Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.809
Filter
1.
J Environ Sci (China) ; 147: 597-606, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39003074

ABSTRACT

Harnessing bacteria for superoxide production in bioremediation holds immense promise, yet its practical application is hindered by slow production rates and the relatively weak redox potential of superoxide. This study delves into a cost-effective approach to amplify superoxide production using an Arthrobacter strain, a prevalent soil bacterial genus. Our research reveals that introducing a carbon source along with specific iron-binding ligands, including deferoxamine (DFO), diethylenetriamine pentaacetate (DTPA), citrate, and oxalate, robustly augments microbial superoxide generation. Moreover, our findings suggest that these iron-binding ligands play a pivotal role in converting superoxide into hydroxyl radicals by modulating the electron transfer rate between Fe(III)/Fe(II) and superoxide. Remarkably, among the tested ligands, only DTPA emerges as a potent promoter of this conversion process when complexed with Fe(III). We identify an optimal Fe(III) to DTPA ratio of approximately 1:1 for enhancing hydroxyl radical production within the Arthrobacter culture. This research underscores the efficacy of simultaneously introducing carbon sources and DTPA in facilitating superoxide production and its subsequent conversion to hydroxyl radicals, significantly elevating bioremediation performance. Furthermore, our study reveals that DTPA augments superoxide production in cultures of diverse soils, with various soil microorganisms beyond Arthrobacter identified as contributors to superoxide generation. This emphasizes the universal applicability of DTPA across multiple bacterial genera. In conclusion, our study introduces a promising methodology for enhancing microbial superoxide production and its conversion into hydroxyl radicals. These findings hold substantial implications for the deployment of microbial reactive oxygen species in bioremediation, offering innovative solutions for addressing environmental contamination challenges.


Subject(s)
Arthrobacter , Biodegradation, Environmental , Hydroxyl Radical , Iron , Superoxides , Hydroxyl Radical/metabolism , Superoxides/metabolism , Arthrobacter/metabolism , Iron/metabolism , Ligands , Soil Microbiology , Soil Pollutants/metabolism , Deferoxamine/metabolism
2.
Ther Adv Cardiovasc Dis ; 18: 17539447241277382, 2024.
Article in English | MEDLINE | ID: mdl-39291696

ABSTRACT

BACKGROUND: Reperfusion injury, characterized by oxidative stress and inflammation, poses a significant challenge in cardiac surgery with cardiopulmonary bypass (CPB). Deferoxamine, an iron-chelating compound, has shown promise in mitigating reperfusion injury by inhibiting iron-dependent lipid peroxidation and reactive oxygen species (ROS) production. OBJECTIVES: The objective of our study was to analyze and evaluate both the efficacy and safety of a new and promising intervention, that is, deferoxamine for ischemia-reperfusion injury (I/R). DESIGN: Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines are used to perform the study. DATA SOURCES AND METHODS: We conducted a systematic review following PRISMA guidelines to assess the efficacy and safety of deferoxamine in reducing I/R injury following CPB. A comprehensive search of electronic databases, namely, PubMed, Scopus, and Embase, yielded relevant studies published until August 18, 2023. Included studies evaluated ROS production, lipid peroxidation, cardiac performance, and morbidity outcomes. RESULTS: (a) ROS production: Multiple studies demonstrated a statistically significant decrease in ROS production in patients treated with deferoxamine, highlighting its potential to reduce oxidative stress. (b) Lipid peroxidation: Deferoxamine was associated with decreased lipid peroxidation levels, indicating its ability to protect cardiac tissue from oxidative damage during CPB. (c) Cardiac performance: Some studies reported improvements in left ventricular ejection fraction and wall motion score index with deferoxamine. CONCLUSION: Our review shows that deferoxamine is an efficacious and safe drug that can be used to prevent myocardial I/R injury following CPB. It also highlights the need for trials on a larger scale to develop potential strategies and guidelines on the use of deferoxamine for I/R injury.


Subject(s)
Cardiopulmonary Bypass , Deferoxamine , Myocardial Reperfusion Injury , Oxidative Stress , Reactive Oxygen Species , Humans , Deferoxamine/adverse effects , Deferoxamine/therapeutic use , Cardiopulmonary Bypass/adverse effects , Myocardial Reperfusion Injury/prevention & control , Oxidative Stress/drug effects , Treatment Outcome , Reactive Oxygen Species/metabolism , Male , Lipid Peroxidation/drug effects , Female , Middle Aged , Aged , Adult , Antioxidants/adverse effects , Antioxidants/administration & dosage
3.
Transplant Proc ; 56(8): 1732-1739, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39242312

ABSTRACT

OBJECTIVES: Ferroptosis plays a pivotal role in the pathogenesis of renal ischemia-reperfusion injury, where the processes are mediated by free ferrous ions and mitochondrial-released reactive oxygen species. However, the administration of high doses of cyclosporine A (CsA) or deferoxamine (DFO) poses a significant risk of renotoxicity. In contrast, low doses of DFO act as a ferrous iron chelator, and CsA functions as a mitochondrial reactive oxygen species blocker. This study aims to explore the potential protective effects of donor treatment with low-dose CsA, DFO, or their combination against ischemia-reperfusion injury during renal transplantation in a rat model. MATERIALS AND METHODS: In an ex vivo cold storage (CS) model utilizing renal slices, the impact of incorporating DFO, CsA, and a combination of both into the University of Wisconsin solution was assessed through the measurement of lactate dehydrogenase leakage. Additionally, their potential benefits were investigated in a rat donation after circulatory death (DCD) kidney transplant model, where the extent of damage was evaluated based on graft function, tubular necrosis, and inflammation. RESULTS: The co-administration of DFO and CsA effectively decreased the release of lactate dehydrogenase induced by CS ( P ≥ .05). In the in vivo model, this combined supplementation demonstrated a mitigating effect on reperfusion injury, evidenced by lower blood urea nitrogen levels and acute tubular necrosis scores compared to the control group (allP ≤ .05). Furthermore, the combined treatment significantly reduced apoptotic levels compared to the control group (P ≥ .05). CONCLUSIONS: The combined treatment with DFO and CsA mitigated the cold ischemia-reperfusion injury in the DCD kidney. Hence, this presents a new strategy for the CS of DCD kidney in clinical transplants.


Subject(s)
Cyclosporine , Deferoxamine , Kidney Transplantation , Reperfusion Injury , Animals , Reperfusion Injury/prevention & control , Reperfusion Injury/pathology , Reperfusion Injury/etiology , Kidney Transplantation/adverse effects , Deferoxamine/pharmacology , Rats , Male , Kidney/pathology , Kidney/drug effects , Organ Preservation Solutions , Drug Synergism , Cold Ischemia/adverse effects , Organ Preservation/methods , Disease Models, Animal , Raffinose/pharmacology , Allopurinol
4.
Discov Med ; 36(188): 1869-1879, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39327250

ABSTRACT

BACKGROUND: Susceptibility-weighted imaging (SWI) is a common imaging technique used to identify cerebral microbleeds. Given that spinal cord injury (SCI) often creates an environment that favors ferroptosis, a type of cell death driven by iron, this study aimed to explore the relationship between microbleeds on SWI and ferroptosis, and explore the effect of deferoxamine on SCI. METHODS: Thirty-six rabbits were divided into three groups: sham, SCI, and SCI with deferoxamine (DFO, a ferroptosis inhibitor) treatment (SCI+DFO). Following 48 hours of SCI modeling, the rabbits underwent magnetic resonance imaging (MRI) and SWI examinations. Ferroptosis markers and spinal cord tissue morphology were examined, and the modified Tarlov's score was used to assess neurological function. RESULTS: SWI analysis revealed that rabbits in the SCI group exhibited lower signal intensities and larger microbleed areas compared to the those in the SCI+DFO group (p < 0.05). The SCI+DFO group demonstrated significantly decreased iron and malondialdehyde (MDA) levels, coupled with increased glutathione (GSH) and glutathione peroxidase 4 (GPX4) levels, along with attenuated ferroptosis (p < 0.05). This group also displayed greater Neuronal Nuclei (NeuN) expression, Tarlov's scores, and neurological recovery rates (all p < 0.05). A significant positive correlation was found between the microbleed area and iron content (r = 0.59, p = 0.04), MDA (r = 0.75, p = 0.01), and mitochondrial damage (r = 0.90, p < 0.01). Conversely, a negative correlation was established between the microbleed area and GPX4 levels (r = -0.87, p < 0.01), as well as neurological function recovery (r = -0.62, p = 0.03). CONCLUSION: The extent of microbleeds on SWI following SCI is closely correlated with ferroptosis, and the inhibition of ferroptosis could improve neurologic function. These findings suggest that the area of microbleeds on SWI could potentially serve as a predictive marker for ferroptosis in spinal cord injury.


Subject(s)
Deferoxamine , Ferroptosis , Magnetic Resonance Imaging , Spinal Cord Injuries , Animals , Ferroptosis/drug effects , Rabbits , Spinal Cord Injuries/complications , Spinal Cord Injuries/diagnostic imaging , Spinal Cord Injuries/pathology , Magnetic Resonance Imaging/methods , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Iron/metabolism , Prognosis , Male , Disease Models, Animal , Spinal Cord/diagnostic imaging , Spinal Cord/pathology , Spinal Cord/metabolism
5.
PLoS One ; 19(9): e0310984, 2024.
Article in English | MEDLINE | ID: mdl-39316565

ABSTRACT

Magnetic resonance (MR) imaging is a powerful imaging modality for obtaining anatomical information with high spatial and temporal resolution. In the drug delivery system (DDS) framework, nanoparticles such as liposomes are potential candidates for MR imaging. We validated that RGD peptides are possible targeting molecules for pancreatic cancer with αvß3 integrin expression. This study aimed to evaluate RGD-modified liposomes loaded with ferrioxamine B for pancreatic cancer imaging. We synthesized four types of RGD-modified liposomes encapsulated with ferrioxamine B (SH-, H-, M-, and L-RGD-liposomes). The binding affinity of RGD-modified liposomes was evaluated in a competitive inhibition study using 125I-echistatin. To investigate the pharmacokinetics of RGD-modified liposomes, a biodistribution study using RGD-liposomes labeled with 111In was carried out in a human pancreatic cancer PANC-1 xenograft mouse model. Finally, MR was performed using ferrioxamine-B-loaded liposomes. RGD-liposomes inhibited the binding of 125I-echistatin to RGD. The biodistribution study revealed that 111In-RGD-liposomes accumulated significantly in the liver and spleen. Among the 111In-RGD-liposomes, 111In-H-RGD-liposomes showed the highest tumor-to-normal tissue ratio. In the MR study, H-RGD-liposomes loaded with ferrioxamine B showed higher tumor-to-muscle signal ratios than RKG-liposomes loaded with ferrioxamine B (control). We successfully synthesized RGD-liposomes that can target αvß3 integrin.


Subject(s)
Deferoxamine , Integrin alphaVbeta3 , Liposomes , Magnetic Resonance Imaging , Oligopeptides , Pancreatic Neoplasms , Animals , Liposomes/chemistry , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Integrin alphaVbeta3/metabolism , Deferoxamine/chemistry , Deferoxamine/pharmacokinetics , Deferoxamine/administration & dosage , Magnetic Resonance Imaging/methods , Mice , Humans , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Cell Line, Tumor , Tissue Distribution , Mice, Nude , Disease Models, Animal
6.
Mol Imaging Biol ; 26(5): 847-857, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39174789

ABSTRACT

PURPOSE: Thyroid-stimulating hormone receptor (TSHR) is a G-protein coupled receptor that is highly expressed on benign and malignant thyroid tissues. TSHR binding and activation has long been a component of thyroid cancer molecular imaging and radiotherapy, by promoting expression of the sodium-iodide symporter (NIS) and incorporation of I-131 into thyroid hormones. Here, we report the radiosynthesis and preclinical evaluation of a Zirconium-89 (89Zr) labeled TSHR antibody to serve as a positron emission tomography (PET) diagnostic correlate for therapeutic agents targeting TSHR without reliance on NIS. PROCEDURES: TSHR human monoclonal antibody K1-70 was conjugated to chelator desferrioxamine-p-benzyl-isothiocyanate, followed by labeling with Zr-89, yielding the radiotracer 89Zr-DFO-TSHR-Ab. The in vitro cellar uptake and binding affinity of 89Zr-DFO-TSHR-Ab were analyzed in three new TSHR stable overexpressing tumor cell lines and their corresponding wild types (WT) with low or no TSHR expression. 89Zr-DFO-TSHR-Ab PET/CT imaging of TSHR expression was evaluated in tumor mouse models bearing one TSHR-positive tumor and other negative control with or without the coinjection of antibody K1-70, and then verified by radiotracer biodistribution study and tumor immunohistochemistry (IHC). RESULTS: The conjugate DFO-TSHR-Ab was labeled with Zr-89 at 37 °C for 60 min and purified by PD-10 column in radiochemical yields of 68.8 ± 9.9%, radiochemical purities of 98.7 ± 0.8%, and specific activities of 19.1 ± 2.7 mCi/mg (n = 5). In vitro cell studies showed 89Zr-DFO-TSHR-Ab had significantly high uptake on TSHR expressing tumor cells with nanomolar affinity and high potency. Preclinical PET/CT imaging revealed that 89Zr-DFO-TSHR-Ab selectively detected TSHR expressing thyroid tumors and displayed improved in vivo performance with the coinjection of unlabeled TSHR antibody K1-70 leading to higher uptake in TSHR expressing tumors than parental WT tumors and physiologic tissues; this observation was confirmed by the biodistribution and immunostaining analyses. CONCLUSIONS: We synthesized 89Zr-labeled antibody K1-70 as a new radiopharmaceutical for PET imaging of TSHR. 89Zr-DFO-TSHR-Ab has high radioactive uptake and retention in TSHR expressing tumors and cleared quickly from most background tissues in mouse models. Our study demonstrated that 89Zr-DFO-TSHR-Ab has the potential for PET imaging of TSHR-positive thyroid cancer and monitoring TSHR-targeted therapy.


Subject(s)
Antibodies, Monoclonal , Positron-Emission Tomography , Radioisotopes , Receptors, Thyrotropin , Thyroid Neoplasms , Zirconium , Zirconium/chemistry , Animals , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Positron-Emission Tomography/methods , Humans , Receptors, Thyrotropin/metabolism , Cell Line, Tumor , Radioisotopes/chemistry , Tissue Distribution , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Mice , Mice, Nude , Deferoxamine/chemistry , Female
7.
Sci Rep ; 14(1): 19273, 2024 08 20.
Article in English | MEDLINE | ID: mdl-39164295

ABSTRACT

Extracellular vesicles (EVs) from mesenchymal stem cells (MSCs), specifically those preconditioned with deferoxamine (DFO) in canine adipose tissue-derived MSCs (cAT-MSCs), were explored for treating autoimmune diseases. This study assessed the effects of DFO-preconditioned EVs (EVDFO) in an experimental autoimmune encephalomyelitis (EAE) mouse model. cAT-MSCs were treated with DFO for 48 h, after which EVs were isolated. EAE mice received intranasal EV or EVDFO treatments and were euthanized following histopathologic analysis; RNA and protein expression levels were measured. Histologically, EV and EVDFO groups showed a significant reduction in inflammatory cell infiltration and demyelination. Immunofluorescence revealed increased CD206 and Foxp3 expression, indicating elevated M2 macrophages and regulatory T (Treg) cells, particularly in the EVDFO group. Treg cells also notably increased in the spleen of EVDFO -treated mice. STAT3 and pSTAT3 proteins were upregulated in the EAE groups compared to the naïve group. However, following EV treatment, STAT3 expression decreased compared to the EAE group, whereas pSTAT3 expression was similar in both the EV and EAE groups. In conclusion, EVDFO treatment resulted in reduced STAT3 expression, suggesting its role in T cell regulation and the potential of EVDFO in modulating the STAT3 pathway for reducing inflammation more effectively than non-preconditioned EVs.


Subject(s)
Deferoxamine , Encephalomyelitis, Autoimmune, Experimental , Extracellular Vesicles , Inflammation , Mesenchymal Stem Cells , STAT3 Transcription Factor , T-Lymphocytes, Regulatory , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , STAT3 Transcription Factor/metabolism , Mice , Dogs , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Mesenchymal Stem Cells/metabolism , Inflammation/pathology , Female , Disease Models, Animal
8.
Colloids Surf B Biointerfaces ; 244: 114160, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39142232

ABSTRACT

The delay of diabetic wound healing puts a huge burden on the society. The key factors hindering wound healing include bacterial infection, unresolved inflammation and poorly generated blood vessels. In this paper, glycidyl trimethyl ammonium chloride (GTA) was grafted to chitosan (CS) to obtain quaternary ammonium grafted chitosan (QCS) with enhanced antibacterial performance, and then cross-linked by dialdehyde terminated poly(ethylene oxide) (PEO DA) to construct QCS/PEO DA hydrogel with tissue adhesion, biodegradation and self-healing properties. The QCS/PEO DA hydrogel is loaded with tannin acid (TA) and deferoxamine (DFO) to enhance antioxidant property and angiogenesis. At the same time, the TA and DFO loaded TA@DFO/hydrogel preserved the biocompatibility and biodegradability of chitosan. Moreover, the multifunctional hydrogel behaved excellent hemostatic properties in mice model and significantly promoted the healing efficacy of diabetic wounds. Overall, the TA@DFO/hydrogel is promising anti-infection dressing material for diabetic wound healing.


Subject(s)
Anti-Bacterial Agents , Chitosan , Deferoxamine , Diabetes Mellitus, Experimental , Hydrogels , Quaternary Ammonium Compounds , Tannins , Wound Healing , Chitosan/chemistry , Chitosan/pharmacology , Wound Healing/drug effects , Tannins/chemistry , Tannins/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Mice , Deferoxamine/pharmacology , Deferoxamine/chemistry , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Microbial Sensitivity Tests , Male , Drug Carriers/chemistry , Staphylococcus aureus/drug effects , Humans , Escherichia coli/drug effects
9.
J Hazard Mater ; 478: 135542, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39154481

ABSTRACT

Epidemiological studies have shown that coke oven emissions (COEs) affect the deterioration of asthma, but has not been proven by experimental results. In this study, we found for the first time that COEs exacerbate allergen house dust mite (HDM)-induced allergic asthma in the mouse model. The findings reveal that airway inflammation, airway remodeling and allergic reaction were aggravated in the COE + HDM combined exposure group compared with the individual exposure group. Mechanism studies indicated higher levels of iron and MDA in the COE + HDM combined exposure group, along with increased expression of Ptgs2 and reduced GPX4 expression. Iron chelator deferoxamine (DFO) effectively inhibited ferroptosis induced by COE synergistically with HDM in vitro. Further studies highlighted the role of ferritinophagy in the COE + HDM-induced ferroptosis. 3-methyladenine (3-MA) could inhibit ferroptosis in the COE + HDM exposure group. Interestingly, we injected DFO intraperitoneally into mice in the combined exposure group and found DFO could significantly inhibit the COE-exacerbated ferroptosis and allergic asthma. Our findings link ferroptosis with COE-exacerbated allergic asthma, implying that ferroptosis may have important therapeutic potential for asthma in patients with occupational exposure of COE.


Subject(s)
Asthma , Epithelial Cells , Ferroptosis , Mice, Inbred BALB C , Animals , Ferroptosis/drug effects , Asthma/chemically induced , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Pyroglyphidae/immunology , Mice , Deferoxamine/pharmacology , Female , Air Pollutants/toxicity , Iron/metabolism , Cyclooxygenase 2/metabolism
10.
Medicine (Baltimore) ; 103(34): e39142, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39183421

ABSTRACT

BACKGROUND: Deferoxamine is a potent iron chelator that could remove iron from the virus, and severe acute respiratory syndrome coronavirus 2 requires iron to replication. Also, deferoxamine has antioxidant and cytokine-modulating effects. Therefore, we evaluated the efficacy and safety of deferoxamine in patients with moderate coronavirus disease 2019 (COVID-19). METHODS: In this randomized controlled trial, patients with moderate COVID-19 were randomly assigned in a 1:1 ratio to the deferoxamine group (received a solution of 500 mg deferoxamine divided into 4 doses a day through a nebulizer for 7 days) and the control group. The main outcomes were viral clearance, oxygen saturation (SPO2), body temperature, and respiratory rate (RR). Intensive care unit admission, hospital length of stay, and hospital mortality were also assessed. RESULTS: A total of 62 patients, with 30 in the deferoxamine group and 32 in the control group, were randomly assigned. There was no statistically significant improvement in viral clearance after the intervention ended in the deferoxamine group (36.7%) compared to the control group (34.4%). The results showed there was no significant difference between the analyzed groups in terms of SPO2, body temperature, RR, and the number of patients with a worse prognosis (SPO2 < 96%, temperature ≥ 37.5 °C, or RR ≥ 16/min) at the end of the study. There were no significant differences seen between the groups in terms of intensive care unit admission, hospital length of stay, hospital mortality, and the occurrence of adverse medication events during the follow-up period. CONCLUSION: Deferoxamine had no significant impact on improving moderately ill patients with COVID-19. However, it was well-tolerated in the patients, and this intervention demonstrated a safe profile of adverse events.


Subject(s)
COVID-19 Drug Treatment , Deferoxamine , SARS-CoV-2 , Humans , Deferoxamine/therapeutic use , Deferoxamine/administration & dosage , Male , Female , Middle Aged , COVID-19/mortality , Aged , Length of Stay/statistics & numerical data , Treatment Outcome , Hospital Mortality , Adult , Oxygen Saturation , Body Temperature/drug effects
11.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 38(8): 1001-1009, 2024 Aug 15.
Article in Chinese | MEDLINE | ID: mdl-39175324

ABSTRACT

Objective: To compare the effects of hypoxia-inducible drugs using deferoxamine (DFO) and accordion technique (AT) on activating the hypoxia-inducible factor 1α (HIF-1α)/vascular endothelial growth factor (VEGF) signaling pathway to promote bone regeneration and remodelling during consolidation phase of distraction osteogenesis (DO). Methods: Forty-five specific-pathogen-free adult male Sprague-Dawley (SD) rats were randomly divided into the control group, DFO group, and AT group, with 15 rats in each group. All rats underwent osteotomy to establish a right femur DO model. Then, continuous distraction was started for 10 days after 5 days of latency in each group. During the consolidation phase after distraction, no intervention was performed in the control group; DFO was locally perfused into the distraction area in the DFO group starting at the 3rd week of consolidation phase; cyclic stress stimulation was given in the AT group starting at the 3rd week of consolidation phase. The general condition of rats in each group was observed. X-ray films were conducted at the end of the distraction phase and at the 2nd, 4th, and 6th weeks of the consolidation phase to observe the calcification in the distraction area. At the 4th and 6th weeks of the consolidation phase, peripheral blood was taken for ELISA detection (HIF-1α, VEGF, CD31, and Osterix), femoral specimens were harvested for gross observation, histological staining (HE staining), and immunohistochemical staining [HIF-1α, VEGF, osteopontin (OPN), osteocalcin (OCN)]. At the 6th week of the consolidation phase, Micro-CT was used to observe the new bone mineral density (BMD), bone volume/tissue volume (BV/TV), trabecular separation (Tb.Sp), trabecular number (Tb.N), and trabecular thickness (Tb.Th) in the distraction area, and biomechanical test (ultimate load, elastic modulus, energy to failure, and stiffness) to detect bone regeneration in the distraction area. Results: The rats in all groups survived until the termination of the experiment. ELISA showed that the contents of HIF-1α, VEGF, CD31, and Osterix in the serum of the AT group were significantly higher than those of the DFO group and control group at the 4th and 6th weeks of the consolidation phase ( P<0.05). General observation, X-ray films, Micro-CT, and biomechanical test showed that bone formation in the femoral distraction area was significantly better in the DFO group and AT group than in the control group, and complete recanalization of the medullary cavity was achieved in the AT group, and BMD, BV/TV, Tb.Sp, Tb.N, and Tb.Th, as well as ultimate load, elastic modulus, energy to failure, and stiffness in the distraction area, were better in the AT group than in the DFO group and control group, and the differences were significant ( P<0.05). HE staining showed that trabecular bone formation and maturation in the distraction area were better in the AT group than in the DFO group and control group. Immunohistochemical staining showed that at the 4th week of consolidation phase, the expression levels of HIF-1α, VEGF, OCN, and OPN in the distraction area of the AT group were significantly higher than those of the DFO group and control group ( P<0.05); however, at 6th week of consolidation phase, the above indicators were lower in the AT group than in the DFO group and control group, but there was no significant difference between groups ( P>0.05). Conclusion: Both continuous local perfusion of DFO in the distraction area and AT during the consolidation phase can activate the HIF-1α/VEGF signaling pathway. However, AT is more effective than local perfusion of DFO in promoting the process of angiogenesis, osteogenesis, and bone remodelling.


Subject(s)
Bone Regeneration , Deferoxamine , Hypoxia-Inducible Factor 1, alpha Subunit , Osteogenesis, Distraction , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A , Animals , Osteogenesis, Distraction/methods , Male , Rats , Deferoxamine/pharmacology , Bone Regeneration/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Vascular Endothelial Growth Factor A/metabolism , Femur , Signal Transduction/drug effects , Osteogenesis/drug effects , Bone Density/drug effects , Osteotomy/methods
12.
Bioorg Med Chem ; 112: 117842, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39173538

ABSTRACT

The decline of antibiotics efficacy worldwide has recently reached a critical point urging for the development of new strategies to regain upper hand on multidrug resistant bacterial strains. In this context, the raise of photodynamic therapy (PDT), initially based on organic photosensitizers (PS) and more recently on organometallic PS, offers promising perspectives. Many PS exert their biological effects through the generation of reactive oxygen species (ROS) able to freely diffuse into and to kill surrounding bacteria. Hijacking of the bacterial iron-uptake systems with siderophore-PS conjugates would specifically target pathogens. Here, we report the synthesis of unprecedented conjugates between the siderophore desferrioxamine B (DFOB) and an antibacterial iridium(III) PS. Redox properties of the new conjugates have been determined at excited states and compared to that of an antibacterial iridium PS previously reported by our groups. Tested on nosocomial pathogen Pseudomonas aeruginosa and other bacteria, these conjugates demonstrated significant inhibitory activity when activated with blue LED light. Ir(III) conjugate and iridium free DFOB-2,2'-dipyridylamine ligands were crystallized in complex with FoxA, the outer membrane transporter involved in DFOB uptake in P. aeruginosa and revealed details of the binding mode of these unprecedented conjugates.


Subject(s)
Anti-Bacterial Agents , Coordination Complexes , Deferoxamine , Iridium , Light , Microbial Sensitivity Tests , Pseudomonas aeruginosa , Siderophores , Iridium/chemistry , Iridium/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Deferoxamine/pharmacology , Deferoxamine/chemistry , Deferoxamine/chemical synthesis , Siderophores/chemistry , Siderophores/pharmacology , Siderophores/chemical synthesis , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Pseudomonas aeruginosa/drug effects , Molecular Structure , Structure-Activity Relationship , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/chemical synthesis , Dose-Response Relationship, Drug
13.
Mol Pharm ; 21(9): 4324-4335, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39164886

ABSTRACT

Positron emission tomography (PET) is a powerful tool for investigating the in vivo behavior of drug delivery systems. We aimed to assess the biodistribution of extracellular vesicles (EVs), nanosized vesicles secreted by cells isolated from various human cell sources using PET. EVs were isolated from mesenchymal stromal cells (MSCs) (MSC EVs), human macrophages (Mϕ EVs), and a melanoma cell line (A375 EVs) by centrifugation and were conjugated with deferoxamine for radiolabeling with Zr-89. PET using conjugated and radiolabeled EVs evaluated their in vivo biodistribution and tissue tropisms. Our study also investigated differences in mouse models, utilizing immunocompetent and immunocompromised mice and an A375 xenograft tumor model. Lastly, we investigated the impact of different labeling techniques on the observed EV biodistribution, including covalent surface modification and membrane incorporation. PET showed that all tested EVs exhibited extended in vivo circulation and generally low uptake in the liver, spleen, and lungs. However, Mϕ EVs showed high liver uptake, potentially attributable to the intrinsic tissue tropism of these EVs from the surface protein composition. MSC EV biodistribution differed between immunocompetent and immunodeficient mice, with increased spleen uptake observed in the latter. PET using A375 xenografts demonstrated efficient tumor uptake of EVs, but no preferential tissue-specific tropism of A375 EVs was found. Biodistribution differences between labeling techniques showed that surface-conjugated EVs had preferential blood circulation and low liver, spleen, and lung uptake compared to membrane integration. This study demonstrates the potential of EVs as effective drug carriers for various diseases, highlights the importance of selecting appropriate cell sources for EV-based drug delivery, and suggests that EV tropism can be harnessed to optimize therapeutic efficacy. Our findings indicate that the cellular source of EVs, labeling technique, and animal model can influence the observed biodistribution.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Positron-Emission Tomography , Animals , Humans , Extracellular Vesicles/metabolism , Tissue Distribution , Mice , Positron-Emission Tomography/methods , Cell Line, Tumor , Mesenchymal Stem Cells/metabolism , Macrophages/metabolism , Zirconium/chemistry , Zirconium/pharmacokinetics , Deferoxamine/chemistry , Deferoxamine/pharmacokinetics , Radioisotopes/chemistry , Radioisotopes/pharmacokinetics , Drug Delivery Systems/methods , Xenograft Model Antitumor Assays , Female , Melanoma/metabolism , Melanoma/diagnostic imaging
14.
Sci Adv ; 10(33): eado3919, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39141742

ABSTRACT

Postoperative rehemorrhage following intracerebral hemorrhage surgery is intricately associated with a high mortality rate, yet there is now no effective clinical treatment. In this study, we developed a hemoglobin (Hb)-responsive in situ implantable DNA hydrogel comprising Hb aptamers cross-linked with two complementary chains and encapsulating deferoxamine mesylate (DFO). Functionally, the hydrogel generates signals upon postoperative rehemorrhage by capturing Hb, demonstrating a distinctive "self-diagnosis" capability. In addition, the ongoing capture of Hb mediates the gradual disintegration of the hydrogel, enabling the on-demand release of DFO without compromising physiological iron-dependent functions. This process achieves self-treatment by inhibiting the ferroptosis of neurocytes. In a collagenase and autologous blood injection model-induced mimic postoperative rehemorrhage model, the hydrogel exhibited a 5.58-fold increase in iron absorption efficiency, reducing hematoma size significantly (from 8.674 to 4.768 cubic millimeters). This innovative Hb-responsive DNA hydrogel not only offers a therapeutic intervention for postoperative rehemorrhage but also provides self-diagnosis feedback, holding notable promise for enhancing clinical outcomes.


Subject(s)
Cerebral Hemorrhage , Hemoglobins , Hydrogels , Cerebral Hemorrhage/diagnosis , Cerebral Hemorrhage/drug therapy , Hydrogels/chemistry , Hemoglobins/metabolism , Animals , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Deferoxamine/chemistry , DNA/metabolism , Humans , Male , Rats , Disease Models, Animal , Ferroptosis/drug effects , Iron/metabolism , Postoperative Hemorrhage/etiology , Postoperative Hemorrhage/diagnosis , Aptamers, Nucleotide/pharmacology , Aptamers, Nucleotide/chemistry
15.
Methods Enzymol ; 702: 121-145, 2024.
Article in English | MEDLINE | ID: mdl-39155108

ABSTRACT

Siderophores are low-molecular-weight organic bacterial and fungal secondary metabolites that form high affinity complexes with Fe(III). These Fe(III)-siderophore complexes are part of the siderophore-mediated Fe(III) uptake mechanism, which is the most widespread strategy used by microbes to access sufficient iron for growth. Microbial competition for limited iron is met by biosynthetic gene clusters that encode for the biosynthesis of siderophores with variable molecular scaffolds and iron binding motifs. Some classes of siderophores have well understood biosynthetic pathways, which opens opportunities to further expand structural and property diversity using precursor-directed biosynthesis (PDB). PDB involves augmenting culture medium with non-native substrates to compete against native substrates during metabolite assembly. This chapter provides background information and technical details of conducting a PDB experiment towards producing a range of different analogues of the archetypal hydroxamic acid siderophore desferrioxamine B. This includes processes to semi-purify the culture supernatant and the use of liquid chromatography-tandem mass spectrometry for downstream analysis of analogues and groups of constitutional isomers.


Subject(s)
Siderophores , Siderophores/biosynthesis , Siderophores/chemistry , Siderophores/metabolism , Tandem Mass Spectrometry/methods , Deferoxamine/metabolism , Deferoxamine/chemistry , Chromatography, Liquid/methods , Biosynthetic Pathways , Multigene Family , Iron/metabolism , Iron/chemistry , Culture Media/chemistry , Culture Media/metabolism
16.
PLoS One ; 19(8): e0306255, 2024.
Article in English | MEDLINE | ID: mdl-39121099

ABSTRACT

BACKGROUND: Iron (Fe) supplementation is a critical component of anemia therapy for patients with chronic kidney disease (CKD). However, serum Fe, ferritin, and transferrin saturation, used to guide Fe replacement, are far from optimal, as they can be influenced by malnutrition and inflammation. Currently, there is a trend of increasing Fe supplementation to target high ferritin levels, although the long-term risk has been overlooked. METHODS: We prospectively enrolled 28 patients with CKD on hemodialysis with high serum ferritin (> 1000 ng/ml) and tested the effects of 1-year deferoxamine treatment, accompanied by withdrawal of Fe administration, on laboratory parameters (Fe status, inflammatory and CKD-MBD markers), heart, liver, and iliac crest Fe deposition (quantitative magnetic resonance imaging [MRI]), and bone biopsy (histomorphometry and counting of the number of Fe positive cells in the bone marrow). RESULTS: MRI parameters showed that none of the patients had heart iron overload, but they all presented iron overload in the liver and bone marrow, which was confirmed by bone histology. After therapy, ferritin levels decreased, although neither hemoglobin levels nor erythropoietin dose was changed. A significant decrease in hepcidin and FGF-23 levels was observed. Fe accumulation was improved in the liver and bone marrow, reaching normal values only in the bone marrow. No significant changes in turnover, mineralization or volume were observed. CONCLUSIONS: Our data suggest that treatment with deferoxamine was safe and could improve Fe accumulation, as measured by MRI and histomorphometry. Whether MRI is considered a standard tool for investigating bone marrow Fe accumulation requires further investigation. Registry and the registration number of clinical trial: ReBEC (Registro Brasileiro de Ensaios Clinicos) under the identification RBR-3rnskcj available at: https://ensaiosclinicos.gov.br/pesquisador.


Subject(s)
Bone Marrow , Deferoxamine , Ferritins , Iron Overload , Iron , Liver , Renal Dialysis , Humans , Male , Female , Iron Overload/drug therapy , Iron Overload/etiology , Iron Overload/metabolism , Bone Marrow/metabolism , Bone Marrow/drug effects , Bone Marrow/pathology , Ferritins/blood , Ferritins/metabolism , Liver/metabolism , Liver/drug effects , Liver/pathology , Liver/diagnostic imaging , Middle Aged , Deferoxamine/therapeutic use , Deferoxamine/administration & dosage , Iron/metabolism , Aged , Magnetic Resonance Imaging , Prospective Studies , Renal Insufficiency, Chronic/therapy , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/blood , Fibroblast Growth Factor-23 , Hepcidins/metabolism
17.
Clin Pharmacol Drug Dev ; 13(9): 1051-1060, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38973337

ABSTRACT

Myocardial reperfusion injury (MRI) accounts for up to 50% of the final size in acute myocardial infarction and other conditions associated with ischemia-reperfusion. Currently, there is still no therapy to prevent MRI, but it is well known that oxidative stress has a key role in its mechanism. We previously reduced MRI in rats through a combined antioxidant therapy (CAT) of ascorbic acid, N-acetylcysteine, and deferoxamine. This study determines the safety and pharmacokinetics of CAT in a Phase I clinical trial. Healthy subjects (n = 18) were randomized 2:1 to CAT or placebo (NaCl 0.9% i.v.). Two different doses/infusion rates of CATs were tested in a single 90-minute intravenous infusion. Blood samples were collected at specific times for 180 minutes to measure plasma drug concentrations (ascorbic acid, N-acetylcysteine, and deferoxamine) and oxidative stress biomarkers. Adverse events were registered during infusion and followed for 30 days. Both CAT1 and CAT2 significantly increased the CAT drug concentrations compared to placebo (P < .05). Most of the pharmacokinetic parameters were similar between CAT1 and CAT2. In total, 6 adverse events were reported, all nonserious and observed in CAT1. The ferric-reducing ability of plasma (an antioxidant biomarker) increased in both CAT groups compared to placebo (P < .001). The CAT is safe in humans and a potential treatment for patients with acute myocardial infarction undergoing reperfusion therapy.


Subject(s)
Acetylcysteine , Antioxidants , Ascorbic Acid , Deferoxamine , Myocardial Reperfusion Injury , Oxidative Stress , Humans , Antioxidants/pharmacokinetics , Antioxidants/administration & dosage , Antioxidants/adverse effects , Antioxidants/pharmacology , Male , Acetylcysteine/administration & dosage , Acetylcysteine/pharmacokinetics , Acetylcysteine/adverse effects , Ascorbic Acid/administration & dosage , Ascorbic Acid/pharmacokinetics , Ascorbic Acid/adverse effects , Adult , Oxidative Stress/drug effects , Female , Deferoxamine/pharmacokinetics , Deferoxamine/administration & dosage , Deferoxamine/adverse effects , Healthy Volunteers , Young Adult , Infusions, Intravenous , Middle Aged , Double-Blind Method , Drug Therapy, Combination , Biomarkers/blood
18.
Biomed Environ Sci ; 37(6): 617-627, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38988112

ABSTRACT

Objective: The aim of this study was to explore the role and mechanism of ferroptosis in SiO 2-induced cardiac injury using a mouse model. Methods: Male C57BL/6 mice were intratracheally instilled with SiO 2 to create a silicosis model. Ferrostatin-1 (Fer-1) and deferoxamine (DFO) were used to suppress ferroptosis. Serum biomarkers, oxidative stress markers, histopathology, iron content, and the expression of ferroptosis-related proteins were assessed. Results: SiO 2 altered serum cardiac injury biomarkers, oxidative stress, iron accumulation, and ferroptosis markers in myocardial tissue. Fer-1 and DFO reduced lipid peroxidation and iron overload, and alleviated SiO 2-induced mitochondrial damage and myocardial injury. SiO 2 inhibited Nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream antioxidant genes, while Fer-1 more potently reactivated Nrf2 compared to DFO. Conclusion: Iron overload-induced ferroptosis contributes to SiO 2-induced cardiac injury. Targeting ferroptosis by reducing iron accumulation or inhibiting lipid peroxidation protects against SiO 2 cardiotoxicity, potentially via modulation of the Nrf2 pathway.


Subject(s)
Disease Models, Animal , Ferroptosis , Iron Overload , Mice, Inbred C57BL , Myocytes, Cardiac , Silicon Dioxide , Silicosis , Animals , Ferroptosis/drug effects , Male , Mice , Iron Overload/metabolism , Silicon Dioxide/toxicity , Silicosis/metabolism , Silicosis/drug therapy , Silicosis/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Deferoxamine/pharmacology , Phenylenediamines/pharmacology , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Iron/metabolism , Cyclohexylamines/pharmacology
19.
Int Immunopharmacol ; 139: 112619, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39024748

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is a prevalent age-related disease characterized by the gradual deterioration of cartilage. The involvement of chondrocyte senescence is crucial in the pathogenesis of OA. Desferoxamine (DFO) is an iron chelator with therapeutic potential in various diseases. However, the relationship of chondrocyte senescence and iron homeostasis is largely unknown. METHODS: Chondrocyte senescence was induced using tert-butyl hydroperoxide (TBHP), and the impact of DFO on chondrocyte senescence and iron metabolism was assessed through techniques such as western blotting, qRT-PCR, and ß-Galactosidase staining. To assess the impact of DFO on chondrocyte senescence and the progression of osteoarthritis (OA), the surgical destabilization of the medial meniscus model was established. RESULTS: In chondrocytes, TBHP administration resulted in elevated expression of P16, P21, and P53, as well as alterations in SA-ß-gal staining. Nevertheless, DFO effectively mitigated chondrocyte senescence induced by TBHP, and reversed the decrease in collagen II expression and increase in MMP13 expression caused by TBHP. Mechanismly, TBHP induced NCOA4 expression and iron release in chondrocytes. Excessive iron could induce chondrocyte senescence, whereas, DFO could inhibit NCOA4 expression and restore ferritin level, and chelate excessive iron. Importantly, intra-articular injection of DFO enhanced collagen II expression and reduced expression of P16, P21, and MMP13 of cartilage in OA mice, and delayed cartilage degeneration. CONCLUSIONS: Overall, this study provides evidence that DFO has the potential to alleviate chondrocyte senescence induced by TBHP and slow down the progression of osteoarthritis (OA) by effectively chelating excessive iron. These findings suggest that iron chelation could be a promising therapeutic strategy for treating OA.


Subject(s)
Cellular Senescence , Chondrocytes , Deferoxamine , Homeostasis , Iron , Osteoarthritis , Chondrocytes/drug effects , Chondrocytes/metabolism , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Animals , Osteoarthritis/drug therapy , Osteoarthritis/pathology , Osteoarthritis/metabolism , Iron/metabolism , Cellular Senescence/drug effects , Homeostasis/drug effects , Mice , Cells, Cultured , Male , Mice, Inbred C57BL , Disease Progression , tert-Butylhydroperoxide , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 13/genetics , Humans , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Disease Models, Animal
20.
ACS Appl Mater Interfaces ; 16(33): 43892-43906, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39046193

ABSTRACT

With the swift evolution of multidrug-resistant bacteria resulting from the intense and inappropriate use of antibiotics, there is a pressing need for innovative solutions. In this study, a thermosensitive hydrogel was developed for efficient bacterial inhibition and promotion of wound healing. The antibacterial chitosan (CS) thermosensitive hydrogel, cross-linked with two-dimensional photothermal nanomaterial black phosphorus (BP) nanosheets through electrostatic interactions, effectively encapsulates and sustains the release of angiogenic drug deferoxamine mesylate (DFO). This facilitates the acceleration of re-epithelialization and neovascularization by enhancing cell migration and proliferation. Following near-infrared (NIR) treatment, this hydrogel demonstrates rapid eradication of the most common multidrug-resistant bacteria encountered in clinical settings, achieved through physical disruption of bacterial membranes and photothermal therapies. Noteworthy is the significant upregulation of IL-19 expression via STAT3 signaling pathways by the BP/CS-DFO hydrogel in a full-thickness wound model. This results in the polarization of the anti-inflammatory M2 macrophage phenotype, altering the microenvironment to a pro-healing state and enhancing extracellular matrix deposition and blood vessel formation. In conclusion, the BP/CS-DFO hydrogel shows immense promise as a potential clinical candidate for wound healing and antimicrobial therapy. Its innovative design and multifunctional capabilities position it as a valuable asset in combating antibiotic resistance and enhancing efficiency in wound healing.


Subject(s)
Anti-Bacterial Agents , Chitosan , Deferoxamine , Hydrogels , Phosphorus , Wound Healing , Wound Healing/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Phosphorus/chemistry , Mice , Animals , Chitosan/chemistry , Chitosan/pharmacology , Deferoxamine/chemistry , Deferoxamine/pharmacology , Nanostructures/chemistry , RAW 264.7 Cells , Microbial Sensitivity Tests , Humans , Staphylococcus aureus/drug effects , Cell Proliferation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL