Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.105
Filtrar
1.
J Neurosci Res ; 102(4): e25334, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38656648

RESUMO

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.


Assuntos
Astrócitos , Diferenciação Celular , Deficiências de Ferro , Oligodendroglia , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Oligodendroglia/metabolismo , Oligodendroglia/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Proteínas de Transporte de Cátions/metabolismo , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Ratos , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Células Precursoras de Oligodendrócitos/metabolismo , Desferroxamina/farmacologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Ferro/metabolismo
2.
J Cell Mol Med ; 28(8): e18306, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38613357

RESUMO

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.


Assuntos
Desferroxamina , Síndrome da Fibrose por Radiação , Camundongos , Animais , Camundongos Endogâmicos C57BL , Desferroxamina/farmacologia , Desferroxamina/uso terapêutico , Pele , Perfusão
3.
Environ Sci Technol ; 58(8): 3974-3984, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38306233

RESUMO

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.


Assuntos
Shewanella putrefaciens , Urânio , Biomineralização , Desferroxamina/metabolismo , Desferroxamina/farmacologia , Shewanella putrefaciens/metabolismo , Sideróforos/metabolismo , Sideróforos/farmacologia , Urânio/química , Compostos de Ferro/química
4.
Int Immunopharmacol ; 129: 111662, 2024 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-38340421

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 1 , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Ratos , Humanos , Animais , Desferroxamina/farmacologia , Desferroxamina/uso terapêutico , Diabetes Mellitus Tipo 1/terapia , Secretoma , Citocinas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Imunomodulação , Cordão Umbilical , Regeneração
5.
Brain Res Bull ; 207: 110878, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38218407

RESUMO

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.


Assuntos
Lesões Encefálicas Traumáticas , Ferroptose , Humanos , Camundongos , Animais , Desferroxamina/farmacologia , Células Endoteliais/metabolismo , Ferroptose/fisiologia , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/metabolismo , Ferro/metabolismo
6.
Int J Mol Sci ; 25(2)2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38255991

RESUMO

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.


Assuntos
Quitosana , Humanos , Animais , Camundongos , Desferroxamina/farmacologia , Quelantes , Transporte Biológico , Ferro
7.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167024, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38242180

RESUMO

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).


Assuntos
Desferroxamina , Ototoxicidade , Humanos , terc-Butil Hidroperóxido/toxicidade , terc-Butil Hidroperóxido/metabolismo , Desferroxamina/farmacologia , Ototoxicidade/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Células Ciliadas Auditivas/metabolismo , Ferro/metabolismo
8.
Transplant Proc ; 56(1): 223-227, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38199859

RESUMO

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.


Assuntos
Transplante de Fígado , Soluções para Preservação de Órgãos , Humanos , Ratos , Animais , Óxido de Deutério/farmacologia , Desferroxamina/farmacologia , Fígado , Soluções para Preservação de Órgãos/farmacologia , Reperfusão , Glutationa/farmacologia , Alopurinol/farmacologia , Insulina/farmacologia , Rafinose/farmacologia , Preservação de Órgãos , Adenosina
9.
Eur J Pharmacol ; 960: 176153, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-38059446

RESUMO

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.


Assuntos
Desferroxamina , Sobrecarga de Ferro , Camundongos , Masculino , Animais , Desferroxamina/farmacologia , Catalase , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Sobrecarga de Ferro/tratamento farmacológico , Sobrecarga de Ferro/patologia , Quelantes de Ferro/farmacologia , Quelantes de Ferro/uso terapêutico , Ferro , Superóxido Dismutase
10.
Shanghai Kou Qiang Yi Xue ; 32(4): 356-362, 2023 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-38044727

RESUMO

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.


Assuntos
Quitosana , Células-Tronco Mesenquimais , Ratos , Animais , Osteogênese/genética , Desferroxamina/farmacologia , Desferroxamina/metabolismo , Microesferas , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Alginatos/farmacologia , Células Cultivadas
11.
PLoS One ; 18(12): e0295257, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38100448

RESUMO

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.


Assuntos
Desferroxamina , Ferro , Ferro/metabolismo , Desferroxamina/farmacologia , Desferroxamina/metabolismo , Sideróforos/genética , Metais , Quelantes
12.
Sci Rep ; 13(1): 20145, 2023 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-37978208

RESUMO

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.


Assuntos
Ferroptose , Traumatismo por Reperfusão , Baixa Visão , Humanos , Animais , Ratos , Desferroxamina/farmacologia , Desferroxamina/uso terapêutico , Reperfusão , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/tratamento farmacológico , Ferritinas , Glutationa , Transferrinas , Espécies Reativas de Oxigênio
13.
Toxicol Appl Pharmacol ; 479: 116727, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37863361

RESUMO

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.


Assuntos
Cardiomiopatias , Ferroptose , Sobrecarga de Ferro , Ratos , Humanos , Masculino , Animais , Desferroxamina/metabolismo , Desferroxamina/farmacologia , Desferroxamina/uso terapêutico , Necroptose , Volume Sistólico , Ratos Wistar , Função Ventricular Esquerda , Apoptose , Sobrecarga de Ferro/tratamento farmacológico , Sobrecarga de Ferro/metabolismo , Ferro/metabolismo , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/prevenção & controle , Cardiomiopatias/induzido quimicamente , Mitocôndrias , Miócitos Cardíacos/metabolismo
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 43(9): 1577-1584, 2023 Sep 20.
Artigo em Chinês | MEDLINE | ID: mdl-37814872

RESUMO

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.


Assuntos
Medula Óssea , Desferroxamina , Camundongos , Animais , Caspase 3 , Desferroxamina/farmacologia , Raios X , Inibidores de Poli(ADP-Ribose) Polimerases , Fator A de Crescimento do Endotélio Vascular , Irradiação Corporal Total , Células da Medula Óssea , Peso Corporal
15.
Reprod Domest Anim ; 58(12): 1702-1711, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37818658

RESUMO

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.


Assuntos
Preservação do Sêmen , Sêmen , Masculino , Animais , Antioxidantes/farmacologia , Desferroxamina/farmacologia , Motilidade dos Espermatozoides , Espermatozoides , Ácido Ascórbico/farmacologia , Glutationa/farmacologia , Patos , Preservação do Sêmen/veterinária , Preservação do Sêmen/métodos , Criopreservação/veterinária , Análise do Sêmen/veterinária , Crioprotetores/farmacologia
16.
Biomolecules ; 13(8)2023 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-37627331

RESUMO

Diabetic nephropathy (DN) is one of the most devastating diabetic microvascular complications. It has previously been observed that iron metabolism levels are abnormal in diabetic patients. However, the mechanism by which iron metabolism levels affect DN is poorly understood. This study was designed to evaluate the role of iron-chelator deferoxamine (DFO) in the improvement of DN. Here, we established a DN rat model induced by diets high in carbohydrates and fat and streptozotocin (STZ) injection. Our data demonstrated that DFO treatment for three weeks greatly attenuated renal dysfunction as evidenced by decreased levels of urinary albumin, blood urea nitrogen, and serum creatinine, which were elevated in DN rats. Histopathological observations showed that DFO treatment improved the renal structures of DN rats and preserved podocyte integrity by preventing the decrease of transcripts of nephrin and podocin. In addition, DFO treatment reduced the overexpression of fibronectin 1, collagen I, IL-1ß, NF-κB, and MCP-1 in DN rats, as well as inflammatory cell infiltrates and collagenous fibrosis. Taken together, our findings unveiled that iron chelation via DFO injection had a protective impact on DN by alleviating inflammation and fibrosis, and that it could be a potential therapeutic strategy for DN.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Animais , Ratos , Nefropatias Diabéticas/tratamento farmacológico , Desferroxamina/farmacologia , Inflamação/tratamento farmacológico , Fibrose , Quelantes de Ferro/farmacologia , Quelantes de Ferro/uso terapêutico , Ferro
17.
Int J Biol Macromol ; 253(Pt 1): 126602, 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-37652316

RESUMO

Electrospinning is an advanced method used for developing wound dressings. Biopolymer-based electrospun mats have been extensively studied in tissue engineering due to their similarity to the extracellular matrix. In this study, electrospun poly(vinyl alcohol)/chitosan/silk fibroin (PChS) mat demonstrated improved mechanical properties, including tensile strength, strain at break, and Young's modulus, compared to electrospun poly(vinyl alcohol) and poly(vinyl alcohol)/chitosan mats. Similarly, the swelling capability, thermal stability, and hydrophilicity were higher in the PChS mat compared to the other ones. Hence, the PChS mat was selected for further investigation. Ciprofloxacin (CIP) was added to the PChS electrospinning solution at 5 % and 10 % concentration, and deferoxamine (DFO) was immobilized on CIP-loaded mats at 1 and 2 g/L concentration using a polydopamine linker. Evaluating mats with the dimensions of 1 × 1 cm2 showed that those containing 5 % and 10 % CIP exhibited bactericidal activity against Escherichia coli and Staphylococcus aureus. Moreover, Human dermal fibroblast cells were compatible with the fabricated mats, as confirmed by the MTT assay. Finally, drug-loaded mats had a positive effect on wound healing in a scratch test, and mats with 10 % CIP and 2 g/L DFO showed the highest effect on promoting wound healing, indicating potential for use as a wound dressing.


Assuntos
Quitosana , Nanofibras , Humanos , Ciprofloxacina/farmacologia , Quitosana/farmacologia , Antibacterianos/farmacologia , Desferroxamina/farmacologia , Seda/farmacologia , Álcool de Polivinil/farmacologia , Cicatrização
18.
Elife ; 122023 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-37267120

RESUMO

The reasons for poor healing of pressure injuries are poorly understood. Vascular ulcers are worsened by extracellular release of hemoglobin, so we examined the impact of myoglobin (Mb) iron in murine muscle pressure injuries (mPI). Tests used Mb-knockout or treatment with deferoxamine iron chelator (DFO). Unlike acute injuries from cardiotoxin, mPI regenerated poorly with a lack of viable immune cells, persistence of dead tissue (necro-slough), and abnormal deposition of iron. However, Mb-knockout or DFO-treated mPI displayed a reversal of the pathology: decreased tissue death, decreased iron deposition, decrease in markers of oxidative damage, and higher numbers of intact immune cells. Subsequently, DFO treatment improved myofiber regeneration and morphology. We conclude that myoglobin iron contributes to tissue death in mPI. Remarkably, a large fraction of muscle death in untreated mPI occurred later than, and was preventable by, DFO treatment, even though treatment started 12 hr after pressure was removed. This demonstrates an opportunity for post-pressure prevention to salvage tissue viability.


Assuntos
Ferro , Lesão por Pressão , Camundongos , Humanos , Animais , Desferroxamina/farmacologia , Mioglobina , Necrose , Músculos
19.
Curr Drug Targets ; 24(8): 688-696, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37278033

RESUMO

INTRODUCTION: Several studies demonstrated that deferoxamine, an iron chelator, can improve inflammatory alterations in adipose tissue induced by obesity. Obesity alterations in adipose tissue are also associated with tissue remodeling, and deferoxamine has anti-fibrosis action previously described in sites like the skin and liver. METHODS: In this work, we analyzed deferoxamine effects on adipose tissue fibro-inflammation during obesity induced by diet in mice. in vitro approaches with fibroblasts and macrophages were also carried out to elucidate deferoxamine activity. RESULTS: Our results demonstrated that in addition to exerting anti-inflammatory effects, reducing the cytokine production in adipose tissue of obese mice and by human monocyte differentiated in macrophage in vitro, deferoxamine can alter metalloproteinases expression and extracellular matrix production in vivo and in vitro. CONCLUSION: Deferoxamine could be an alternative to control fibro-inflammation in obese adipose tissue, contributing to the metabolic improvements previously described.


Assuntos
Desferroxamina , Resistência à Insulina , Humanos , Animais , Camundongos , Desferroxamina/farmacologia , Desferroxamina/uso terapêutico , Desferroxamina/metabolismo , Tecido Adiposo , Obesidade/metabolismo , Inflamação/metabolismo , Fígado/metabolismo , Camundongos Endogâmicos C57BL
20.
Neuroscience ; 524: 149-157, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37286159

RESUMO

Deferoxamine (DFO) is a potent iron chelator for clinical treatment of various diseases. Recent studies have also shown its potential to promote vascular regeneration during peripheral nerve regeneration. However, the effect of DFO on the Schwann cell function and axon regeneration remains unclear. In this study, we investigated the effects of different concentrations of DFO on Schwann cell viability, proliferation, migration, expression of key functional genes, and axon regeneration of dorsal root ganglia (DRG) through a series of in vitro experiments. We found that DFO improves Schwann cell viability, proliferation, and migration in the early stages, with an optimal concentration of 25 µM. DFO also upregulates the expression of myelin-related genes and nerve growth-promoting factors in Schwann cells, while inhibiting the expression of Schwann cell dedifferentiation genes. Moreover, the appropriate concentration of DFO promotes axon regeneration in DRG. Our findings demonstrate that DFO, with suitable concentration and duration of action, can positively affect multiple stages of peripheral nerve regeneration, thereby improving the effectiveness of nerve injury repair. This study also enriches the theory of DFO promoting peripheral nerve regeneration and provides a basis for the design of sustained-release DFO nerve grafts.


Assuntos
Regeneração Nervosa , Traumatismos dos Nervos Periféricos , Humanos , Regeneração Nervosa/fisiologia , Gânglios Espinais , Axônios , Desferroxamina/metabolismo , Desferroxamina/farmacologia , Células Cultivadas , Células de Schwann/metabolismo , Fatores de Crescimento Neural/metabolismo , Traumatismos dos Nervos Periféricos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...