Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 853
Filter
1.
Int J Mol Sci ; 25(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38791185

ABSTRACT

Acetylsalicylic acid or aspirin is the most commonly used drug in the world and is taken daily by millions of people. There is increasing evidence that chronic administration of low-dose aspirin of about 75-100 mg/day can cause iron deficiency anaemia (IDA) in the absence of major gastric bleeding; this is found in a large number of about 20% otherwise healthy elderly (>65 years) individuals. The mechanisms of the cause of IDA in this category of individuals are still largely unknown. Evidence is presented suggesting that a likely cause of IDA in this category of aspirin users is the chelation activity and increased excretion of iron caused by aspirin chelating metabolites (ACMs). It is estimated that 90% of oral aspirin is metabolized into about 70% of the ACMs salicyluric acid, salicylic acid, 2,5-dihydroxybenzoic acid, and 2,3-dihydroxybenzoic acid. All ACMs have a high affinity for binding iron and ability to mobilize iron from different iron pools, causing an overall net increase in iron excretion and altering iron balance. Interestingly, 2,3-dihydroxybenzoic acid has been previously tested in iron-loaded thalassaemia patients, leading to substantial increases in iron excretion. The daily administration of low-dose aspirin for long-term periods is likely to enhance the overall iron excretion in small increments each time due to the combined iron mobilization effect of the ACM. In particular, IDA is likely to occur mainly in populations such as elderly vegetarian adults with meals low in iron content. Furthermore, IDA may be exacerbated by the combinations of ACM with other dietary components, which can prevent iron absorption and enhance iron excretion. Overall, aspirin is acting as a chelating pro-drug similar to dexrazoxane, and the ACM as combination chelation therapy. Iron balance, pharmacological, and other studies on the interaction of iron and aspirin, as well as ACM, are likely to shed more light on the mechanism of IDA. Similar mechanisms of iron chelation through ACM may also be implicated in patient improvements observed in cancer, neurodegenerative, and other disease categories when treated long-term with daily aspirin. In particular, the role of aspirin and ACM in iron metabolism and free radical pathology includes ferroptosis, and may identify other missing links in the therapeutic effects of aspirin in many more diseases. It is suggested that aspirin is the first non-chelating drug described to cause IDA through its ACM metabolites. The therapeutic, pharmacological, toxicological and other implications of aspirin are incomplete without taking into consideration the iron binding and other effects of the ACM.


Subject(s)
Anemia, Iron-Deficiency , Aspirin , Iron Chelating Agents , Iron , Humans , Aspirin/therapeutic use , Aspirin/metabolism , Anemia, Iron-Deficiency/metabolism , Anemia, Iron-Deficiency/drug therapy , Iron/metabolism , Iron Chelating Agents/therapeutic use , Iron Chelating Agents/metabolism , Salicylic Acid/metabolism , Gentisates/metabolism , Hippurates/metabolism , Hydroxybenzoates
2.
Food Chem ; 451: 139493, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38703728

ABSTRACT

Iron chelating peptides have been widely utilized as iron supplements due to their excellent absorption capacity, However, the high cost and cumbersome manufacturing process of these peptides significantly limit their industrial application. In this study, fermentation was used for the first time to prepare iron chelating peptides. Bacillus altitudinis 3*1-3 was selected as the most suitable strain from 50 strains. The hydrolysates of fermented scallop skirts showed excellent iron-chelating capacity (9.39 mg/g). Aspartic acid, glutamic acid, and histidine are crucial for the binding of peptides to ferrous ions. The heptapeptide (FEDPEFE) forms six binding bonds with ferrous irons. Compared with ferrous sulfate, peptide-ferrous chelate showed more stability in salt solution and simulated gastrointestinal juice (p < 0.05). Furthermore, the fermentation method could save >50% of the cost compared with the enzymatic method. The results can provide a theoretical basis for the preparation of ferrous-chelated peptides using the fermentation method.


Subject(s)
Bacillus , Fermentation , Iron Chelating Agents , Pectinidae , Peptides , Animals , Pectinidae/chemistry , Pectinidae/metabolism , Pectinidae/microbiology , Peptides/chemistry , Peptides/metabolism , Iron Chelating Agents/chemistry , Iron Chelating Agents/metabolism , Bacillus/metabolism , Bacillus/chemistry , Iron/chemistry , Iron/metabolism
3.
Metallomics ; 16(4)2024 04 05.
Article in English | MEDLINE | ID: mdl-38449344

ABSTRACT

Iron is a vital metal for most biological functions in tissues, and its concentration is exquisitely regulated at the cellular level. During the process of differentiation, keratinocytes in the epidermis undergo a noticeable reduction in iron content. Conversely, psoriatic lesions, characterized by disruptions in epidermal differentiation, frequently reveal an excessive accumulation of iron within keratinocytes that have undergone differentiation. In this study, we clarified the significance of attenuated cellular iron content in the intricate course of epidermal differentiation. We illustrated this phenomenon through the utilization of hinokitiol, an iron chelator derived from the heartwood of Taiwanese hinoki, which forcibly delivers iron into cells independent of the intrinsic iron-regulation systems. While primary cultured keratinocytes readily succumbed to necrotic cell death by this iron chelator, mild administration of the hinokitiol-iron complex modestly disrupts the process of differentiation in these cells. Notably, keratinocyte model cells HaCaT and anaplastic skin rudiments exhibit remarkable resilience against the cytotoxic impact of hinokitiol, and the potent artificial influx of iron explains a suppressive effect selectively on epidermal differentiation. Moreover, the augmentation of iron content induced by the overexpression of divalent metal transporter 1 culminates in the inhibition of differentiation in HaCaT cells. Consequently, the diminution in cellular iron content emerges as an important determinant influencing the trajectory of keratinocyte differentiation.


Subject(s)
Iron , Keratinocytes , Tropolone/analogs & derivatives , Iron/metabolism , Keratinocytes/metabolism , Monoterpenes/metabolism , Epidermis/physiology , Cell Differentiation/physiology , Iron Chelating Agents/metabolism
4.
J Vis Exp ; (204)2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38465932

ABSTRACT

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.


Subject(s)
Iron Chelating Agents , Iron , Iron Chelating Agents/pharmacology , Iron Chelating Agents/metabolism , Iron/metabolism , Fluoresceins/metabolism , Fluorescent Dyes
5.
Article in English | MEDLINE | ID: mdl-38423705

ABSTRACT

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.


Subject(s)
Benzoates , Iron Overload , Humans , Deferasirox/pharmacology , Deferasirox/therapeutic use , Benzoates/pharmacology , Benzoates/therapeutic use , Benzoates/metabolism , Triazoles/therapeutic use , Triazoles/pharmacokinetics , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Iron Chelating Agents/metabolism , Iron Overload/drug therapy , Iron/metabolism , Iron/therapeutic use
6.
Food Funct ; 14(13): 6049-6061, 2023 Jul 03.
Article in English | MEDLINE | ID: mdl-37313959

ABSTRACT

Iron deficiency (ID) is the biggest cause of anemia. This pilot study aimed to investigate the effects of food-derived oligopeptide iron chelates on ameliorating liver injury and restoring gut microbiota homeostasis in iron-deficiency anemia (IDA) female rats. Female Sprague-Dawley rats at 21 days old were selected and randomly divided into a control group (N = 4) and an ID model group (N = 16). The ID model group was fed an iron-deficient diet containing 4 mg kg-1 iron for 28 days to generate the IDA rat model and then randomly subdivided into four groups (N = 4 for each group): ID group, ferrous sulfate group, marine fish oligopeptide iron chelate (MCOP-Fe) group, and whey protein oligopeptide iron chelate (WPP-Fe) group. Iron supplements were given to rats in the three intervention groups once per day via intragastric administration for three weeks. After iron supplementation, the hemoglobin levels in the three intervention groups were significantly improved, with the MCOP-Fe and WPP-Fe groups returning to normal. The ALT and AST levels in the ID group increased significantly, while levels in all intervention groups decreased to normal levels. Liver glutathione in the WPP-Fe group was increased, while the activity of superoxide dismutase also tended to be higher. In addition, 16S rRNA gene sequencing showed that IDA resulted in changes to intestinal microbiota. After intervention, the WPP-Fe group showed increased alpha diversity of intestinal microbes. Therefore, MCOP-Fe and WPP-Fe may improve the iron status of IDA female rats as well as ameliorate liver damage, with WPP-Fe showing a greater potential in improving gut microbiota imbalance.


Subject(s)
Anemia, Iron-Deficiency , Gastrointestinal Microbiome , Iron Deficiencies , Rats , Female , Animals , Iron/metabolism , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/metabolism , Pilot Projects , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Rats, Sprague-Dawley , Oligopeptides/metabolism , Liver/metabolism , Iron Chelating Agents/metabolism
7.
Int J Biochem Cell Biol ; 159: 106419, 2023 06.
Article in English | MEDLINE | ID: mdl-37086817

ABSTRACT

Iron chelators, such as deferoxamine, exert an anticancer effect by altering the activity of biomolecules critical for regulation of the cell cycle, cell metabolism, and apoptotic processes. Thus, iron chelators are sometimes used in combination with radio- and/or chemotherapy in the treatment of cancer. The possibility that deferoxamine could induce a program of senescence similar to radio- and/or chemotherapy, fostering adaptation in the treatment of cancer cells, is not fully understood. Using established biochemical techniques, biomarkers linked to lipid composition, and coherent anti-Stokes Raman scattering microscopy, we demonstrated that hepatocellular carcinoma-derived HepG2 cells survive after deferoxamine treatment, acquiring phenotypic traits and representative hallmarks of senescent cells. The results support the view that deferoxamine acts in HepG2 cells to produce oxidative stress-induced senescence by triggering sequential mitochondrial and lysosomal dysfunction accompanied by autophagy blockade. We also focused on the lipidome of senescent cells after deferoxamine treatment. Using mass spectrometry, we found that the deferoxamine-induced senescent cells presented marked remodeling of the phosphoinositol, sulfatide, and cardiolipin profiles, which all play a central role in cell signaling cascades, intracellular membrane trafficking, and mitochondria functions. Detection of alterations in glycosphingolipid sulfate species suggested modifications in ceramide generation, and turnover is frequently described in cancer cell survival and resistance to chemotherapy. Blockade of ceramide generation may explain autophagic default, resistance to apoptosis, and the onset of senescence.


Subject(s)
Deferoxamine , Sulfoglycosphingolipids , Humans , Deferoxamine/pharmacology , Deferoxamine/metabolism , Sulfoglycosphingolipids/metabolism , Sulfoglycosphingolipids/pharmacology , Hep G2 Cells , Iron Chelating Agents/pharmacology , Iron Chelating Agents/metabolism , Mitochondria/metabolism , Cellular Senescence
8.
Lett Appl Microbiol ; 76(2)2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36731874

ABSTRACT

Acinetobacter baumannii (AB) has become multidrug-resistant (MDR) in recent years, and, currently, there are limited effective treatment options. Nutrient metals (e.g. iron) are essential to the metabolic functions of AB. This study examined the impact of iron chelation on the growth of AB in vitro and in vivo. Susceptible and MDR-AB bloodstream isolates (n = 9) were recovered from different patients between 2011 and 2018. Clonal diversity was ascertained by Fourier-transform infrared spectroscopy. In vitro bacterial densities were measured over 20 h to determine growth profiles. Variable amounts of a chelating agent [deferiprone (DFP)] were added to create a concentration gradient. Galleria mellonella larvae were inoculated with an isolate, with and without DFP. Quantitative culture was used to ascertain the bacterial burden of aggregate larvae immediately and 4 h post-infection. Increasing concentrations of DFP caused a transient and concentration-dependent hindrance to in vitro growth, compared to the no-treatment group. In vivo bacterial burden immediately post-infection in both groups was comparable. After 4 h, the burden was much higher in the control group comparatively (8.7 and 6.7 log CFU g-1). These results support that micro-nutrient limitation has the potential of being a novel approach for treating high-risk infections due to MDR-AB.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Moths , Animals , Humans , Anti-Bacterial Agents/pharmacology , Acinetobacter Infections/microbiology , Moths/microbiology , Larva/microbiology , Iron Chelating Agents/metabolism , Iron Chelating Agents/pharmacology , Drug Resistance, Multiple, Bacterial , Microbial Sensitivity Tests
9.
Mol Neurobiol ; 60(2): 749-767, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36357615

ABSTRACT

Coumarins are plant-derived polyphenolic compounds belonging to the benzopyrones family, possessing wide-ranging pharmaceutical applications including cytoprotection, which may translate into therapeutic potential for multiple diseases, including Parkinson's disease (PD). Here we demonstrate the neuroprotective potential of a new polyhydroxyl coumarin, N-(1,3-dihydroxy-2-(hydroxymethyl)propan-2-yl)-2-(7-hydroxy-2-oxo-2H-chromen-4-yl)acetamide (CT51), against the mitochondrial toxin 1-methyl-4-phenylpyridinium (MPP+). MPP+'s mechanism of toxicity relates to its ability to inhibit complex I of the mitochondrial electron transport chain (METC), leading to adenosine triphosphate (ATP) depletion, increased reactive oxygen species (ROS) production, and apoptotic cell death, hence mimicking PD-related neuropathology. Dopaminergic differentiated human neuroblastoma cells were briefly pretreated with CT51, followed by toxin exposure. CT51 significantly restored somatic cell viability and neurite processes; hence, the drug targets cell bodies and axons thereby preserving neural function and circuitry against PD-related damage. Moreover, MPP+ emulates the iron dyshomeostasis affecting dopaminergic neurons in PD-affected brains, whilst CT51 was previously revealed as an effective iron chelator that preferentially partitions to mitochondria. We extend these findings by characterising the drug's interactive effects at the METC level. CT51 did not improve mitochondrial coupling efficiency. However, voltammetric measurements and high-resolution respirometry analysis revealed that CT51 acts as an antioxidant agent. Also, the neuronal protection afforded by CT51 associated with downregulating MPP+-induced upregulated expression of hypoxia-inducible factor 1 alpha (HIF-1α), a protein which regulates iron homeostasis and protects against certain forms of oxidative stress after translocating to mitochondria. Our findings support the further development of CT51 as a dual functioning iron chelator and antioxidant antiparkinsonian agent.


Subject(s)
Neuroprotective Agents , Parkinson Disease , Humans , Dopaminergic Neurons/metabolism , Parkinson Disease/pathology , Antioxidants/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Iron/metabolism , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Iron Chelating Agents/metabolism , Hypoxia-Inducible Factor 1/metabolism , Hypoxia-Inducible Factor 1/pharmacology , Hypoxia-Inducible Factor 1/therapeutic use , 1-Methyl-4-phenylpyridinium/toxicity , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/metabolism , Cell Line, Tumor
10.
J Physiol Pharmacol ; 73(3)2022 Jun.
Article in English | MEDLINE | ID: mdl-36302532

ABSTRACT

Acute cardiac pathologies represent one of the leading causes of death, while iron metabolism is recognized to be implicated in reactive oxygen species production, lipid peroxidation, and inflammation. The aim of the present study was to assess iron chelation effects in isoproterenol (ISO) induced acute cardiac stress. We divided male Wistar rats into preventive and secondary treatment groups, with the active arm consisting in deferiprone (DFP), a lipid permeable chelator. Mortality of ISO was 10-18.18% in both preventive and secondary groups. We analyzed serum and myocardial tissue parameters of inflammation, iron dynamics, and lipid peroxidation, accompanied by ultramicroscopy, histological, and ultrasound-derived parameters of left ventricular function. Results reveal that ISO-mediated lipid peroxidation and inflammation are alleviated by administration of DFP, with negligible effect on systemic ferroregulation dynamics and global ventricular function (as assessed by ultrasound). DFP administration after cardiovascular stress is associated with a decrease in lipid peroxidation and inflammation, without an improvement in gross left ventricular parameters.


Subject(s)
Myocardium , Animals , Male , Rats , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Iron/metabolism , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Iron Chelating Agents/metabolism , Isoproterenol/pharmacology , Isoproterenol/metabolism , Lipid Peroxidation , Myocardium/metabolism , Oxidative Stress , Rats, Wistar
11.
Int J Biol Macromol ; 219: 11-20, 2022 Oct 31.
Article in English | MEDLINE | ID: mdl-35931292

ABSTRACT

Iron deficiency anemia (IDA) is a common micronutrient deficiency. Tegillarca granosa (T. granosa) is a good source of iron due to its high content of hemoglobin. The present study aimed to determine the effects of glycosylation on structure, physicochemical characteristics and iron bioavailability of hemoglobin. Using Box-Behnken design and response surface methodology, the optimal conditions for hemoglobin-chitosan glycosylation were obtained: 61.8 °C, pH 6.3, hemoglobin/chitosan mass ratio of 4.3 and reaction time of 15 min. The formation of hemoglobin-chitosan conjugates was verified by SDS-PAGE and fluorescence spectroscopy. The surface hydrophobicity of hemoglobin was reduced by 20.90-65.05 % after glycosylation, along with the observations of elevated water-holding capacity, likely owing to the introduction of hydrophilic groups. Antioxidant capacity of glycosylated products (0.41-0.66 µM Trolox/mg protein) was markedly greater than that of original protein (0.06 µM Trolox/mg protein) due to the formation of brown polymers with antioxidant activity. In addition, glycosylation improved in vitro digestibility of hemoglobin by 41.15-69.09 %, which could be attributed to less ß-sheet in secondary structures. Moreover, hemoglobin (324.38 ng/mg) exhibited better iron absorption than FeSO4 (121.63 ng/mg), with the value being further enhanced by glycosylation (442.73 ng/mg), which may be due to the improved protein digestibility and iron-chelating capacity.


Subject(s)
Arcidae , Chitosan , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Biological Availability , Chitosan/metabolism , Glycosylation , Hemoglobins/chemistry , Iron/metabolism , Iron Chelating Agents/metabolism , Micronutrients/metabolism , Water/metabolism
12.
J Adv Res ; 39: 89-101, 2022 07.
Article in English | MEDLINE | ID: mdl-35777919

ABSTRACT

BACKGROUND: Bacterial infections involving multidrug-resistant Gram-negative bacteria have become critically involved in the current antibiotic crisis. This, together with the bacterial evolution ability, prioritizes the discovery of new antibiotics. Research on microbial iron acquisition pathways and metabolites, particularly siderophores, has highlighted hopeful aspects for the design of advanced antimicrobial approaches. Moreover, exploiting siderophores machinery to treat diseases associated with iron overload and cancer is of additional interest for the therapeutic arena. AIM OF REVIEW: This review highlights and provides a renewed perspective on the evolutionary path of siderophores, from primordial siderophores to new iron chelating agents, stimulating the field to build on the past and shape the future. KEY SCIENTIFIC CONCEPTS OF REVIEW: The effectiveness of siderophore-mimicking antibiotics appears to be high and selective for Gram-negative pathogens, rendering multidrug-resistant (MDR) bacteria susceptible to killing. Herein, cefiderocol, a new siderophore antibiotic, is well positioned in the clinic to treat MDR infections instigated by Gram-negative bacteria, particularly urinary tract infections and pneumonia. This siderophore has a mode of action based on a "Trojan horse" strategy, using the iron uptake systems for efficient bacterial penetration and killing. Recent progress has also been achieved concerning new iron chelating compounds to treat diseases associated with iron overload and cancer. Though these compounds still face great challenges for a clinical application, their promising results open up new doors for the design and development of innovative iron chelating compounds, taking benefit from the structurally diverse nature of siderophores.


Subject(s)
Iron Overload , Siderophores , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteria/metabolism , Humans , Iron , Iron Chelating Agents/metabolism , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Iron Overload/drug therapy , Siderophores/metabolism , Siderophores/pharmacology , Siderophores/therapeutic use
13.
mBio ; 13(4): e0149822, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35770947

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen responsible for acute and chronic infections in immunocompromised hosts. This organism is known to compete efficiently against coinfecting microorganisms, due in part to the secretion of antimicrobial molecules and the synthesis of siderophore molecules with high affinity for iron. P. aeruginosa possess a large repertoire of TonB-dependent transporters for the uptake of its own, as well as xenosiderophores released from other bacteria or fungi. Here, we show that P. aeruginosa is also capable of utilizing plant-derived polyphenols as an iron source. We found that exclusively plant-derived phenols containing a catechol group (i.e., chlorogenic acid, caffeic acid, quercetin, luteolin) induce the expression of the TonB-dependent transporters PiuA or PirA. This induction requires the two-component system PirR-PirS. Chlorogenic acid in its Fe(III)-loaded form was actively transported by PiuA and PirA and supported growth under iron-limiting conditions. Coincidentally, PiuA and PirA are also the main TonB transporters for the recently approved siderophore-drug conjugate cefiderocol. Surprisingly, quercetin supplementation increased the susceptibility of P. aeruginosa to siderophore-drug conjugates, due to induction of piuA and pirA expression mediated by the PirR-PirS two-component system. These findings suggest a potential novel therapeutic application for these biologically active dietary polyphenols. IMPORTANCE Iron is an essential element for living organisms. Most bacteria synthesize species-specific iron chelators, called siderophores, able to capture iron from their host or the environment. Pseudomonas aeruginosa, an opportunistic pathogen, produces two endogenous siderophores but is able to acquire iron also via xenosiderophores, produced by other bacteria or fungi, using a set of conserved TonB transporters. Here, we show that P. aeruginosa is also able to use plant metabolites, like quercetin and chlorogenic acid, as siderophores. These metabolites possess an iron-chelating catechol group and are recognized and transported by the TonB transporters PirA and PiuA. Since these transporters also promote the specific uptake of siderophore-drug conjugates, P. aeruginosa exposed to these plant catechols becomes hypersusceptible to this novel class of antibiotics. This unexpected finding suggests a potential therapeutic application for quercetin and chlorogenic acid, which were mainly investigated for their antioxidant and anti-inflammatory properties.


Subject(s)
Pseudomonas aeruginosa , Siderophores , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Catechols/metabolism , Catechols/pharmacology , Chlorogenic Acid/metabolism , Chlorogenic Acid/pharmacology , Ferric Compounds/metabolism , Iron/metabolism , Iron Chelating Agents/metabolism , Membrane Transport Proteins/metabolism , Pseudomonas aeruginosa/metabolism , Quercetin/metabolism , Siderophores/metabolism
14.
Free Radic Biol Med ; 188: 337-350, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35752374

ABSTRACT

Excessive iron has emerged in a large population of patients suffering from degenerative or hematological diseases with a common outcome, osteoporosis. However, its underlying mechanism remains to be clarified in order to formulate effective prevention and intervention against the loss of bone-forming osteoblasts. We show herein that increased intracellular iron by ferric ammonium citrate (FAC) mimicking the so-called non-transferrin bound iron concentrations leads to ferroptosis and impaired osteoblast differentiation. FAC upregulates the expression of Trfr and DMT1 genes to increase iron uptake, accumulating intracellular labile ferrous iron for iron overload status. Then, the excessive ferrous iron generates reactive oxygen species (ROS) and lipid peroxidation products (LPO), causing ferroptosis with its typical mitochondrial morphological changes, such as shrinkaged and condensed membrane with diminution and loss of crista and outer membrane rupture. We further examined that ferroptosis is the main cause responsible for FAC-disrupted osteoblast differentiation, although apoptosis and senescence are concurrently induced as well. Mechanistically, we revealed that iron dose-dependently down-regulates the expression of Wnt target genes and inhibits the transcription of Wnt reporter TopFlash construct, so as to inhibit the canonical Wnt signaling. Wnt agonist, ferroptosis inhibitor, or antioxidant melatonin reverses iron-inhibited canonical Wnt signaling to restore osteoblast differentiation by reducing ROS and LPO production to prevent ferroptosis notably without reducing iron overload. This study proposes a working model against excessive iron-induced osteoporosis: iron chelator deferoxamine or the above three drugs prevent ferroptosis, restore traditional Wnt signaling to maintain osteoblast differentiation no matter whether iron overload is removed or not. Additionally, iron chelator should be used to a suitable extent because iron itself is necessary for osteogenic differentiation.


Subject(s)
Ferroptosis , Iron Overload , Osteoporosis , Humans , Iron/metabolism , Iron Chelating Agents/metabolism , Iron Chelating Agents/pharmacology , Iron Overload/genetics , Iron Overload/metabolism , Osteoblasts , Osteogenesis/genetics , Osteoporosis/metabolism , Reactive Oxygen Species/metabolism , Wnt Signaling Pathway
15.
Anaerobe ; 75: 102582, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35533828

ABSTRACT

Desulfovibrio spp. is a commensal sulfate reducing bacterium that is present in small numbers in the gastrointestinal tract. Increased concentrations of Desulfovibrio spp. (blooms) have been reported in patients with inflammatory bowel disease and irritable bowel syndrome. Since stress has been reported to exacerbate symptoms of these chronic diseases, this study examined whether the stress catecholamine norepinephrine (NE) promotes Desulfovibrio growth. Norepinephrine-stimulated growth has been reported in other bacterial taxa, and this effect may depend on the availability of the micronutrient iron. OBJECTIVES: This study tested whether norepinephrine exposure affects the in vitro growth of Desulfovibrio vulgaris in an iron dependent manner. METHODS: DSV was incubated in a growth medium with and without 1 µm of norepinephrine. An additional growth assay added the iron chelator deferoxamine in NE exposed DSV. Iron regulatory genes were assessed with and without the treatment of NE and Deferoxamine. RESULTS: We found that norepinephrine significantly increased growth of D. vulgaris. Norepinephrine also increased bacterial production of hydrogen sulfide. Additionally, norepinephrine significantly increased bacterial expression in three of the four tested iron regulatory genes. The iron chelator deferoxamine inhibited growth of D. vulgaris in a dose-dependent manner and reversed the effect of norepinephrine on proliferation of D. vulgaris and on bacterial expression of iron regulatory genes. CONCLUSION: The data presented in this work suggests that promotion of D. vulgaris growth by norepinephrine is iron dependent.


Subject(s)
Desulfovibrio vulgaris , Desulfovibrio , Deferoxamine/metabolism , Deferoxamine/pharmacology , Desulfovibrio/metabolism , Desulfovibrio vulgaris/genetics , Humans , Iron/metabolism , Iron Chelating Agents/metabolism , Iron Chelating Agents/pharmacology , Norepinephrine/metabolism , Norepinephrine/pharmacology
16.
Toxicol Mech Methods ; 32(7): 530-541, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35313783

ABSTRACT

Aluminum (Al), a neurotoxic element, can induce Alzheimer's disease-like (AD-like) changes by triggering neuronal death. Iron homeostasis disturbance has also been implicated in Alzheimer's disease (AD), and excess iron exacerbates oxidative damage and cognitive defects. Ferroptosis is a nonapoptotic form of cell death dependent upon intracellular iron. However, the involvement of neuronal death induced by aluminum maltolate (Al(mal)3) in the pathogenesis of AD remains elusive. In this study, the results of three different behavioral experiments suggested that the learning and memory ability deteriorated and autonomous activity declined of these rats that exposed Al(mal)3 were alleviated by deferoxamine (DFO). Transmission electron microscope observations showed that the membrane was ruptured, and the membrane density increased and ridge disappearance (the most prominent characteristic of ferroptosis) in the perinuclear and cytoplasmic compartments of the hippocampal neurons were perceived in the exposure group, while the DFO group and 18 µM/kg Al(mal)3+DFO group were alleviated compared with 18 µM/kg Al(mal)3. In addition, DFO prevented oxidative stress, such as increased glutathione (GSH) and decreased malondialdehyde (MDA) and reactive oxygen species (ROS), while the latter two indexes had the same changing tendency as the total iron of brain tissue. These data indicated that Al(mal)3 could cause ferroptosis in Sprague-Dawley (SD) rat neurons, which was inhibited by DFO via reducing the content of iron and increasing the ability of cells to resist oxidative damage.


Subject(s)
Alzheimer Disease , Ferroptosis , Aluminum/toxicity , Animals , Brain/metabolism , Deferoxamine/metabolism , Deferoxamine/pharmacology , Iron/metabolism , Iron/toxicity , Iron Chelating Agents/metabolism , Iron Chelating Agents/pharmacology , Neurons/metabolism , Oxidative Stress , Rats , Rats, Sprague-Dawley
17.
Neurobiol Dis ; 168: 105693, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35304229

ABSTRACT

Brain iron is precisely regulated, and disrupted brain iron homeostasis is implicated in neuropsychological disease. Mounting evidence connects the iron status of the substantia nigra (SN) with locomotion-related neural symptomatology. Researchers in this field have long speculated that iron deficiency in the SN directly causes the high-locomotion symptoms observed in neuropsychiatric disorders. However, no direct experimental evidence of a causal relationship has been presented. To explore the relationship between iron deficiency in the SN and locomotion-related phenotypes, we stereotaxically injected the well-documented iron chelator, deferiprone (DFP) into the SN of mice to induce regional brain iron deprivation and subsequently performed behavioral tests. Altered expression of iron metabolism-related molecules was detected in the brain regions with interventions, and behavioral changes were observed. Targeted iron chelation effectively decreased the local iron content of the SN. Among the brain regions examined, only DFP injected into the SN resulted in the hyperlocomotion phenotype. Upon SN iron chelation, transferrin receptor (Tfr) expression was found to be upregulated. Conversely, viral vector-mediated SN-Tfr knockdown was sufficient to induce SN iron deficiency and mimic the hyperlocomotion phenotype. All locomotion changes had a significant negative correlation with iron alteration in the SN. Furthermore, SN iron disturbance also contributed to poor sleep efficiency. Thus, SN iron deficiency directly contributed to triggering both hyperlocomotion and sleep disturbances. This study offers a promising research and therapeutic direction for iron-linked neuropsychiatric diseases.


Subject(s)
Iron Deficiencies , Animals , Mice , Iron/metabolism , Iron Chelating Agents/metabolism , Phenotype , Substantia Nigra/metabolism
18.
Exp Biol Med (Maywood) ; 247(7): 574-583, 2022 04.
Article in English | MEDLINE | ID: mdl-35130741

ABSTRACT

Iron overload can be the result of either dysregulated iron metabolism in the case of hereditary hemochromatosis or repeated blood transfusions in the case of secondary hemochromatosis (e.g. in ß-thalassemia and sickle cell anemia patients). Under iron overload conditions, transferrin (Tf) saturation leads to an increase in non-Tf bound iron which can result in the generation of reactive oxygen species (ROS). These excess ROS can damage cellular components, resulting in the dysfunction of vital organs including iron overload cardiomyopathy (IOC). Multiple studies have demonstrated that L-type and T-type calcium channels are the main routes for iron uptake in the heart, and that calcium channel blockers, given either individually or in combination with standard iron chelators, confer cardioprotective effects under iron overload conditions. Treatment with antioxidants may also provide therapeutic benefits. Interestingly, recent studies have suggested that mitochondrial dynamics and regulated cell death (RCD) pathways are potential targets for pharmacological interventions against iron-induced cardiomyocyte injury. In this review, the potential therapeutic roles of iron chelators, antioxidants, iron uptake/metabolism modulators, mitochondrial dynamics modulators, and inhibitors of RCD pathways in IOC are summarized and discussed.


Subject(s)
Cardiomyopathies , Hemochromatosis , Iron Overload , Antioxidants/therapeutic use , Cardiomyopathies/drug therapy , Hemochromatosis/complications , Hemochromatosis/drug therapy , Hemochromatosis/metabolism , Humans , Iron/metabolism , Iron Chelating Agents/metabolism , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Iron Overload/complications , Iron Overload/drug therapy , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism
19.
mBio ; 12(6): e0327921, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34903054

ABSTRACT

Diverse 2-pyridone alkaloids have been identified with an array of biological and pharmaceutical activities, including the development of drugs. However, the biosynthetic regulation and chemical ecology of 2-pyridones remain largely elusive. Here, we report the inductive activation of the silent polyketide synthase-nonribosomal peptide synthetase (PKS-NRPS) (tenS) gene cluster for the biosynthesis of the tenellin-type 2-pyridones in the insect-pathogenic fungus Beauveria bassiana when cocultured with its natural competitor fungus Metarhizium robertsii. A pathway-specific transcription factor, tenR, was identified, and the overexpression of tenR well expanded the biosynthetic mechanism of 15-hydroxytenellin (15-HT) and its derivatives. In particular, a tandemly linked glycosyltransferase-methyltransferase gene pair located outside the tenS gene cluster was verified to mediate the rare and site-specific methylglucosylation of 15-HT at its N-OH residue. It was evident that both tenellin and 15-HT can chelate iron, which could benefit B. bassiana to outcompete M. robertsii in cocultures and to adapt to iron-replete and -depleted conditions. Relative to the wild-type strain, the deletion of tenS had no obvious negative effect on fungal virulence, but the overexpression of tenR could substantially increase fungal pathogenicity toward insect hosts. The results of this study well advance the understanding of the biosynthetic machinery and chemical ecology of 2-pyridones. IMPORTANCE Different 2-pyridones have been identified, with multiple biological activities but unclear chemical ecology. We found that the silent tenS gene cluster was activated in the insect pathogen Beauveria bassiana when the fungus was cocultured with its natural competitor Metarhizium robertsii. It was established that the gene cluster is regulated by a pathway-specific regulator, tenR, and the overexpression of this transcription factor expanded the biosynthetic machinery of the tenellin 2-pyridones. It was also found that the paired genes located outside the tenS cluster contribute to the site-specific methylglucosylation of the main compound 15-hydroxytenellin. Both tenellin and 15-hydroxytenellin can chelate and sequester iron to benefit the producing fungus to compete for different niches. This study well advances the biosynthetic mechanism and chemical ecology of 2-pyridones.


Subject(s)
Beauveria/metabolism , Iron Chelating Agents/metabolism , Metarhizium/metabolism , Pyridones/metabolism , Beauveria/enzymology , Beauveria/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Iron/metabolism , Metarhizium/enzymology , Metarhizium/genetics , Multigene Family , Peptide Synthases/genetics , Peptide Synthases/metabolism , Polyketide Synthases/genetics , Polyketide Synthases/metabolism , Pyridones/chemistry
20.
Bull Exp Biol Med ; 171(5): 606-610, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34617179

ABSTRACT

In a relatively isolated system of avian embryo, the metabolism of NO, a component of the dinitrosyl iron complexes (DNIC), the main NO donor in most tissues, depends on the ligands that make up the complex. This fact corroborates the earlier hypothesis that these ligands perform a regulatory function in NO metabolism. It is also shown that nitrite injected into the embryo is not oxidized to nitrate like NO in DNIC, but is accumulated outside the amniotic sac. Normally, nitrite is present in an embryo in trace amounts. These facts suggest that NO in the embryo is transferred from the donor molecule to a target in the embryo tissues further transformed with minimum oxidation to nitrite.


Subject(s)
Iron Chelating Agents/pharmacology , Iron/metabolism , Iron/pharmacology , Nitrogen Oxides/metabolism , Nitrogen Oxides/pharmacology , Animals , Catalase/antagonists & inhibitors , Catalase/drug effects , Catalase/metabolism , Chick Embryo , Citric Acid/pharmacology , Embryonic Development/drug effects , Glutathione , Hemoglobins/chemistry , Hemoglobins/metabolism , Hemoglobins/pharmacology , Iron/chemistry , Iron/physiology , Iron Chelating Agents/metabolism , Ligands , Nitrates/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/metabolism , Nitrites/metabolism , Nitrogen Oxides/chemistry , Oxidation-Reduction/drug effects , Phenanthrolines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...