and BSIH represent promising lead molecules and warrant further studies in more complex animal models.


Iron Chelating Agents , Iron Overload , Animals , Catecholamines/pharmacology , Deferasirox/pharmacology , Deferiprone/pharmacology , Deferoxamine/pharmacology , Dopamine/pharmacology , Iron/pharmacology , Iron Chelating Agents/pharmacology , Oxidative Stress , Oxidopamine/pharmacology , PC12 Cells , Rats
...