Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.673
Filtrar
1.
Yakugaku Zasshi ; 144(6): 627-631, 2024.
Artigo em Japonês | MEDLINE | ID: mdl-38825471

RESUMO

Cefiderocol is a novel siderophore-conjugated cephalosporin with a catechol residue acting as an iron chelator. Cefiderocol forms a chelating complex with ferric iron and is transported rapidly into bacterial cells through iron-uptake systems. As a result, cefiderocol shows good activity against Gram-negative bacteria, including carbapenem-resistant isolates that are causing significant global health issues. Cefiderocol has been approved for clinical use in the United States and Europe, where it is being used to treat infection caused by carbapenem-resistant Gram-negative pathogens.


Assuntos
Antibacterianos , Cefiderocol , Cefalosporinas , Bactérias Gram-Negativas , Sideróforos , Cefalosporinas/farmacologia , Cefalosporinas/química , Sideróforos/química , Humanos , Antibacterianos/farmacologia , Antibacterianos/química , Bactérias Gram-Negativas/efeitos dos fármacos , Quelantes de Ferro/farmacologia , Ferro/metabolismo , Farmacorresistência Bacteriana , Descoberta de Drogas , Carbapenêmicos/farmacologia , Infecções por Bactérias Gram-Negativas/tratamento farmacológico
2.
J Oleo Sci ; 73(7): 991-999, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38945927

RESUMO

In this study, we evaluated the cancer cell killing activity of koji mold-derived extracts using several solvents. The koji mold lipid extract (KML) exhibited potent cytotoxicity against a human leukemia cell line. Fractionation of the KML via silica gel chromatography revealed the presence of active components in fraction (Fr.) 6. Cytotoxic effects of Fr. 6 were inhibited by the ferroptosis inhibitors, ferrostatin-1 and SRS11-92, and the iron chelator, deferoxamine. Interestingly, ferroptosis inhibitors failed to prevent the KML-induced cell death. Fr. 6 decreased the expression of glutathione peroxidase 4 (GPx4) and increased the level of peroxidized plasma membrane lipids. Furthermore, Fr. 6 decreased the intracellular glutathione levels. Overall, our results suggest that Fr. 6 included in KML induces ferroptosis in HL-60 cells.


Assuntos
Ferroptose , Glutationa , Peroxidação de Lipídeos , Oxirredução , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Humanos , Células HL-60 , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Ferroptose/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Glutationa/metabolismo , Oxirredução/efeitos dos fármacos , Desferroxamina/farmacologia , Cicloexilaminas/farmacologia , Lipídeos , Fenilenodiaminas/farmacologia , Lipídeos de Membrana/metabolismo , Quelantes de Ferro/farmacologia
3.
Int J Biochem Cell Biol ; 173: 106610, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38879152

RESUMO

Colorectal cancer (CRC) is the third most common type of cancer in the world. It is characterized by complex crosstalk between various signaling pathways, as a result of which it is highly challenging to identify optimal therapeutic targets and design treatment strategies. In this study, we tested the effect of 700 compounds on the CRC cell line HT-29 by using the sulforhodamine B assay and screened out 17 compounds that exhibited high toxicity (indicated by an inhibition rate of ≥75 % when applied at a concentration of 10 µM) against the HT-29 cell line. Next, we investigated the mechanisms underlying the effects of these 17 highly toxic compounds. The results of ferroptosis analysis and electron microscopy showed that compounds 575 and 578 were able to significantly reverse RSL3-induced increase in ferroptosis, while compound 580 had a less pronounced ferroptosis-regulating effect. In subsequent experiments, western blotting showed that compounds 575, 578, and 580, which belong to a class of meroterpene-like compounds that affect ferroptosis, do not induce autophagy or apoptosis in the CRC cell line. Instead, Fe2+ chelation experiments showed that these three compounds can serve as iron chelators by chelating Fe2+ at a 1:1 (chelator: Fe2+) ratio. Specifically, the aldehyde and hydroxyl groups of the benzene ring in these compounds may chelate Fe2+, thus reducing Fe2+ levels in cells and inhibiting ferroptosis. These results indicate that these novel meroterpene-like compounds are potential therapeutic small-molecule candidates for targeting ferroptosis in tumors.


Assuntos
Ferroptose , Quelantes de Ferro , Ferro , Ferroptose/efeitos dos fármacos , Humanos , Quelantes de Ferro/farmacologia , Quelantes de Ferro/química , Células HT29 , Ferro/metabolismo , Terpenos/farmacologia , Terpenos/química , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Apoptose/efeitos dos fármacos
4.
Cardiovasc Diabetol ; 23(1): 186, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38812011

RESUMO

BACKGROUND: Vascular calcification (VC) is an independent risk factor for cardiovascular diseases. Recently, ferroptosis has been recognised as a novel therapeutic target for cardiovascular diseases. Although an association between ferroptosis and vascular calcification has been reported, the role and mechanism of iron overload in vascular calcification are still poorly understood. Specifically, further in-depth research is required on whether metalloproteins SLC39a14 and SLC39a8 are involved in ferroptosis induced by iron overload. METHODS: R language was employed for the differential analysis of the dataset, revealing the correlation between ferroptosis and calcification. The experimental approaches encompassed both in vitro and in vivo studies, incorporating the use of iron chelators and models of iron overload. Additionally, gain- and loss-of-function experiments were conducted to investigate iron's effects on vascular calcification comprehensively. Electron microscopy, immunofluorescence, western blotting, and real-time polymerase chain reaction were used to elucidate how Slc39a14 and Slc39a8 mediate iron overload and promote calcification. RESULTS: Ferroptosis was observed in conjunction with vascular calcification (VC); the association was consistently confirmed by in vitro and in vivo studies. Our results showed a positive correlation between iron overload in VSMCs and calcification. Iron chelators are effective in reversing VC and iron overload exacerbates this process. The expression levels of the metal transport proteins Slc39a14 and Slc39a8 were significantly upregulated during calcification; the inhibition of their expression alleviated VC. Conversely, Slc39a14 overexpression exacerbates calcification and promotes intracellular iron accumulation in VSMCs. CONCLUSIONS: Our research demonstrates that iron overload occurs during VC, and that inhibition of Slc39a14 and Slc39a8 significantly relieves VC by intercepting iron overload-induced ferroptosis in VSMCs, providing new insights into the VC treatment.


Assuntos
Proteínas de Transporte de Cátions , Modelos Animais de Doenças , Ferroptose , Quelantes de Ferro , Camundongos Endogâmicos C57BL , Músculo Liso Vascular , Miócitos de Músculo Liso , Calcificação Vascular , Ferroptose/efeitos dos fármacos , Calcificação Vascular/metabolismo , Calcificação Vascular/patologia , Animais , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Transporte de Cátions/genética , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Músculo Liso Vascular/patologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Quelantes de Ferro/farmacologia , Quelantes de Ferro/uso terapêutico , Transdução de Sinais , Masculino , Humanos , Ferro/metabolismo , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/patologia
5.
J Med Chem ; 67(11): 8630-8641, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38747630

RESUMO

A novel Fe(III) complex, Fe-tBPCDTA, was synthesized and explored as a potential contrast agent for MRI. Compared to established agents like Fe-EDTA and Fe-tCDTA, Fe-tBPCDTA exhibited moderate relaxivity (r1 = 1.17 s-1·mmol-1) due to its enhanced second-sphere mechanism. It also displayed improved kinetic inertness, lower cytotoxicity, and enhanced redox stability. In vivo studies demonstrated its function as an extracellular fluid agent, providing tumor contrast comparable to that of Gd-DTPA at a higher dosage. Complete renal clearance occurred within 24 h. These findings suggest Fe-tBPCDTA as a promising candidate for further development as a safe and effective extracellular MRI contrast agent.


Assuntos
Meios de Contraste , Quelantes de Ferro , Imageamento por Ressonância Magnética , Organofosfonatos , Meios de Contraste/química , Meios de Contraste/síntese química , Imageamento por Ressonância Magnética/métodos , Animais , Humanos , Organofosfonatos/química , Organofosfonatos/síntese química , Quelantes de Ferro/química , Quelantes de Ferro/farmacologia , Quelantes de Ferro/síntese química , Compostos Férricos/química , Camundongos , Linhagem Celular Tumoral , Quelantes/química , Quelantes/síntese química
6.
Int Immunopharmacol ; 135: 112318, 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38795598

RESUMO

Ferroptosis of intestinal epithelial cells (IECs) had been identified as a key factor in the development of ulcerative colitis (UC). Therefore, targeted inhibition of ferroptosis may provide a new strategy for the treatment of UC. Isorhamnetin (ISO) was an O-methylated flavonol with therapeutic effects on a variety of diseases, such as cardiovascular disease, neurological disorders and tumors. However, the role and mechanism of ISO in ferroptosis and associated colitis were rarely investigated. In this study, we demonstrated that ISO could effectively alleviate intestinal inflammation by inhibiting ferroptosis of IECs in DSS-induced mice. Moreover, our results shown that ISO acted as a potent and common ferroptosis inhibitor in multiple human and murine cell lines. Mechanistically, ISO inhibited ferroptosis independent of its previously reported targets MEK1 and PI3K, but alleviated oxidative stress by targeting and activating NRF2. Furthermore, ISO could also directly chelate iron to hinder ferroptosis. In conclusion, our study indicated that ISO as a novel potential ferroptosis inhibitor, providing a promising therapeutic strategy for ferroptosis-related colitis.


Assuntos
Ferroptose , Heme Oxigenase-1 , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2 , Quercetina , Transdução de Sinais , Animais , Ferroptose/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Quercetina/farmacologia , Quercetina/análogos & derivados , Quercetina/uso terapêutico , Humanos , Camundongos , Heme Oxigenase-1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Colite/tratamento farmacológico , Colite/induzido quimicamente , Colite/metabolismo , Sulfato de Dextrana , Ferro/metabolismo , Quelantes de Ferro/farmacologia , Quelantes de Ferro/uso terapêutico , Linhagem Celular , Masculino , Estresse Oxidativo/efeitos dos fármacos , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/patologia , Colite Ulcerativa/induzido quimicamente
7.
Phytomedicine ; 129: 155700, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38704914

RESUMO

BACKGROUND: Myoglobin (Mb) induced death of renal tubular epithelial cells (RTECs) is a major pathological factor in crush syndrome-related acute kidney injury (CS-AKI). It is unclear whether ferroptosis is involved and could be a target for treatment. PURPOSE: This study aimed to evaluate the potential therapeutic effects of combining the natural small molecule rosemarinic acid (RA) and the iron chelator deferasirox (Dfe) on CS-AKI through inhibition of ferroptosis. METHODS: Sequencing data were downloaded from the GEO database, and differential expression analysis was performed using the R software limma package. The CS-AKI mouse model was constructed by squeezing the bilateral thighs of mice for 16 h with 1.5 kg weight. TCMK1 and NRK-52E cells were induced with 200 µM Mb and then treated with RA combined with Dfe (Dfe + RA, both were 10 µM). Functional and pathological changes in mouse kidney were evaluated by glomerular filtration rate (GFR) and HE pathology. Immunofluorescence assay was used to detect Mb levels in kidney tissues. The expression levels of ACSL4, GPX4, Keap1, and Nrf2 were analyzed by WB. RESULTS: We found that AKI mice in the GSE44925 cohort highly expressed the ferroptosis markers ACSL4 and PTGS2. CS-AKI mice showed a rapid decrease in GFR, up-regulation of ACSL4 expression in kidney tissue, and down-regulation of GPX4 expression, indicating activation of the ferroptosis pathway. Mb was found to deposit in renal tubules, and it has been proven to cause ferroptosis in TCMK1 and NRK-52E cells in vitro. We found that Dfe had a strong iron ion scavenging effect and inhibited ACSL4 expression. RA could disrupt the interaction between Keap1 andNrf2, stabilize Nrf2, and promote its nuclear translocation, thereby exerting antioxidant effects. The combination of Dfe and RA effectively reversed Mb induced ferroptosis in RTECs. CONCLUSION: In conclusion, we found that RA combined with Dfe attenuated CS-AKI by inhibiting Mb-induced ferroptosis in RTECs via activating the Nrf2/Keap1 pathway.


Assuntos
Injúria Renal Aguda , Cinamatos , Deferasirox , Depsídeos , Ferroptose , Proteína 1 Associada a ECH Semelhante a Kelch , Fator 2 Relacionado a NF-E2 , Ácido Rosmarínico , Animais , Ferroptose/efeitos dos fármacos , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Injúria Renal Aguda/tratamento farmacológico , Depsídeos/farmacologia , Camundongos , Deferasirox/farmacologia , Masculino , Cinamatos/farmacologia , Modelos Animais de Doenças , Quelantes de Ferro/farmacologia , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular , Camundongos Endogâmicos C57BL
8.
G3 (Bethesda) ; 14(7)2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38805688

RESUMO

Nature has been a rich source of pharmaceutical compounds, producing 80% of our currently prescribed drugs. The feijoa plant, Acca sellowiana, is classified in the family Myrtaceae, native to South America, and currently grown worldwide to produce feijoa fruit. Feijoa is a rich source of bioactive compounds with anticancer, anti-inflammatory, antibacterial, and antifungal activities; however, the mechanism of action of these compounds is largely not known. Here, we used chemical genetic analyses in the model organism Saccharomyces cerevisiae to investigate the mechanism of action of a feijoa-derived ethanol adduct of vescalagin (EtOH-vescalagin). Genome-wide barcode sequencing analysis revealed yeast strains lacking genes in iron metabolism, zinc metabolism, retromer function, or mitochondrial function were hypersensitive to 0.3 µM EtOH-vescalagin. This treatment increased expression of iron uptake proteins at the plasma membrane, which was a compensatory response to reduced intracellular iron. Likewise, EtOH-vescalagin increased expression of the Cot1 protein in the vacuolar membrane that transports zinc into the vacuole to prevent cytoplasmic accumulation of zinc. Each individual subunit in the retromer complex was required for the iron homeostatic mechanism of EtOH-vescalagin, while only the cargo recognition component in the retromer complex was required for the zinc homeostatic mechanism. Overexpression of either retromer subunits or high-affinity iron transporters suppressed EtOH-vescalagin bioactivity in a zinc-replete condition, while overexpression of only retromer subunits increased EtOH-vescalagin bioactivity in a zinc-deficient condition. Together, these results indicate that EtOH-vescalagin bioactivity begins with extracellular iron chelation and proceeds with intracellular transport of zinc via the retromer complex. More broadly, this is the first report of a bioactive compound to further characterize the poorly understood interaction between zinc metabolism and retromer function.


Assuntos
Etanol , Frutas , Homeostase , Taninos Hidrolisáveis , Ferro , Saccharomyces cerevisiae , Zinco , Zinco/metabolismo , Taninos Hidrolisáveis/farmacologia , Taninos Hidrolisáveis/metabolismo , Ferro/metabolismo , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/efeitos dos fármacos , Etanol/metabolismo , Frutas/metabolismo , Quelantes de Ferro/farmacologia , Genômica/métodos
9.
Int J Mol Sci ; 25(9)2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38731873

RESUMO

The supply and control of iron is essential for all cells and vital for many physiological processes. All functions and activities of iron are expressed in conjunction with iron-binding molecules. For example, natural chelators such as transferrin and chelator-iron complexes such as haem play major roles in iron metabolism and human physiology. Similarly, the mainstay treatments of the most common diseases of iron metabolism, namely iron deficiency anaemia and iron overload, involve many iron-chelator complexes and the iron-chelating drugs deferiprone (L1), deferoxamine (DF) and deferasirox. Endogenous chelators such as citric acid and glutathione and exogenous chelators such as ascorbic acid also play important roles in iron metabolism and iron homeostasis. Recent advances in the treatment of iron deficiency anaemia with effective iron complexes such as the ferric iron tri-maltol complex (feraccru or accrufer) and the effective treatment of transfusional iron overload using L1 and L1/DF combinations have decreased associated mortality and morbidity and also improved the quality of life of millions of patients. Many other chelating drugs such as ciclopirox, dexrazoxane and EDTA are used daily by millions of patients in other diseases. Similarly, many other drugs or their metabolites with iron-chelation capacity such as hydroxyurea, tetracyclines, anthracyclines and aspirin, as well as dietary molecules such as gallic acid, caffeic acid, quercetin, ellagic acid, maltol and many other phytochelators, are known to interact with iron and affect iron metabolism and related diseases. Different interactions are also observed in the presence of essential, xenobiotic, diagnostic and theranostic metal ions competing with iron. Clinical trials using L1 in Parkinson's, Alzheimer's and other neurodegenerative diseases, as well as HIV and other infections, cancer, diabetic nephropathy and anaemia of inflammation, highlight the importance of chelation therapy in many other clinical conditions. The proposed use of iron chelators for modulating ferroptosis signifies a new era in the design of new therapeutic chelation strategies in many other diseases. The introduction of artificial intelligence guidance for optimal chelation therapeutic outcomes in personalised medicine is expected to increase further the impact of chelation in medicine, as well as the survival and quality of life of millions of patients with iron metabolic disorders and also other diseases.


Assuntos
Quelantes de Ferro , Sobrecarga de Ferro , Humanos , Sobrecarga de Ferro/tratamento farmacológico , Sobrecarga de Ferro/metabolismo , Quelantes de Ferro/uso terapêutico , Quelantes de Ferro/farmacologia , Anemia Ferropriva/tratamento farmacológico , Anemia Ferropriva/metabolismo , Ferro/metabolismo , Animais , Deferiprona/uso terapêutico , Deferiprona/farmacologia
10.
Free Radic Biol Med ; 221: 273-282, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-38740102

RESUMO

Defective mitochondria and autophagy, as well as accumulation of lipid and iron in WDR45 mutant fibroblasts, is related to beta-propeller protein-associated neurodegeneration (BPAN). In this study, we found that enlarged lysosomes in cells derived from patients with BPAN had low enzyme activity, and most of the enlarged lysosomes had an accumulation of iron and oxidized lipid. Cryo-electron tomography revealed elongated lipid accumulation, and spectrometry-based elemental analysis showed that lysosomal iron and oxygen accumulation superimposed with lipid aggregates. Lysosomal lipid aggregates superimposed with autofluorescence as free radical generator, lipofuscin. To eliminate free radical stress by iron accumulation in cells derived from patients with BPAN, we investigated the effects of the iron chelator, 2,2'-bipyridine (bipyridyl, BIP). To study whether the defects in patient-derived cells can be rescued by an iron chelator BIP, we tested whether the level of iron and reactive oxygen species (ROS) in the cells and genes related to oxidative stress were rescued BIP treatment. Although BIP treatment decreased some iron accumulation in the cytoplasm and mitochondria, the accumulation of iron in the lysosomes and levels of cellular ROS were unaffected. In addition, the change of specific RNA levels related to free radical stress in patient fibroblasts was not rescued by BIP. To alleviate free radical stress, we investigated whether l-serine can regulate abnormal structures in cells derived from patients with BPAN through the regulation of free radical stress. l-serine treatment alleviated increase of enlarged lysosomes and iron accumulation and rescued impaired lysosomal activity by reducing oxidized lipid accumulation in the lysosomes of the cells. Lamellated lipids in the lysosomes of the cells were identified as lipofuscin through correlative light and electron microscopy, and l-serine treatment reduced the increase of lipofuscin. These data suggest that l-serine reduces oxidative stress-mediated lysosomal lipid oxidation and iron accumulation by rescuing lysosomal activity.


Assuntos
Fibroblastos , Ferro , Lipofuscina , Lisossomos , Estresse Oxidativo , Espécies Reativas de Oxigênio , Serina , Humanos , Lisossomos/metabolismo , Lisossomos/efeitos dos fármacos , Lipofuscina/metabolismo , Ferro/metabolismo , Fibroblastos/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Serina/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Distrofias Neuroaxonais/metabolismo , Distrofias Neuroaxonais/patologia , Distrofias Neuroaxonais/tratamento farmacológico , Distrofias Neuroaxonais/genética , 2,2'-Dipiridil/farmacologia , 2,2'-Dipiridil/análogos & derivados , Quelantes de Ferro/farmacologia
11.
Adv Sci (Weinh) ; 11(25): e2305895, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38671590

RESUMO

Myocardial infarction (MI), a consequence of coronary artery occlusion, triggers the degradation of ferritin, resulting in elevated levels of free iron in the heart and thereby inducing ferroptosis. Targeting myocardial ferroptosis through the chelation of excess iron has therapeutic potential for MI treatment. However, iron chelation in post ischemic injury areas using conventional iron-specific chelators is hindered by ineffective myocardial intracellular chelation, rapid clearance, and high systemic toxicity. A chitosan-desferrioxamine nanosponge (CDNS) is designed by co-crosslinking chitosan and deferoxamine through noncovalent gelation to address these challenges. This architecture facilitates direct iron chelation regardless of deferoxamine (DFO) release due to its sponge-like porous hydrogel structure. Upon cellular internalization, CDNS can effectively chelate cellular iron and facilitate the efflux of captured iron, thereby inhibiting ferroptosis and associated oxidative stress and lipid peroxidation. In MI mouse models, myocardial injection of CDNS promotes sustainable retention and the suppression of ferroptosis in the infarcted heart. This intervention improves cardiac function and alleviates adverse cardiac remodeling post-MI, leading to decreased oxidative stress and the promotion of angiogenesis due to ferroptosis inhibition by CDNS in the infarcted heart. This study reveals a nanosponge-based nanomedicine targeting myocardial ferroptosis with efficient iron chelation and efflux, offering a promising MI treatment.


Assuntos
Modelos Animais de Doenças , Ferroptose , Quelantes de Ferro , Infarto do Miocárdio , Ferroptose/efeitos dos fármacos , Animais , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Camundongos , Quelantes de Ferro/farmacologia , Quelantes de Ferro/uso terapêutico , Desferroxamina/farmacologia , Quitosana , Ferro/metabolismo , Nanopartículas , Masculino , Estresse Oxidativo/efeitos dos fármacos , Camundongos Endogâmicos C57BL
12.
Eur J Med Res ; 29(1): 253, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38659000

RESUMO

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


Assuntos
Produtos Biológicos , Insuficiência Cardíaca , Ferro , Humanos , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/tratamento farmacológico , Ferro/metabolismo , Produtos Biológicos/uso terapêutico , Produtos Biológicos/farmacologia , Animais , Ferroptose/efeitos dos fármacos , Quelantes de Ferro/uso terapêutico , Quelantes de Ferro/farmacologia , Antioxidantes/uso terapêutico
13.
Int J Mol Sci ; 25(8)2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38673849

RESUMO

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


Assuntos
Anemia Falciforme , Farmacogenética , Talassemia beta , Humanos , Anemia Falciforme/genética , Anemia Falciforme/tratamento farmacológico , Talassemia beta/genética , Talassemia beta/tratamento farmacológico , Farmacogenética/métodos , Hemoglobina Fetal/genética , gama-Globinas/genética , Quelantes de Ferro/uso terapêutico , Quelantes de Ferro/farmacologia
14.
Clin Immunol ; 263: 110205, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38575044

RESUMO

Increasing clinical data show that the imbalance of host metallome is closely associated with different kinds of disease, however, the intrinsic mechanisms of action of metals in immunity and pathogenesis of disease remain largely undefined. There is lack of multiplexed profiling system to integrate the metalloproteome-immunoproteome information at systemic level for exploring the roles of metals in immunity and disease pathogenesis. In this study, we build up a metal-coding assisted multiplexed proteome assay platform for serum metalloproteomic and immunoproteomic profiling. By taking COVID-19 as a showcase, we unbiasedly uncovered the most evident modulation of iron-related proteins, i.e., Ft and Tf, in serum of severe COVID-19 patients, and the value of Ft/Tf could work as a robust biomarker for COVID-19 severity stratification, which overtakes the well-established clinical risk factors (cytokines). We further uncovered a tight association of transferrin with inflammation mediator IL-10 in COVID-19 patients, which was proved to be mainly governed by the monocyte/macrophage of liver, shedding light on new pathophysiological and immune regulatory mechanisms of COVID-19 disease. We finally validated the beneficial effects of iron chelators as anti-viral agents in SARS-CoV-2-infected K18-hACE2 mice through modulation of iron dyshomeostasis and alleviating inflammation response. Our findings highlight the critical role of liver-mediated iron dysregulation in COVID-19 disease severity, providing solid evidence on the involvement of iron-related proteins in COVID-19 pathophysiology and immunity.


Assuntos
COVID-19 , Ferro , Proteoma , SARS-CoV-2 , COVID-19/imunologia , Humanos , Animais , SARS-CoV-2/imunologia , Camundongos , Ferro/metabolismo , Proteômica/métodos , Transferrina/metabolismo , Metaloproteínas/imunologia , Metaloproteínas/metabolismo , Masculino , Feminino , Biomarcadores/sangue , Biomarcadores/metabolismo , Quelantes de Ferro/uso terapêutico , Quelantes de Ferro/farmacologia , Interleucina-10/imunologia , Interleucina-10/metabolismo , Pessoa de Meia-Idade
15.
J Pharmacol Sci ; 155(2): 44-51, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38677785

RESUMO

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


Assuntos
Hemina , Hemoglobinas , Ferro , Epitélio Pigmentado da Retina , Humanos , Morte Celular/efeitos dos fármacos , Linhagem Celular , Cicloexilaminas/farmacologia , Hemina/farmacologia , Hemoglobinas/metabolismo , Ferro/metabolismo , Quelantes de Ferro/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Fenilenodiaminas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/patologia
16.
Neurochem Int ; 176: 105725, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38561151

RESUMO

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


Assuntos
Anticonvulsivantes , Encéfalo , Deferasirox , Epilepsia , Quelantes de Ferro , Ferro , Proteínas de Membrana , Animais , Masculino , Camundongos , Anticonvulsivantes/farmacologia , Anticonvulsivantes/uso terapêutico , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Deferasirox/farmacologia , Epilepsia/tratamento farmacológico , Epilepsia/metabolismo , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Ferro/metabolismo , Quelantes de Ferro/farmacologia , Quelantes de Ferro/uso terapêutico , Excitação Neurológica/efeitos dos fármacos , Pentilenotetrazol/toxicidade , Ratos Sprague-Dawley , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo
17.
J Vis Exp ; (204)2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38465932

RESUMO

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


Assuntos
Quelantes de Ferro , Ferro , Quelantes de Ferro/farmacologia , Quelantes de Ferro/metabolismo , Ferro/metabolismo , Fluoresceínas/metabolismo , Corantes Fluorescentes
18.
Int J Biol Macromol ; 263(Pt 2): 130387, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38401586

RESUMO

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


Assuntos
Doença de Alzheimer , Ferroptose , Doenças Neurodegenerativas , Ratos , Masculino , Animais , Doença de Alzheimer/tratamento farmacológico , Espermidina/farmacologia , Espermidina/metabolismo , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Peptídeos beta-Amiloides/metabolismo , Estresse Oxidativo , Ciprofloxacina/farmacologia , Quelantes de Ferro/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo
19.
Artigo em Inglês | MEDLINE | ID: mdl-38423705

RESUMO

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


Assuntos
Benzoatos , Sobrecarga de Ferro , Humanos , Deferasirox/farmacologia , Deferasirox/uso terapêutico , Benzoatos/farmacologia , Benzoatos/uso terapêutico , Benzoatos/metabolismo , Triazóis/uso terapêutico , Triazóis/farmacocinética , Quelantes de Ferro/farmacologia , Quelantes de Ferro/uso terapêutico , Quelantes de Ferro/metabolismo , Sobrecarga de Ferro/tratamento farmacológico , Ferro/metabolismo , Ferro/uso terapêutico
20.
Microbiol Spectr ; 12(2): e0259423, 2024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38230926

RESUMO

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


Assuntos
Anti-Infecciosos , Micoses , Animais , Humanos , Antifúngicos/farmacologia , Fluconazol/farmacologia , Ferro , Candida , Micoses/microbiologia , Candida albicans , Anti-Infecciosos/farmacologia , Azóis/farmacologia , Candida glabrata , Quelantes de Ferro/farmacologia , Farmacorresistência Fúngica , Testes de Sensibilidade Microbiana , Mamíferos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...