Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.110
1.
Molecules ; 29(9)2024 Apr 29.
Article En | MEDLINE | ID: mdl-38731540

Deferoxamine, an iron chelator used to treat diseases caused by excess iron, has had a Food and Drug Administration-approved status for many years. A large number of studies have confirmed that deferoxamine can reduce inflammatory response and promote angiogenesis. Blood vessels play a crucial role in sustaining vital life by facilitating the delivery of immune cells, oxygen, and nutrients, as well as eliminating waste products generated during cellular metabolism. Dysfunction in blood vessels may contribute significantly to the development of life-threatening diseases. Anti-angiogenesis therapy and pro-angiogenesis/angiogenesis strategies have been frequently recommended for various diseases. Herein, we describe the mechanism by which deferoxamine promotes angiogenesis and summarize its application in chronic wounds, bone repair, and diseases of the respiratory system. Furthermore, we discuss the drug delivery system of deferoxamine for treating various diseases, providing constructive ideas and inspiration for the development of new treatment strategies.


Deferoxamine , Neovascularization, Physiologic , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Humans , Animals , Neovascularization, Physiologic/drug effects , Regeneration/drug effects , Wound Healing/drug effects , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Angiogenesis
2.
ACS Appl Mater Interfaces ; 16(19): 24384-24397, 2024 May 15.
Article En | MEDLINE | ID: mdl-38709640

Vascularization and inflammation management are essential for successful bone regeneration during the healing process of large bone defects assisted by artificial implants/fillers. Therefore, this study is devoted to the optimization of the osteogenic microenvironment for accelerated bone healing through rapid neovascularization and appropriate inflammation inhibition that were achieved by applying a tantalum oxide (TaO)-based nanoplatform carrying functional substances at the bone defect. Specifically, TaO mesoporous nanospheres were first constructed and then modified by functionalized metal ions (Mg2+) with the following deferoxamine (DFO) loading to obtain the final product simplified as DFO-Mg-TaO. Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) revealed that the product was homogeneously dispersed hollow nanospheres with large specific surface areas and mesoporous shells suitable for loading Mg2+ and DFO. The biological assessments indicated that DFO-Mg-TaO could enhance the adhesion, proliferation, and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). The DFO released from DFO-Mg-TaO promoted angiogenetic activity by upregulating the expressions of hypoxia-inducible factor-1 (HIF-1α) and vascular endothelial growth factor (VEGF). Notably, DFO-Mg-TaO also displayed anti-inflammatory activity by reducing the expressions of pro-inflammatory factors, benefiting from the release of bioactive Mg2+. In vivo experiments demonstrated that DFO-Mg-TaO integrated with vascular regenerative, anti-inflammatory, and osteogenic activities significantly accelerated the reconstruction of bone defects. Our findings suggest that the optimized DFO-Mg-TaO nanospheres are promising as multifunctional fillers to speed up the bone healing process.


Bone Regeneration , Deferoxamine , Magnesium , Mesenchymal Stem Cells , Oxides , Tantalum , Deferoxamine/chemistry , Deferoxamine/pharmacology , Bone Regeneration/drug effects , Tantalum/chemistry , Animals , Oxides/chemistry , Oxides/pharmacology , Magnesium/chemistry , Magnesium/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Neovascularization, Physiologic/drug effects , Rats , Mice , Rats, Sprague-Dawley , Cell Proliferation/drug effects , Angiogenesis
3.
Biomed Mater ; 19(4)2024 May 21.
Article En | MEDLINE | ID: mdl-38697149

To effectively address underlying issues and enhance the healing process of hard-to-treat soft tissue defects, innovative therapeutic approaches are required. One promising strategy involves the incorporation of bioactive substances into biodegradable scaffolds to facilitate synergistic tissue regeneration, particularly in vascular regeneration. In this study, we introduce a composite hydrogel design that mimics the extracellular matrix by covalently combining gelatin and hyaluronic acid (HA), with the encapsulation of deferoxamine nanoparticles (DFO NPs) for potential tissue regeneration applications. Crosslinked hydrogels were fabricated by controlling the ratio of HA in the gelatin-based hydrogels, resulting in improved mechanical properties, enhanced degradation ability, and optimised porosity, compared with hydrogel formed by gelatin alone. The DFO NPs, synthesized using a double emulsion method with poly (D,L-lactide-co-glycolide acid), exhibited a sustained release of DFO over 12 d. Encapsulating the DFO NPs in the hydrogel enabled controlled release over 15 d. The DFO NPs, composite hydrogel, and the DFO NPs loaded hydrogel exhibited excellent cytocompatibility and promoted cell proliferationin vitro. Subcutaneous implantation of the composite hydrogel and the DFO NPs loaded hydrogel demonstrated biodegradability, tissue integration, and no obvious adverse effects, evidenced by histological analysis. Furthermore, the DFO NPs loaded composite hydrogel exhibited accelerated wound closure and promoted neovascularisation and granular formation when tested in an excisional skin wound model in mice. These findings highlight the potential of our composite hydrogel system for promoting the faster healing of diabetes-induced skin wounds and oral lesions through its ability to modulate tissue regeneration processes.


Biomimetic Materials , Deferoxamine , Gelatin , Hyaluronic Acid , Hydrogels , Nanoparticles , Gelatin/chemistry , Deferoxamine/chemistry , Deferoxamine/pharmacology , Animals , Hydrogels/chemistry , Hyaluronic Acid/chemistry , Nanoparticles/chemistry , Mice , Biomimetic Materials/chemistry , Cell Proliferation/drug effects , Wound Healing/drug effects , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Biocompatible Materials/chemistry , Humans , Porosity , Regeneration , Biomimetics
4.
J Cell Mol Med ; 28(8): e18306, 2024 Apr.
Article En | MEDLINE | ID: mdl-38613357

Topical patch delivery of deferoxamine (DFO) has been studied as a treatment for this fibrotic transformation in irradiated tissue. Efficacy of a novel cream formulation of DFO was studied as a RIF therapeutic in unwounded and excisionally wounded irradiated skin. C57BL/6J mice underwent 30 Gy of radiation to the dorsum followed by 4 weeks of recovery. In a first experiment, mice were separated into six conditions: DFO 50 mg cream (D50), DFO 100 mg cream (D100), soluble DFO injections (DI), DFO 1 mg patch (DP), control cream (Vehicle), and irradiated untreated skin (IR). In a second experiment, excisional wounds were created on the irradiated dorsum of mice and then divided into four treatment groups: DFO 100 mg Cream (W-D100), DFO 1 mg patch (W-DP), control cream (W-Vehicle), and irradiated untreated wounds (W-IR). Laser Doppler perfusion scans, biomechanical testing, and histological analysis were performed. In irradiated skin, D100 improved perfusion compared to D50 or DP. Both D100 and DP enhanced dermal characteristics, including thickness, collagen density and 8-isoprostane staining compared to untreated irradiated skin. D100 outperformed DP in CD31 staining, indicating higher vascular density. Extracellular matrix features of D100 and DP resembled normal skin more closely than DI or control. In radiated excisional wounds, D100 facilitated faster wound healing and increased perfusion compared to DP. The 100 mg DFO cream formulation rescued RIF of unwounded irradiated skin and improved excisional wound healing in murine skin relative to patch delivery of DFO.


Deferoxamine , Radiation Fibrosis Syndrome , Mice , Animals , Mice, Inbred C57BL , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Skin , Perfusion
5.
J Neurosci Res ; 102(4): e25334, 2024 Apr.
Article En | MEDLINE | ID: mdl-38656648

Iron deficiency (ID) has been shown to affect central nervous system (CNS) development and induce hypomyelination. Previous work from our laboratory in a gestational ID model showed that both oligodendrocyte (OLG) and astrocyte (AST) maturation was impaired. To explore the contribution of AST iron to the myelination process, we generated an in vitro ID model by silencing divalent metal transporter 1 (DMT1) in AST (siDMT1 AST) or treating AST with Fe3+ chelator deferoxamine (DFX; DFX AST). siDMT1 AST showed no changes in proliferation but remained immature. Co-cultures of oligodendrocyte precursors cells (OPC) with siDMT1 AST and OPC cultures incubated with siDMT1 AST-conditioned media (ACM) rendered a reduction in OPC maturation. These findings correlated with a decrease in the expression of AST-secreted factors IGF-1, NRG-1, and LIF, known to promote OPC differentiation. siDMT1 AST also displayed increased mitochondrial number and reduced mitochondrial size as compared to control cells. DFX AST also remained immature and DFX AST-conditioned media also hampered OPC maturation in culture, in keeping with a decrease in the expression of AST-secreted growth factors IGF-1, NRG-1, LIF, and CNTF. DFX AST mitochondrial morphology and number showed results similar to those observed in siDMT1 AST. In sum, our results show that ID, induced through two different methods, impacts AST maturation and mitochondrial functioning, which in turn hampers OPC differentiation.


Astrocytes , Cell Differentiation , Iron Deficiencies , Oligodendroglia , Astrocytes/metabolism , Astrocytes/drug effects , Oligodendroglia/metabolism , Oligodendroglia/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Cation Transport Proteins/metabolism , Coculture Techniques , Culture Media, Conditioned/pharmacology , Rats , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/metabolism , Deferoxamine/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Iron/metabolism
6.
Environ Sci Technol ; 58(8): 3974-3984, 2024 Feb 27.
Article En | MEDLINE | ID: mdl-38306233

In contaminated water and soil, little is known about the role and mechanism of the biometabolic molecule siderophore desferrioxamine-B (DFO) in the biogeochemical cycle of uranium due to complicated coordination and reaction networks. Here, a joint experimental and quantum chemical investigation is carried out to probe the biomineralization of uranyl (UO22+, referred to as U(VI) hereafter) induced by Shewanella putrefaciens (abbreviated as S. putrefaciens) in the presence of DFO and Fe3+ ion. The results show that the production of mineralized solids {hydrogen-uranium mica [H2(UO2)2(PO4)2·8H2O]} via S. putrefaciens binding with UO22+ is inhibited by DFO, which can both chelate preferentially UO22+ to form a U(VI)-DFO complex in solution and seize it from U(VI)-biominerals upon solvation. However, with Fe3+ ion introduced, the strong specificity of DFO binding with Fe3+ causes re-emergence of biomineralization of UO22+ {bassetite [Fe(UO2)2(PO4)2·8(H2O)]} by S. putrefaciens, owing to competitive complexation between Fe3+ and UO22+ for DFO. As DFO possesses three hydroxamic functional groups, it forms hexadentate coordination with Fe3+ and UO22+ ions via these functional groups. The stability of the Fe3+-DFO complex is much higher than that of U(VI)-DFO, resulting in some DFO-released UO22+ to be remobilized by S. putrefaciens. Our finding not only adds to the understanding of the fate of toxic U(VI)-containing substances in the environment and biogeochemical cycles in the future but also suggests the promising potential of utilizing functionalized DFO ligands for uranium processing.


Shewanella putrefaciens , Uranium , Biomineralization , Deferoxamine/metabolism , Deferoxamine/pharmacology , Shewanella putrefaciens/metabolism , Siderophores/metabolism , Siderophores/pharmacology , Uranium/chemistry , Iron Compounds/chemistry
7.
Int Immunopharmacol ; 129: 111662, 2024 Mar 10.
Article En | MEDLINE | ID: mdl-38340421

This study aimed to examine the effects of the secretome released by human umbilical cord-mesenchymal stem cells (MSC) as a result of preconditioning with deferoxamine (DFX), a hypoxia mimetic agent, on type 1 diabetes (T1D), by comparing it with the secretome produced by untreated MSCs. Initially, the levels of total protein, IL4, IL10, IL17, and IFNγ in the conditioned medium (CM) obtained from MSCs subjected to preconditioning with 150 µM DFX (DFX-CM) were analyzed in comparison to CM derived from untreated MSCs (N-CM). Subsequently, the CMs were administered to rats with T1D within a specific treatment plan. Following the sacrification, immunomodulation was evaluated by measuring serum cytokine levels and assessing the regulatory T cell (Treg) ratio in spleen mononuclear cells. Additionally, ß-cell mass was determined in the islets by immunohistochemical labeling of NK6 Homeobox 1 (Nkx6.1), Pancreatic duodenal homeobox-1 (Pdx1), and insulin antibodies in pancreatic sections. In vitro findings indicated that the secretome levels of MSCs were enhanced by preconditioning with DFX. In vivo, the use of DFX-CM significantly increased the Treg population, and accordingly, the level of inflammatory cytokines decreased. In ß-cell marker labeling, D + DFX-CM showed significantly increased PDX1 and insulin immunoreactivity. In conclusion, while the factors released by MSCs without external stimulation had limited therapeutic effects, substantial improvements in immunomodulation and ß-cell regeneration were seen with DFX-preconditioned cell-derived CM.


Diabetes Mellitus, Type 1 , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Rats , Humans , Animals , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Diabetes Mellitus, Type 1/therapy , Secretome , Cytokines/metabolism , Mesenchymal Stem Cells/metabolism , Immunomodulation , Umbilical Cord , Regeneration
8.
Adv Healthc Mater ; 13(12): e2303134, 2024 May.
Article En | MEDLINE | ID: mdl-38348511

The effective repair of large bone defects remains a major challenge due to its limited self-healing capacity. Inspired by the structure and function of the natural periosteum, an electrospun biomimetic periosteum is constructed to programmatically promote bone regeneration using natural bone healing mechanisms. The biomimetic periosteum is composed of a bilayer with an asymmetric structure in which an aligned electrospun poly(ε-caprolactone)/gelatin/deferoxamine (PCL/GEL/DFO) layer mimics the outer fibrous layer of the periosteum, while a random coaxial electrospun PCL/GEL/aspirin (ASP) shell and PCL/silicon nanoparticles (SiNPs) core layer mimics the inner cambial layer. The bilayer controls the release of ASP, DFO, and SiNPs to precisely regulate the inflammatory, angiogenic, and osteogenic phases of bone repair. The random coaxial inner layer can effectively antioxidize, promoting cell recruitment, proliferation, differentiation, and mineralization, while the aligned outer layer can promote angiogenesis and prevent fibroblast infiltration. In particular, different stages of bone repair are modulated in a rat skull defect model to achieve faster and better bone regeneration. The proposed biomimetic periosteum is expected to be a promising candidate for bone defect healing.


Biomimetic Materials , Bone Regeneration , Periosteum , Polyesters , Bone Regeneration/drug effects , Animals , Periosteum/drug effects , Rats , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Polyesters/chemistry , Rats, Sprague-Dawley , Deferoxamine/pharmacology , Deferoxamine/chemistry , Gelatin/chemistry , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Delayed-Action Preparations/pharmacokinetics , Osteogenesis/drug effects , Skull/drug effects , Skull/injuries , Male , Nanoparticles/chemistry , Tissue Engineering/methods , Cell Differentiation/drug effects , Tissue Scaffolds/chemistry
9.
Int J Mol Sci ; 25(2)2024 Jan 11.
Article En | MEDLINE | ID: mdl-38255991

Deferoxamine (DFO) is a water-soluble iron chelator used pharmacologically for the management of patients with transfusional iron overload. However, DFO is not cell-permeable and has a short plasma half-life, which necessitates lengthy parenteral administration with an infusion pump. We previously reported the synthesis of chitosan (CS) nanoparticles for sustained slow release of DFO. In the present study, we developed solid dispersions and nanoparticles of a carboxymethyl water-soluble chitosan derivative (CMCS) for improved DFO encapsulation and release. CS dispersions and nanoparticles with DFO have been prepared by ironical gelation using sodium triphosphate (TPP) and were examined for comparison purposes. The successful presence of DFO in CMCS polymeric dispersions and nanoparticles was confirmed through FTIR measurements. Furthermore, the formation of CMCS nanoparticles led to inclusion of DFO in an amorphous state, while dispersion of DFO in the polymeric matrix led to a decrease in its crystallinity according to X-ray diffraction (XRD) and differential scanning calorimetry (DSC) results. An in vitro release assay indicated sustained release of DFO from CS and CMCS nanoparticles over 48 h and 24 h, respectively. Application of CMCS-DFO dispersions to murine RAW 264.7 macrophages or human HeLa cervical carcinoma cells triggered cellular responses to iron deficiency. These were exemplified in the induction of the mRNA encoding transferrin receptor 1, the major iron uptake protein, and the suppression of ferritin, the iron storage protein. Our data indicate that CMCS-DFO nanoparticles release bioactive DFO that causes effective iron chelation in cultured cells.


Chitosan , Humans , Animals , Mice , Deferoxamine/pharmacology , Chelating Agents , Biological Transport , Iron
10.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167024, 2024 03.
Article En | MEDLINE | ID: mdl-38242180

Oxidative stress is the common mechanism of sensorineural hearing loss (SNHL) caused by many factors, such as noise, drugs and ageing. Here, we used tert-butyl hydroperoxide (t-BHP) to cause oxidative stress damage in HEI-OC1 cells and in an in vitro cochlear explant model. We observed lipid peroxidation, iron accumulation, mitochondrial shrinkage and vanishing of mitochondrial cristae, which caused hair cell ferroptosis, after t-BHP exposure. Moreover, the number of TUNEL-positive cells in cochlear explants and HEI-OC1 cells increased significantly, suggesting that t-BHP caused the apoptosis of hair cells. Administration of deferoxamine (DFOM) significantly attenuated t-BHP-induced hair cell loss and disordered hair cell arrangement in cochlear explants as well as HEI-OC1 cell death, including via apoptosis and ferroptosis. Mechanistically, we found that DFOM treatment reduced t-BHP-induced lipid peroxidation, iron accumulation and mitochondrial pathological changes in hair cells, consequently mitigating apoptosis and ferroptosis. Moreover, DFOM treatment alleviated GSH depletion caused by t-BHP and activated the Nrf2 signalling pathway to exert a protective effect. Furthermore, we confirmed that the protective effect of DFOM mainly depended on its ability to chelate iron by constructing Fth1 knockout (KO), TfR1 KO and Nrf2 KO HEI-OC1 cell lines using CRISPR/Cas9 technology and a Flag-Fth1 (overexpression) HEI-OC1 cell line using the FlpIn™ System. Our findings suggest that DFOM is a potential drug for SNHL treatment due to its ability to inhibit apoptosis and ferroptosis by chelating iron and scavenging reactive oxygen species (ROS).


Deferoxamine , Ototoxicity , Humans , tert-Butylhydroperoxide/toxicity , tert-Butylhydroperoxide/metabolism , Deferoxamine/pharmacology , Ototoxicity/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Hair Cells, Auditory/metabolism , Iron/metabolism
11.
Transplant Proc ; 56(1): 223-227, 2024.
Article En | MEDLINE | ID: mdl-38199859

The University of Wisconsin (UW) solution is the most effective preservation solution currently used; however, to safely use expanded-criteria donor grafts, a new cold storage solution that alleviates graft injury more effectively is required. We prepared a heavy water (D2O)-containing buffer, Dsol, and observed strong protective effects during extended cold storage of rat hearts and livers. In the current study, we modified Dsol (mDsol) and tested its efficacy. The aim of the present study was to determine whether mDsol could protect the rat liver more effectively than the UW solution and to clarify the roles of D2O and deferoxamine (DFX). Rat livers were subjected to cold storage for 48 hours in test solutions: UW, mDsol, mDsol without D2O or DFX (mDsol-D2O[-], mDsol-DFX[-]), and subsequently reperfused on an isolated perfused rat liver for 90 minutes at 37°C. In the UW group, the liver was dehydrated during cold storage and rapidly expanded during reperfusion. Accordingly, the cumulative weight change was the highest in the UW group, together with augmented portal veinous resistance and ALT leakage and decreased oxygen consumption rate and bile production. These changes were significantly suppressed in the mDsol-treated group. In the mDsol-D2O(-) and mDsol-DFX(-) groups offered partial protection. In conclusion, mDsol appeared to be superior to the UW solution for simple cold storage of the rat liver, presumably due to improved microcirculation in the early phase of reperfusion. Both heavy water and deferoxamine are essential for alleviating seamless organ swelling that occurs during cold storage and subsequent reperfusion.


Liver Transplantation , Organ Preservation Solutions , Humans , Rats , Animals , Deuterium Oxide/pharmacology , Deferoxamine/pharmacology , Liver , Organ Preservation Solutions/pharmacology , Reperfusion , Glutathione/pharmacology , Allopurinol/pharmacology , Insulin/pharmacology , Raffinose/pharmacology , Organ Preservation , Adenosine
12.
Brain Res Bull ; 207: 110878, 2024 Feb.
Article En | MEDLINE | ID: mdl-38218407

Cerebrovascular dysfunction resulting from traumatic brain injury (TBI) significantly contributes to poor patient outcomes. Recent studies revealed the involvement of iron metabolism in neuronal survival, yet its effect on vasculature remains unclear. This study aims to explore the impact of endothelial ferroptosis on cerebrovascular function in TBI. A Controlled Cortical Impact (CCI) model was established in mice, resulting in a significant increase in iron-related proteins such as TfR1, FPN1, and FTH, as well as oxidative stress biomarker 4HNE. This was accompanied by a decline in expression of the ferroptosis inhibitor GPX4. Moreover, Perls' staining and nonhemin iron content assay showed iron overload in brain microvascular endothelial cells (BMECs) and the ipsilateral cortex. Immunofluorescence staining revealed more FTH-positive cerebral endothelial cells, consistent with impaired perfusion vessel density and cerebral blood flow. As a specific iron chelator, deferoxamine (DFO) treatment inhibited such ferroptotic proteins expression and the accumulation of lipid-reactive oxygen species following CCI, enhancing glutathione peroxidase (GPx) activity. DFO treatment significantly reduced iron deposition in BMECs and brain tissue, and increased density of the cerebral capillaries as well. Consequently, DFO treatment led to improvements in cerebral blood flow (as measured by laser speckle imaging) and behavioral performance (as measured by the neurological severity scores, rotarod test, and Morris water maze test). Taken together, our results indicated that TBI induces remarkable iron disorder and endothelial ferroptosis, and DFO treatment may help maintain iron homeostasis and protect vascular function. This may provide a novel therapeutic strategy to prevent cerebrovascular dysfunction following TBI.


Brain Injuries, Traumatic , Ferroptosis , Humans , Mice , Animals , Deferoxamine/pharmacology , Endothelial Cells/metabolism , Ferroptosis/physiology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Iron/metabolism
13.
Adv Healthc Mater ; 13(8): e2303000, 2024 Mar.
Article En | MEDLINE | ID: mdl-38063809

Inducing cell migration from the edges to the center of a wound, promoting angiogenesis, and controlling bacterial infection are very important for diabetic wound healing. Incorporating growth factors and antibiotics into hydrogels for wound dressing is considered a potential strategy to meet these requirements. However, some present drawbacks greatly slow down their development toward application, such as the short half-life and high price of growth factors, low antibiotic efficiency against drug-resistant bacteria, insufficient ability of hydrogels to promote cell migration, etc. Deferoxamine (DFO) can upregulate the expression of HIF-1α, thus stimulating the secretion of angiogenesis-related endogenous growth factors. Copper sulfide (CuS) nanoparticles possess excellent antibacterial performance combined with photothermal therapy (PTT). Herein, DFO and CuS nanoparticles are incorporated into a biomimetic hydrogel, which mimics the structure and function of the extracellular matrix (ECM), abbreviated as DFO/CuS-ECMgel. This biomimetic hydrogel is expected to be able to promote cell adhesion and migration, be degraded by cell-secreted matrix metalloproteinases (MMPs), and then release DFO and CuS nanoparticles at the wound site to exert their therapeutic effects. As a result, the three crucial requirements for diabetic wound healing, "beneficial for cell adhesion and migration, promoting angiogenesis, effectively killing drug-resistant bacteria," can be achieved simultaneously.


Diabetes Mellitus , Nanoparticles , Humans , Hydrogels/chemistry , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Copper/chemistry , Photothermal Therapy , Biomimetics , Nanoparticles/chemistry , Diabetes Mellitus/drug therapy , Anti-Bacterial Agents/chemistry
14.
Eur J Pharmacol ; 960: 176153, 2023 Dec 05.
Article En | MEDLINE | ID: mdl-38059446

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.


Deferoxamine , Iron Overload , Mice , Male , Animals , Deferoxamine/pharmacology , Catalase , Antioxidants/pharmacology , Antioxidants/therapeutic use , Iron Overload/drug therapy , Iron Overload/pathology , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Iron , Superoxide Dismutase
15.
Shanghai Kou Qiang Yi Xue ; 32(4): 356-362, 2023 Aug.
Article Zh | MEDLINE | ID: mdl-38044727

PURPOSE: To explore the effect of sodium alginate-g-deferoxamine/chitosan (SA-g-DFO/CS) microspheres on proliferation and osteogenic differentiation of rat bone mesenchymal stem cells (BMSCs). METHODS: A kind of SA-g-DFO/CS microsphere was developed through electrostatic interaction between porous chitosan microspheres and sodium alginate chemically grafted on the surface of DFO. Its morphology, porosity rate, pore size and sustained release of DFO in vitro were examined. Rat BMSCs were isolated and co-cultured with microspheres in osteogenic differentiation medium. MTT assay was used to study the influence of cell proliferation, and Calcein-AM/PI staining was used to observe the cell viability. Alkaline phosphatase (ALP) activity assay was conducted. PCR was used to detect the expression of genes related to angiogenesis and osteogenesis. Statistical analysis was performed using SPSS 22.0 software package. RESULTS: The SA-g-DFO/CS porous microspheres were successfully prepared with a sustained re6lease of DFO. Compared with SA/CS microspheres, the SA-g-DFO/CS microspheres were conducive to cell proliferation and differentiation, with the increases in expression level of ALP, related angiogenesis genes HIF-1α, VEGF and osteogenesis genes COLI, OCN. CONCLUSIONS: The SA-g-DFO/CS porous microspheres can provide a new choice for the development of alveolar bone regeneration.


Chitosan , Mesenchymal Stem Cells , Rats , Animals , Osteogenesis/genetics , Deferoxamine/pharmacology , Deferoxamine/metabolism , Microspheres , Cell Differentiation , Mesenchymal Stem Cells/metabolism , Alginates/pharmacology , Cells, Cultured
16.
PLoS One ; 18(12): e0295257, 2023.
Article En | MEDLINE | ID: mdl-38100448

Research into marine iron cycles and biogeochemistry has commonly relied on the use of chelators (including siderophores) to manipulate iron bioavailability. To test whether a commonly used chelator, desferrioxamine B (DFB) caused effects beyond changing the iron-status of cells, cultures of the environmentally relevant marine heterotrophic bacterium, Ruegeria pomeroyii, were grown in media with different concentrations of iron and/or DFB, resulting in a gradient of iron availability. To determine how cells responded, transcriptomes were generated for cells from the different treatments and analyzed to determine how cells reacted to these to perturbations. Analyses were also performed to look for cellular responses specific to the presence of DFB in the culture medium. As expected, cells experiencing different levels of iron availability had different transcriptomic profiles. While many genes related to iron acquisition were differentially expressed between treatments, there were many other genes that were also differentially expressed between different sample types, including those related to the uptake and metabolism of other metals as well as genes related to metabolism of other types of molecules like amino acids and carbohydrates. We conclude that while DFB certainly altered iron availability to cells, it also appears to have had a general effect on the homeostasis of other metals as well as influenced metabolic processes outside of metal acquisition.


Deferoxamine , Iron , Iron/metabolism , Deferoxamine/pharmacology , Deferoxamine/metabolism , Siderophores/genetics , Metals , Chelating Agents
17.
Sci Rep ; 13(1): 20145, 2023 11 17.
Article En | MEDLINE | ID: mdl-37978208

Retinal ischemia‒reperfusion (I/R) injury can cause significant damage to human retinal neurons, greatly compromising their functions. Existing interventions have been proven to have little effect. Ferroptosis is a newly discovered type of programmed cell death that has been found to be involved in the process of ischemia‒reperfusion in multiple organs throughout the body. Studies have shown that it is also present in retinal ischemia‒reperfusion injury. A rat model of retinal ischemia‒reperfusion injury was constructed and treated with deferoxamine. In this study, we found the accumulation of Fe2+, reactive oxygen species (ROS), malondialdehyde (MDA), and the consumption of glutathione (GSH) via ELISA testing; increased expression of transferrin; and decreased expression of ferritin, SLC7A11, and GPX4 via Western blotting (WB) and real-time PCR testing. Structural signs of ferroptosis (mitochondrial shrinkage) were observed across multiple cell types, including retinal ganglion cells (RGCs), photoreceptor cells, and pigment epithelial cells. Changes in visual function were detected by F-VEP and ERG. The results showed that iron and oxidative stress were increased in the retinal ischemia‒reperfusion injury model, resulting in ferroptosis and tissue damage. Deferoxamine protects the structural and functional soundness of the retina by inhibiting ferroptosis through the simultaneous inhibition of hemochromatosis, the initiation of transferrin, and the degradation of ferritin and activating the antioxidant capacity of the System Xc-GSH-GPX4 pathway.


Ferroptosis , Reperfusion Injury , Vision, Low , Humans , Animals , Rats , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Reperfusion , Reperfusion Injury/complications , Reperfusion Injury/drug therapy , Ferritins , Glutathione , Transferrins , Reactive Oxygen Species
18.
Nan Fang Yi Ke Da Xue Xue Bao ; 43(9): 1577-1584, 2023 Sep 20.
Article Zh | MEDLINE | ID: mdl-37814872

OBJECTIVE: To evaluate the effect of deferoxamine (DFO) on bone marrow hematopoietic function in C57 mice exposed to a sublethal dose of X-ray irradiation. METHODS: C57 mice exposed to a sublethal dose (5.4 Gy, 1.0 Gy/min) of total body X-ray irradiation (TBI) were treated with subcutaneous injection of 100 mg/kg DFO, with normal saline as the control, on a daily basis for 10 and 20 consecutive days. Body weight changes of the mice were monitored every 3 days. Five mice were selected from each group at 10 and 20 days for examination of blood cell counts, bone marrow nucleated cell counts, percentage of bone marrow CD34+ cells, bone marrow pathology, and expressions of cleaved PARP-1, cleaved caspase-3, VEGF, GPX4, and SLC7A11 in the nucleated cells. RESULTS: The body weight of the mice decreased significantly on day 3 in TBI and DFO groups (P<0.05), and to the lowest on day 6 in TBI group (P<0.01). Blood cell counts and bone marrow nucleated cell counts of the mice were significantly decreased at 10 and 20 days following TBI (P<0.01). On day 10 following TBI, the mice showed significantly decreased nucleated cells and the presence of adipocytes in the bone marrow, where increased expressions of cleaved PARP-1 and cleaved caspase-3 and lowered expressions of GPX4 and SLC7A11 were detected in the nucleated cells (P<0.05). In the mice exposed to TBI, treatment with DFO significantly increased CD34+ cell percentage (P<0.001), decreased the expressions of cleaved PARP-1 and cleaved caspase-3, and increased the expressions of GPX4, SLC7A11 and VEGF in the bone marrow nucleated cells (P<0.05). DFO treatment significantly increased blood cell counts and bone marrow nucleated cells in mice at 20 days following TBI (P<0.05). CONCLUSION: DFO improves bone marrow hematopoiesis in mice with sublethal-dose TBI by inhibiting apoptosis and ferroptosis of bone marrow nucleated cells and promoting VEGF expression and CD34+ cell proliferation.


Bone Marrow , Deferoxamine , Mice , Animals , Caspase 3 , Deferoxamine/pharmacology , X-Rays , Poly(ADP-ribose) Polymerase Inhibitors , Vascular Endothelial Growth Factor A , Whole-Body Irradiation , Bone Marrow Cells , Body Weight
19.
Toxicol Appl Pharmacol ; 479: 116727, 2023 11 15.
Article En | MEDLINE | ID: mdl-37863361

Iron overload cardiomyopathy (IOC) is the leading cause of death in cases of iron overload in patients. Previous studies demonstrated that iron overload led to cardiomyocyte dysfunction and death through multiple pathways including apoptosis, necroptosis and ferroptosis. However, the dominant cell death pathway in the iron-overloaded heart needs clarification. We tested the hypothesis that ferroptosis, an iron-dependent cell death, plays a dominant role in IOC, and ferroptosis inhibitor exerts greater efficacy than inhibitors of apoptosis and necroptosis on improving cardiac function in iron-overloaded rats. Iron dextran was injected intraperitoneally into male Wistar rats for four weeks to induce iron overload. Then, the rats were divided into 5 groups: treated with vehicle, apoptosis inhibitor (z-VAD-FMK), necroptosis inhibitor (Necrostatin-1), ferroptosis inhibitor (Ferrostatin-1) or iron chelator (deferoxamine) for 2 weeks. Cardiac function, mitochondrial function, apoptosis, necroptosis and ferroptosis were determined. The increased expression of apoptosis-, necroptosis- and ferroptosis-related proteins, were associated with impaired cardiac and mitochondrial function in iron-overloaded rats. All cell death inhibitors attenuated cardiac apoptosis, necroptosis and ferroptosis in iron-overloaded rats. Ferrostatin-1 was more effective than the other drugs in diminishing mitochondrial dysfunction and Bax/Bcl-2 ratio. Moreover, both Ferrostatin-1 and deferoxamine reversed iron overload-induced cardiac dysfunction as indicated by restored left ventricular ejection fraction and E/A ratio, whereas z-VAD-FMK and Necrostatin-1 only partially improved this parameter. These results indicated that ferroptosis could be the predominant form of cardiomyocyte death in IOC, and that inhibiting ferroptosis might be a potential novel treatment for IOC.


Cardiomyopathies , Ferroptosis , Iron Overload , Rats , Humans , Male , Animals , Deferoxamine/metabolism , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Necroptosis , Stroke Volume , Rats, Wistar , Ventricular Function, Left , Apoptosis , Iron Overload/drug therapy , Iron Overload/metabolism , Iron/metabolism , Cardiomyopathies/drug therapy , Cardiomyopathies/prevention & control , Cardiomyopathies/chemically induced , Mitochondria , Myocytes, Cardiac/metabolism
20.
Reprod Domest Anim ; 58(12): 1702-1711, 2023 Dec.
Article En | MEDLINE | ID: mdl-37818658

The aim of this study was to evaluate the effect of antioxidant supplementation in diluted semen from Muscovy drakes after the induction of oxidative stress (OS) on the sperm motility, kinematic parameters and biochemical markers - lipid peroxidation (LPO) levels and total glutathione (tGSH) concentration. The pooled semen was distributed equally into three parts, diluted (1:3 v/v) with IMV Canadyl, HIA-1 or AU, and stored at 4°C for 6 h. Later, the semen was equilibrated at 20-25°C for 15 min, and divided in Eppendorf tubes. The sperm samples (final concentration of 50 × 106 sperm cells/mL per sample) were incubated at 37°C for 30 min in the absence (-) or presence (+) of 0.1 mM FeSO4 + 0.5 mM H2 O2 (Fenton system) and the following combinations of antioxidants: ascorbic acid + Trolox (A + T); ascorbic acid + Desferal (A + D); Trolox + Desferal (T + D) and ascorbic acid + Trolox + Desferal (A + T + D), all of them in a final concentration of 0.1 mM. Thus, the total number of samples was 30 and in each one, the sperm motility, velocity parameters, LPO and tGSH were determined. The motility and kinematic parameters of the diluted semen with added antioxidants were restored by up to 20% after inducing OS via the Fenton reaction. Dual combinations of antioxidants (A + T, A + D, and T + D) lowered LPO levels, but not equally across different extenders. After the induction of OS, the tGSH levels in diluted semen with IMV-Canadyl were not affected by the added antioxidants. Whereas antioxidant combinations in diluted semen with HIA-1 or AU had a beneficial effect and partially restored tGSH levels. In conclusion, the results showed that the extender IMV-Canadyl is well balanced and protected the Muscovy semen under OS conditions, while the other two extenders HIA-1 and AU can be improved by adding antioxidants.


Semen Preservation , Semen , Male , Animals , Antioxidants/pharmacology , Deferoxamine/pharmacology , Sperm Motility , Spermatozoa , Ascorbic Acid/pharmacology , Glutathione/pharmacology , Ducks , Semen Preservation/veterinary , Semen Preservation/methods , Cryopreservation/veterinary , Semen Analysis/veterinary , Cryoprotective Agents/pharmacology
...