Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.661
Filtrar
1.
Int J Mol Sci ; 25(9)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38731873

RESUMEN

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.


Asunto(s)
Quelantes del Hierro , Sobrecarga de Hierro , Humanos , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/metabolismo , Quelantes del Hierro/uso terapéutico , Quelantes del Hierro/farmacología , Anemia Ferropénica/tratamiento farmacológico , Anemia Ferropénica/metabolismo , Hierro/metabolismo , Animales , Deferiprona/uso terapéutico , Deferiprona/farmacología
2.
J Pharmacol Sci ; 155(2): 44-51, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38677785

RESUMEN

Subretinal hemorrhages result in poor vision and visual field defects. During hemorrhage, several potentially toxic substances are released from iron-based hemoglobin and hemin, inducing cellular damage, the detailed mechanisms of which remain unknown. We examined the effects of excess intracellular iron on retinal pigment epithelial (RPE) cells. A Fe2+ probe, SiRhoNox-1 was used to investigate Fe2+ accumulation after treatment with hemoglobin or hemin in the human RPE cell line ARPE-19. We also evaluated the production of reactive oxygen species (ROS) and lipid peroxidation. Furthermore, the protective effect of-an iron chelator, 2,2'-bipyridyl (BP), and ferrostatin-1 (Fer-1) on the cell damage, was evaluated. Fe2+ accumulation increased in the hemoglobin- or hemin-treated groups, as well as intracellular ROS production and lipid peroxidation. In contrast, BP treatment suppressed RPE cell death, ROS production, and lipid peroxidation. Pretreatment with Fer-1 ameliorated cell death in a concentration-dependent manner and suppressed ROS production and lipid peroxidation. Taken together, these findings indicate that hemoglobin and hemin, as well as subretinal hemorrhage, may induce RPE cell damage and visual dysfunction via intracellular iron accumulation.


Asunto(s)
Hemina , Hemoglobinas , Hierro , Peroxidación de Lípido , Especies Reactivas de Oxígeno , Epitelio Pigmentado de la Retina , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/patología , Hemina/farmacología , Humanos , Hierro/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Hemoglobinas/metabolismo , Línea Celular , Quelantes del Hierro/farmacología , Ciclohexilaminas/farmacología , Fenilendiaminas/farmacología , Muerte Celular/efectos de los fármacos
3.
Eur J Med Res ; 29(1): 253, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38659000

RESUMEN

The progression of heart failure (HF) is complex and involves multiple regulatory pathways. Iron ions play a crucial supportive role as a cofactor for important proteins such as hemoglobin, myoglobin, oxidative respiratory chain, and DNA synthetase, in the myocardial energy metabolism process. In recent years, numerous studies have shown that HF is associated with iron dysmetabolism, and deficiencies in iron and overload of iron can both lead to the development of various myocarditis diseases, which ultimately progress to HF. Iron toxicity and iron metabolism may be key targets for the diagnosis, treatment, and prevention of HF. Some iron chelators (such as desferrioxamine), antioxidants (such as ascorbate), Fer-1, and molecules that regulate iron levels (such as lactoferrin) have been shown to be effective in treating HF and protecting the myocardium in multiple studies. Additionally, certain natural compounds can play a significant role by mediating the imbalance of iron-related signaling pathways and expression levels. Therefore, this review not only summarizes the basic processes of iron metabolism in the body and the mechanisms by which they play a role in HF, with the aim of providing new clues and considerations for the treatment of HF, but also summarizes recent studies on natural chemical components that involve ferroptosis and its role in HF pathology, as well as the mechanisms by which naturally occurring products regulate ferroptosis in HF, with the aim of providing reference information for the development of new ferroptosis inhibitors and lead compounds for the treatment of HF in the future.


Asunto(s)
Productos Biológicos , Insuficiencia Cardíaca , Hierro , Humanos , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Hierro/metabolismo , Productos Biológicos/uso terapéutico , Productos Biológicos/farmacología , Animales , Ferroptosis/efectos de los fármacos , Quelantes del Hierro/uso terapéutico , Quelantes del Hierro/farmacología , Antioxidantes/uso terapéutico
4.
Neurochem Int ; 176: 105725, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38561151

RESUMEN

Epilepsy constitutes a global health concern, affecting millions of individuals and approximately one-third of patients exhibit drug resistance. Recent investigations have revealed alterations in cerebral iron content in both epilepsy patients and animal models. However, the extant literature lacks a comprehensive exploration into the ramifications of modulating iron homeostasis as an intervention in epilepsy. This study investigated the impact of deferasirox, a iron ion chelator, on epilepsy. This study unequivocally substantiated the antiepileptic efficacy of deferasirox in a kainic acid-induced epilepsy model. Furthermore, deferasirox administration mitigated seizure susceptibility in a pentylenetetrazol-induced kindling model. Conversely, the augmentation of iron levels through supplementation has emerged as a potential exacerbating factor in the precipitating onset of epilepsy. Intriguingly, our investigation revealed a hitherto unreported discovery: ITPRIP was identified as a pivotal modulator of excitatory synaptic transmission, regulating seizures in response to deferasirox treatment. In summary, our findings indicate that deferasirox exerts its antiepileptic effects through the precise targeting of ITPRIP and amelioration of cerebral iron homeostasis, suggesting that deferasirox is a promising and novel therapeutic avenue for interventions in epilepsy.


Asunto(s)
Anticonvulsivantes , Encéfalo , Deferasirox , Epilepsia , Homeostasis , Quelantes del Hierro , Hierro , Deferasirox/farmacología , Hierro/metabolismo , Animales , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Epilepsia/tratamiento farmacológico , Epilepsia/metabolismo , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Masculino , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Ratones , Excitación Neurológica/efectos de los fármacos , Pentilenotetrazol/toxicidad , Ratas Sprague-Dawley
5.
Int J Mol Sci ; 25(8)2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38673849

RESUMEN

In this short review we have presented and discussed studies on pharmacogenomics (also termed pharmacogenetics) of the drugs employed in the treatment of ß-thalassemia or Sickle-cell disease (SCD). This field of investigation is relevant, since it is expected to help clinicians select the appropriate drug and the correct dosage for each patient. We first discussed the search for DNA polymorphisms associated with a high expression of γ-globin genes and identified this using GWAS studies and CRISPR-based gene editing approaches. We then presented validated DNA polymorphisms associated with a high HbF production (including, but not limited to the HBG2 XmnI polymorphism and those related to the BCL11A, MYB, KLF-1, and LYAR genes). The expression of microRNAs involved in the regulation of γ-globin genes was also presented in the context of pharmacomiRNomics. Then, the pharmacogenomics of validated fetal hemoglobin inducers (hydroxyurea, butyrate and butyrate analogues, thalidomide, and sirolimus), of iron chelators, and of analgesics in the pain management of SCD patients were considered. Finally, we discuss current clinical trials, as well as international research networks focusing on clinical issues related to pharmacogenomics in hematological diseases.


Asunto(s)
Anemia de Células Falciformes , Farmacogenética , Talasemia beta , Humanos , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/tratamiento farmacológico , Talasemia beta/genética , Talasemia beta/tratamiento farmacológico , Farmacogenética/métodos , Hemoglobina Fetal/genética , gamma-Globinas/genética , Quelantes del Hierro/uso terapéutico , Quelantes del Hierro/farmacología
6.
J Vis Exp ; (204)2024 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-38465932

RESUMEN

Cancer cells require large amounts of iron to maintain their proliferation. Iron metabolism is considered a hallmark of cancer, making iron a valid target for anti-cancer approaches. The development of novel compounds and the identification of leads for further modification requires that proof of mechanism assays be carried out. There are many assays to evaluate the impact on proliferation; however, the ability to chelate iron is an important and sometimes overlooked end-point measure due to the high costs of equipment and the challenge to quickly and reproducibly quantify the strength of chelation. Here, we describe a quantifiable and inexpensive cell-free fluorescent method to confirm the ability of novel compounds to chelate iron. Our assay relies on the commercially available inexpensive fluorescent dye Calcein, whose fluorescence can be quantified on most fluorescent microtiter plate readers. Calcein is a weak iron chelator, and its fluorescence is quenched when it binds Fe2+/3+; fluorescence is restored when a novel chelator outcompetes Calcein for bound Fe2+/3+. The removal of fluorescent quenching and the resulting increase in fluorescence allows the chelation ability of a novel putative chelator to be determined. Therefore, we offer an inexpensive, high-throughput assay that allows the rapid screening of novel candidate chelator compounds.


Asunto(s)
Quelantes del Hierro , Hierro , Quelantes del Hierro/farmacología , Quelantes del Hierro/metabolismo , Hierro/metabolismo , Fluoresceínas/metabolismo , Colorantes Fluorescentes
7.
Int J Biol Macromol ; 263(Pt 2): 130387, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38401586

RESUMEN

Alzheimer's disease (AD) is a prevalent form of neurodegenerative disease with a complex pathophysiology that remains not fully understood, and the exact mechanism of neurodegeneration is uncertain. Ferroptosis has been linked to the progression of degenerative diseases observed in AD models. The present study is designed to investigate the protective effects of spermidine, a potent antioxidant and iron chelator, and its synergistic interactions with ciprofloxacin, another iron chelator, in modulating ferroptosis and mitigating AD progression in rats. This study investigated AD-related biomarkers like neurotoxic amyloid beta (Aß), arginase I, and serotonin. Spermidine demonstrated an anti-ferroptotic effect in the AD model, evident from the modulation of ferroptosis parameters such as hippocampus iron levels, reduced protein expression of transferrin receptor 1 (TFR1), and arachidonate 15-lipoxygenase (ALOX15). Additionally, the administration of spermidine led to a significant increase in protein expression of phosphorylated nuclear factor erythroid 2-related factor 2 (p-Nrf2) and upregulation of Cystine/glutamate transporter (SLC7A11) gene expression. Moreover, spermidine notably decreased p53 protein levels, acrolein, and gene expression of spermidine/spermine N1-acetyltransferase 1 (SAT1). Overall, our findings suggest that spermidine and/or ciprofloxacin may offer potential benefits against AD by modulating ferroptosis. Furthermore, spermidine enhanced the antioxidant efficacy of ciprofloxacin and reduced its toxic effects.


Asunto(s)
Enfermedad de Alzheimer , Ferroptosis , Enfermedades Neurodegenerativas , Ratas , Masculino , Animales , Enfermedad de Alzheimer/tratamiento farmacológico , Espermidina/farmacología , Espermidina/metabolismo , Antioxidantes/farmacología , Antioxidantes/metabolismo , Péptidos beta-Amiloides/metabolismo , Estrés Oxidativo , Ciprofloxacina/farmacología , Quelantes del Hierro/farmacología , Factor 2 Relacionado con NF-E2/metabolismo
8.
Artículo en Inglés | MEDLINE | ID: mdl-38423705

RESUMEN

Deferasirox is an iron-chelating drug developed by Novartis company for treatment of diseases accompanied by chronic iron overload; such as ß-thalassemia or sickle cell diseases. Owing to its advantages such as high affinity, specificity and wide therapeutic window, it is considered as first line treatment. The current chapter describes the physicochemical characteristics, mode of action, pharmacokinetics, therapeutic applications and synthetic methods for deferasirox. Moreover, it includes Fourier transform infrared spectrometry (FTIR) and nuclear magnetic resonance spectroscopy (NMR) analysis for its functional groups. In addition, the selected analytical methods are summarized to aid the analysts in their routine analysis of deferasirox.


Asunto(s)
Benzoatos , Sobrecarga de Hierro , Humanos , Deferasirox/farmacología , Deferasirox/uso terapéutico , Benzoatos/farmacología , Benzoatos/uso terapéutico , Benzoatos/metabolismo , Triazoles/uso terapéutico , Triazoles/farmacocinética , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Quelantes del Hierro/metabolismo , Sobrecarga de Hierro/tratamiento farmacológico , Hierro/metabolismo , Hierro/uso terapéutico
9.
ACS Infect Dis ; 10(2): 676-687, 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38287902

RESUMEN

Iron, as an essential micronutrient, plays a crucial role in host-pathogen interactions. In order to limit the growth of the pathogen, a common strategy of innate immunity includes withdrawing available iron to interfere with the cellular processes of the microorganism. Against that, unicellular parasites have developed powerful strategies to scavenge iron, despite the effort of the host. Iron-sequestering compounds, such as the approved and potent chelator deferoxamine (DFO), are considered a viable option for therapeutic intervention. Since iron is heavily utilized in the mitochondrion, targeting iron chelators in this organelle could constitute an effective therapeutic strategy. This work presents mitochondrially targeted DFO, mitoDFO, as a candidate against a range of unicellular parasites with promising in vitro efficiency. Intracellular Leishmania infection can be cleared by this compound, and experimentation with Trypanosoma brucei 427 elucidates its possible mode of action. The compound not only affects iron homeostasis but also alters the physiochemical properties of the inner mitochondrial membrane, resulting in a loss of function. Furthermore, investigating the virulence factors of pathogenic yeasts confirms that mitoDFO is a viable candidate for therapeutic intervention against a wide spectrum of microbe-associated diseases.


Asunto(s)
Antiinfecciosos , Hierro , Deferoxamina/química , Antiparasitarios/farmacología , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Mitocondrias
10.
Microbiol Spectr ; 12(2): e0259423, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38230926

RESUMEN

Fungal infections are a growing global health concern due to the limited number of available antifungal therapies as well as the emergence of fungi that are resistant to first-line antimicrobials, particularly azoles and echinocandins. Development of novel, selective antifungal therapies is challenging due to similarities between fungal and mammalian cells. An attractive source of potential antifungal treatments is provided by ecological niches co-inhabited by bacteria, fungi, and multicellular organisms, where complex relationships between multiple organisms have resulted in evolution of a wide variety of selective antimicrobials. Here, we characterized several analogs of one such natural compound, collismycin A. We show that NR-6226C has antifungal activity against several pathogenic Candida species, including C. albicans and C. glabrata, whereas it only has little toxicity against mammalian cells. Mechanistically, NR-6226C selectively chelates iron, which is a limiting factor for pathogenic fungi during infection. As a result, NR-6226C treatment causes severe mitochondrial dysfunction, leading to formation of reactive oxygen species, metabolic reprogramming, and a severe reduction in ATP levels. Using an in vivo model for fungal infections, we show that NR-6226C significantly increases survival of Candida-infected Galleria mellonella larvae. Finally, our data indicate that NR-6226C synergizes strongly with fluconazole in inhibition of C. albicans. Taken together, NR-6226C is a promising antifungal compound that acts by chelating iron and disrupting mitochondrial functions.IMPORTANCEDrug-resistant fungal infections are an emerging global threat, and pan-resistance to current antifungal therapies is an increasing problem. Clearly, there is a need for new antifungal drugs. In this study, we characterized a novel antifungal agent, the collismycin analog NR-6226C. NR-6226C has a favorable toxicity profile for human cells, which is essential for further clinical development. We unraveled the mechanism of action of NR-6226C and found that it disrupts iron homeostasis and thereby depletes fungal cells of energy. Importantly, NR-6226C strongly potentiates the antifungal activity of fluconazole, thereby providing inroads for combination therapy that may reduce or prevent azole resistance. Thus, NR-6226C is a promising compound for further development into antifungal treatment.


Asunto(s)
Antiinfecciosos , Micosis , Animales , Humanos , Antifúngicos/farmacología , Fluconazol/farmacología , Hierro , Candida , Micosis/microbiología , Candida albicans , Antiinfecciosos/farmacología , Azoles/farmacología , Candida glabrata , Quelantes del Hierro/farmacología , Farmacorresistencia Fúngica , Pruebas de Sensibilidad Microbiana , Mamíferos
11.
Inorg Chem ; 63(5): 2401-2417, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38265361

RESUMEN

As cancer cells exhibit an increased uptake of iron, targeting the interaction with iron has become a straightforward strategy in the fight against cancer. This work comprehensively characterizes the chemical properties of 6-methyl-3-{(2E)-2-[1-(2-pyridinyl)ethylidene]hydrazino}-5H-[1,2,4]triazino[5,6-b]indole (VLX600), a clinically investigated iron chelator, in solution. Its protonation processes, lipophilicity, and membrane permeability as well as its complexation with essential metal ions were investigated using UV-visible, electron paramagnetic resonance, and NMR spectroscopic and computational methods. Formation constants revealed the following order of metal binding affinity at pH 7.4: Cu(II) > Fe(II) > Zn(II). The structures of VLX600 (denoted as HL) and the coordination modes in its metal complexes [Cu(II)(LH)Cl2], [Cu(II)(L)(CH3OH)Cl], [Zn(II)(LH)Cl2], and [Fe(II)(LH)2](NO3)2 were elucidated by single-crystal X-ray diffraction. Redox properties of the iron complexes characterized by cyclic voltammetry showed strong preference of VLX600 toward Fe(II) over Fe(III). In vitro cytotoxicity of VLX600 was determined in six different human cancer cell lines, with IC50 values ranging from 0.039 to 0.51 µM. Premixing VLX600 with Fe(III), Zn(II), and Cu(II) salts in stoichiometric ratios had a rather little effect overall, thus neither potentiating nor abolishing cytotoxicity. Together, although clinically investigated as an iron chelator, this is the first comprehensive solution study of VLX600 and its interaction with physiologically essential metal ions.


Asunto(s)
Complejos de Coordinación , Compuestos Férricos , Hidrazonas , Triazoles , Humanos , Cobre/farmacología , Cobre/química , Metales/química , Hierro/química , Iones , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Quelantes del Hierro/farmacología , Compuestos Ferrosos
12.
Neuropharmacology ; 246: 109837, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38184274

RESUMEN

BACKGROUND: Current antidepressants have limitations due to insufficient efficacy and delay before improvement in symptoms. Polymorphisms of the serotonin transporter (5-HTT) gene have been linked to depression (when combined with stressful life events) and altered response to selective serotonergic reuptake inhibitors. We have previously revealed the antidepressant-like properties of the iron chelator deferiprone in the 5-HTT knock-out (KO) mouse model of depression. Furthermore, deferiprone was found to alter neural activity in the prefrontal cortex of both wild-type (WT) and 5-HTT KO mice. METHODS: In the current study, we examined the molecular effects of acute deferiprone treatment in the prefrontal cortex of both genotypes via phosphoproteomics analysis. RESULTS: In WT mice treated with deferiprone, there were 22 differentially expressed phosphosites, with gene ontology analysis implicating cytoskeletal proteins. In 5-HTT KO mice treated with deferiprone, we found 33 differentially expressed phosphosites. Gene ontology analyses revealed phosphoproteins that were predominantly involved in synaptic and glutamatergic signalling. In a drug-naïve cohort (without deferiprone administration), the analysis revealed 21 differentially expressed phosphosites in 5-HTT KO compared to WT mice. We confirmed the deferiprone-induced increase in tyrosine hydroxylase serine 40 residue phosphorylation (pTH-Ser40) (initially revealed in our phosphoproteomics study) by Western blot analysis, with deferiprone increasing pTH-Ser40 expression in WT and 5-HTT KO mice. CONCLUSION: As glutamatergic and synaptic signalling are dysfunctional in 5-HTT KO mice (and are the target of fast-acting antidepressant drugs such as ketamine), these molecular effects may underpin deferiprone's antidepressant-like properties. Furthermore, dopaminergic signalling may also be involved in deferiprone's antidepressant-like properties.


Asunto(s)
Antidepresivos , Hierro , Humanos , Animales , Ratones , Deferiprona , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Transducción de Señal , Quelantes del Hierro/farmacología , Ratones Noqueados
13.
Water Res ; 249: 120966, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38070340

RESUMEN

The effects of solar photo-Fenton (SPF) process mediated by the iron chelate Fe3+ imminodisuccinic acid (Fe:IDS) on both the inactivation of seven relevant pathogens and the potential for antibiotic resistance transfer (degradation of antibiotic resistance genes (ARGs) and after treatment regrowth), in real secondary treated urban wastewater, were investigated for the first time. A comparison with results obtained by sunlight/H2O2 process and Fe3+ ethylenediaminedisuccinic acid (Fe:EDDS) SPF was also carried out. ARGs were quantified by polymerase chain reaction (PCR) in samples before and after (3 h) the treatment. The persistence of the selected pathogens and ARGs was also evaluated in regrowth tests (72 h) under environmentally mimicking conditions. Fe:IDS SPF resulted to be more effective (from 1.4 log removal for Staphylococcus spp. to 4.3 log removal for Escherichia coli) than Fe:EDDS SPF (from 0.8 log removal for Pseudomonas aeruginosa to 2.0 log removal for Total coliphages) and sunlight/H2O2 (from 1.2 log removal for Clostridium perfringens to 3.3 log removal for E. coli) processes for the seven pathogens investigated. Potential pathogens regrowth was also severely affected, as no substantial regrowth was observed, both in presence and absence of catalase. A similar trend was observed for ARGs removal too (until 0.001 fold change expression for qnrS after 3 h). However, a poor effect and a slight increase in fold change was observed after treatment especially for gyrA, mefA and intl1. Overall, the effect of the investigated processes on ARGs was found to be ARG dependent. Noteworthy, coliphages can regrow after sunlight/H2O2 treatment unlike SPF processes, increasing the risk of antibiotic resistance transfer by transduction mechanism. In conclusion, Fe:IDS SPF is an attractive solution for tertiary treatment of urban wastewater in small wastewater treatment plants as it can provide effective disinfection and a higher protection against antibiotic resistance transfer than the other investigated processes.


Asunto(s)
Escherichia coli , Aguas Residuales , Hierro/farmacología , Desinfección/métodos , Peróxido de Hidrógeno/farmacología , Luz Solar , Farmacorresistencia Microbiana , Quelantes del Hierro/farmacología , Antibacterianos/farmacología
14.
Life Sci ; 336: 122328, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38061132

RESUMEN

AIMS: Inflammatory Bowel Disease (IBD) is associated with systemic iron deficiency and has been managed with iron supplements which cause adverse side effects. Conversely, some reports highlight iron depletion to ameliorate IBD. The underlying intestinal response and comparative benefit of iron depletion and supplementation in IBD is unknown. The aims of this work were to characterize and compare the effects of iron supplementation and iron depletion in IBD. MAIN METHODS: IBD was induced in Drosophila melanogaster using 3 % dextran sodium sulfate (DSS) in diet for 7 days. Using this model, we investigated the impacts of acute iron depletion (using bathophenanthroline disulfonate, BPS) and supplementation (using ferrous sulphate, FS), before and after IBD induction, on gut iron homeostasis, cell death, gut permeability, inflammation, antioxidant defence, antimicrobial response and several fly phenotypes. KEY FINDINGS: DSS decreased fly mass (p < 0.001), increased gut permeability (p < 0.001) and shortened lifespan (p = 0.035) compared to control. The DSS-fed flies also showed significantly elevated lipid peroxidation (p < 0.001), and the upregulated expression of apoptotic marker- drice (p < 0.001), tight junction protein - bbg (p < 0.001), antimicrobial peptide - dpta (p = 0.002) and proinflammatory cytokine - upd2 (p < 0.001). BPS significantly (p < 0.05) increased fly mass and lifespan, decreased gut permeability, decreased lipid peroxidation and decreased levels of drice, bbg, dpta and upd2 in IBD flies. This iron chelation (using BPS) showed better protection from DSS-induced IBD than iron supplementation (using FS). Preventive and curative interventions, by BPS or FS, also differed in outcomes. SIGNIFICANCE: This may inform precise management strategies aimed at tackling IBD and its recurrence.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Animales , Ratones , Colitis/inducido químicamente , Drosophila , Drosophila melanogaster , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/metabolismo , Hierro/metabolismo , Suplementos Dietéticos , Quelantes del Hierro/farmacología , Sulfato de Dextran , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Colon/metabolismo
15.
Biometals ; 37(2): 289-304, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38019378

RESUMEN

The redox reactivity of iron is a double-edged sword for cell functions, being either essential or harmful depending on metal concentration and location. Deregulation of iron homeostasis is associated with several clinical conditions, including viral infections. Clinical studies as well as in silico, in vitro and in vivo models show direct effects of several viruses on iron levels. There is support for the strategy of iron chelation as an alternative therapy to inhibit infection and/or viral replication, on the rationale that iron is required for the synthesis of some viral proteins and genes. In addition, abnormal iron levels can affect signaling immune response. However, other studies report different effects of viral infections on iron homeostasis, depending on the class and genotype of the virus, therefore making it difficult to predict whether iron chelation would have any benefit. This review brings general aspects of the relationship between iron homeostasis and the nonspecific immune response to viral infections, along with its relevance to the progress or inhibition of the inflammatory process, in order to elucidate situations in which the use of iron chelators could be efficient as antivirals.


Asunto(s)
Quelantes del Hierro , Virosis , Humanos , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Hierro/metabolismo , Virosis/tratamiento farmacológico
16.
Eur J Pharmacol ; 960: 176153, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38059446

RESUMEN

PURPOSE: Iron toxicity occurs under iron-overloaded settings, such as a high iron diet and blood transfusion, and damages important organs. Vanillin has been proven to have potential iron chelation capability. Given the negative effects of commonly used iron chelators like deferoxamine (DFO), we sought to examine the iron chelation potency of vanillin and evaluate its potential effect in the treatment of iron overload-related disorders. METHODS: 42 male NMRI mice were prepared for this purpose, and except for the negative control group, iron overload conditions were generated in them by injecting iron. Then normal saline (as a control), vanillin, and DFO (n = 7) were subsequently given to iron-overloaded mice. In the following, the activity of antioxidant enzymes catalase and superoxide dismutase were measured in the blood serum, brain, kidney, spleen, lung, and liver tissues of mice. Furthermore, the level of lipid peroxidation was determined by measuring the amount of malondialdehyde. Also, Perl's and H&E staining were used to examine the physiopathology changes of tissues. FINDINGS: Vanillin, a natural antioxidant compound, outperformed deferoxamine, a chemical iron chelator. Along with a decrease in iron content, the activity of catalase and superoxide dismutase enhanced in the iron-overloaded groups that were treated with vanillin. The level of lipid peroxidation was also declined in the iron-overloaded mice receiving vanillin. CONCLUSION: Vanillin can be used as a suitable substitute for chemical chelators with fewer side effects and equivalent efficiency. We encourage the use of this compound as a natural iron chelator following performing additional safety and efficacy studies.


Asunto(s)
Deferoxamina , Sobrecarga de Hierro , Ratones , Masculino , Animales , Deferoxamina/farmacología , Catalasa , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/patología , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Hierro , Superóxido Dismutasa
17.
Int J Mol Sci ; 24(24)2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38139106

RESUMEN

Ferroptosis is a form of cell death that is distinguished from other types of death for its peculiar characteristics of death regulated by iron accumulation, increase in ROS, and lipid peroxidation. In the past few years, experimental evidence has correlated ferroptosis with various pathological processes including neurodegenerative and cardiovascular diseases. Ferroptosis also is involved in several types of cancer because it has been shown to induce tumor cell death. In particular, the pharmacological induction of ferroptosis, contributing to the inhibition of the proliferative process, provides new ideas for the pharmacological treatment of cancer. Emerging evidence suggests that certain mechanisms including the Xc- system, GPx4, and iron chelators play a key role in the regulation of ferroptosis and can be used to block the progression of many diseases. This review summarizes current knowledge on the mechanism of ferroptosis and the latest advances in its multiple regulatory pathways, underlining ferroptosis' involvement in the diseases. Finally, we focused on several types of ferroptosis inducers and inhibitors, evaluating their impact on the cell death principal targets to provide new perspectives in the treatment of the diseases and a potential pharmacological development of new clinical therapies.


Asunto(s)
Ferroptosis , Neoplasias , Humanos , Hierro/metabolismo , Muerte Celular/fisiología , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Neoplasias/metabolismo , Peroxidación de Lípido
18.
Elife ; 122023 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-38153418

RESUMEN

Myelodysplastic syndrome (MDS) is a heterogeneous group of bone marrow stem cell disorders characterized by ineffective hematopoiesis and cytopenias, most commonly anemia. Red cell transfusion therapy for anemia in MDS results in iron overload, correlating with reduced overall survival. Whether the treatment of iron overload benefits MDS patients remains controversial. We evaluate underlying iron-related pathophysiology and the effect of iron chelation using deferiprone on erythropoiesis in NUP98-HOXD13 transgenic mice, a highly penetrant well-established MDS mouse model. Our results characterize an iron overload phenotype with aberrant erythropoiesis in these mice which was reversed by deferiprone-treatment. Serum erythropoietin levels decreased while erythroblast erythropoietin receptor expression increased in deferiprone-treated MDS mice. We demonstrate, for the first time, normalized expression of the iron chaperones Pcbp1 and Ncoa4 and increased ferritin stores in late-stage erythroblasts from deferiprone-treated MDS mice, evidence of aberrant iron trafficking in MDS erythroblasts. Importantly, erythroblast ferritin is increased in response to deferiprone, correlating with decreased erythroblast ROS. Finally, we confirmed increased expression of genes involved in iron uptake, sensing, and trafficking in stem and progenitor cells from MDS patients. Taken together, our findings provide evidence that erythroblast-specific iron metabolism is a novel potential therapeutic target to reverse ineffective erythropoiesis in MDS.


Asunto(s)
Anemia , Sobrecarga de Hierro , Humanos , Ratones , Animales , Eritropoyesis , Deferiprona , Sobrecarga de Hierro/complicaciones , Hierro , Ratones Transgénicos , Ferritinas , Quelantes del Hierro/farmacología
19.
Vet Parasitol ; 324: 110055, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37931475

RESUMEN

BACKGROUND: Babesiosis is an infectious disease caused by protozoa of the apicomplexan phylum, genus Babesia. It is a malaria-like parasitic disease that can be transmitted via tick bites. The apicomplexan phylum of eukaryotic microbial parasites has had detrimental impacts on human and veterinary medicine. There are only a few drugs currently available to treat this disease; however, parasitic strains that are resistant to these commercial drugs are increasing in numbers. Plasmodium and Babesia are closely related as they share similar biological features including mechanisms for host cell invasion and metabolism. Therefore, antimalarial drugs may be useful in the treatment of Babesia infections. In addition to antimalarials, iron chelators also inhibit parasite growth. In this study, we aimed to evaluate the in vitro inhibitory efficacy of iron chelator and different antimalarials in the treatment of Babesia bovis. METHODS: Cytotoxicity of antimalarial drugs; pyrimethamine, artefenomel, chloroquine, primaquine, dihydroarthemisinine, and the iron chelator, 1-(N-acetyl-6-aminohexyl)- 3-hydroxy-2 methylpyridin-4-one (CM1), were evaluated against Madin Darby Bovine Kidney (MDBK) cells and compared to diminazene aceturate, which is the currently available drug for animal babesiosis using an MTT solution. Afterwards, an evaluation of the in vitro growth-inhibitory effects of antimalarial drug concentrations was performed and monitored using a flow cytometer. Half maximal inhibitory concentrations (IC50) of each antimalarial and iron chelator were determined and compared to the antibabesial drug, diminazine aceturate, by interpolation using a curve-fitting technique. Subsequently, the effect of the drug combination was assessed by constructing an isobologram. Values of the sum of fractional inhibitions at 50% inhibition were then estimated. RESULTS: Results indicate that all drugs tested could safely inhibit babesia parasite growth, as high as 2500 µM were non-toxic to mammalian cells. Although no drugs inhibited B. bovis more effectively than diminazine aceturate in this experiment, in vitro growth inhibition results with IC50 values of pyrimethamine 6.25 ± 2.59 µM, artefenomel 2.56 ± 0.67 µM, chloroquine 2.14 ± 0.76 µM, primaquine 22.61 ± 6.72 µM, dihydroarthemisinine 4.65 ± 0.22 µM, 1-(N-acetyl-6-aminohexyl)- 3-hydroxy-2 methylpyridin-4-one (CM1) 9.73 ± 1.90 µM, and diminazine aceturate 0.42 ± 0.01 µM, confirm that all drugs could inhibit B. bovis and could be used as alternative treatments for bovine babesial infection. Furthermore, the efficacy of a combination of the iron chelator, CM1, in combination with artefenomel dihydroarthemisinin or chloroquine, and artefenomel in combination with the iron chelator, CM1, dihydroarthemisinin or chloroquine, exhibited synergism against B. bovis in vitro. CONCLUSION: Our evaluation of the inhibitory efficacy of the iron chelator CM1, antimalarial drugs, and a combination of these drugs against B. bovis could be potentially useful in the development and discovery of a novel drug for the treatment of B. bovis in the future.


Asunto(s)
Antimaláricos , Babesia , Babesiosis , Enfermedades de los Bovinos , Animales , Bovinos , Humanos , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Babesiosis/tratamiento farmacológico , Babesiosis/parasitología , Pirimetamina/farmacología , Primaquina/farmacología , Primaquina/uso terapéutico , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Cloroquina/farmacología , Cloroquina/uso terapéutico , Concentración 50 Inhibidora , Mamíferos , Enfermedades de los Bovinos/tratamiento farmacológico
20.
Sci Rep ; 13(1): 19773, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37957293

RESUMEN

Iron overload (IOL) can cause hepatorenal damage due to iron-mediated oxidative and mitochondrial damage. Remarkably, combining a natural iron chelator with an antioxidant can exert greater efficacy than monotherapy. Thus, the present study aimed to evaluate the efficacy of Chia and CoQ10 to chelate excess iron and prevent hepatorenal oxidative damage in IOL mice. Male Swiss albino mice (n = 49) were randomly assigned to seven groups: control, dietary Chia, CoQ10, IOL, IOL + Chia, IOL + CoQ10, and IOL + Chia + CoQ10. Computational chemistry indicates that the phytic acid found in the Chia seeds is stable, reactive, and able to bind to up to three iron ions (both Fe2+ and Fe3+). IOL induced a significant (P < 0.05) increase in serum iron, ferritin, transferrin, TIBC, TSI, RBCs, Hb, MCV, MCH, WBCs, AST, ALT, creatinine, and MDA. IOL causes a significant (P < 0.05) decrease in UIBC, platelets, and antioxidant molecules (GSH, SOD, CAT, and GR). Also, IOL elicits mitochondrial membrane change depolarization, and DNA fragmentation and suppresses mitochondrial DNA copies. Furthermore, substantial changes in hepatic and renal tissue, including hepatocellular necrosis and apoptosis, glomerular degeneration, glomerular basement membrane thickening, and tubular degeneration, were observed in the IOL group. Dietary Chia and CoQ10 induced significant (P < 0.05) amelioration in all the mentioned parameters. They can mostly repair the abnormal architecture of hepatic and renal tissues induced by IOL, as signified by normal sinusoids, normal central veins, and neither glomerular damage nor degenerated tubules. In conclusion, the combined treatment with Chia + CoQ10 exerts more pronounced efficacy than monotherapy in hepatorenal protection via chelating excess iron and improved cellular antioxidant status and hepatorenal mitochondrial function in IOL mice.


Asunto(s)
Antioxidantes , Sobrecarga de Hierro , Ratones , Masculino , Animales , Antioxidantes/farmacología , Antioxidantes/metabolismo , Ubiquinona/metabolismo , Estrés Oxidativo , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/metabolismo , Hierro/metabolismo , Quelantes del Hierro/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA