Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 223: 116132, 2024 May.
Article in English | MEDLINE | ID: mdl-38492782

ABSTRACT

Cisplatin is an effective chemotherapeutic drug for different cancers, but it also causes severe and permanent hearing loss. Oxidative stress and mitochondrial dysfunction in cochlear hair cells (HCs) have been shown to be important in the pathogenesis of cisplatin-induced hearing loss (CIHL). CDGSH iron sulfur domain 1 (CISD1, also known as mitoNEET) plays a critical role in mitochondrial oxidative capacity and cellular bioenergetics. Targeting CISD1 may improve mitochondrial function in various diseases. However, the role of CISD1 in cisplatin-induced ototoxicity is unclear. Therefore, this study was performed to assess the role of CISD1 in cisplatin-induced ototoxicity. We found that CISD1 expression was significantly increased after cisplatin treatment in both HEI-OC1 cells and cochlear HCs. Moreover, pharmacological inhibition of CISD1 with NL-1 inhibited cell apoptosis and reduced mitochondrial reactive oxygen species accumulation in HEI-OC1 cells and cochlear explants. Inhibition of CISD1 with small interfering RNA in HEI-OC1 cells had similar protective effects. Furthermore, NL-1 protected against CIHL in adult C57 mice, as evaluated by the auditory brainstem response and immunofluorescent staining. Mechanistically, RNA sequencing revealed that NL-1 attenuated CIHL via the PI3K and MAPK pathways. Most importantly, NL-1 did not interfere with the antitumor efficacy of cisplatin. In conclusion, our study revealed that targeting CISD1 with NL-1 reduced reactive oxygen species accumulation, mitochondrial dysfunction, and apoptosis via the PI3K and MAPK pathways in HEI-OC1 cell lines and mouse cochlear explants in vitro, and it protected against CIHL in adult C57 mice. Our study suggests that CISD1 may serve as a novel target for the prevention of CIHL.


Subject(s)
Antineoplastic Agents , Hearing Loss , Mitochondrial Diseases , Ototoxicity , Mice , Animals , Cisplatin/toxicity , Cisplatin/metabolism , Antineoplastic Agents/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Reactive Oxygen Species/metabolism , Ototoxicity/prevention & control , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Apoptosis , Membrane Proteins/metabolism , Iron-Binding Proteins/pharmacology
2.
Toxicol Appl Pharmacol ; 465: 116440, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36870574

ABSTRACT

Ischemia/reperfusion- (I/R-) induced injury is unavoidable and a major risk factor for graft failure and acute rejection following kidney transplantation. However, few effective interventions are available to improve the outcome due to the complicated mechanisms and lack of appropriate therapeutic targets. Hence, this research aimed to explore the effect of the thiazolidinedione (TZD) compounds on I/R-induced kidney damage. One of the main causes of renal I/R injury is the ferroptosis of renal tubular cells. In this study, compared with the antidiabetic TZD pioglitazone (PGZ), we found its derivative mitoglitazone (MGZ) exerted significantly inhibitory effects on erastin-induced ferroptosis by suppressing mitochondrial membrane potential hyperpolarization and lipid ROS production in HEK293 cells. Moreover, MGZ pretreatment remarkably alleviated I/R-induced renal damages by inhibiting cell death and inflammation, upregulating the expression of glutathione peroxidase 4 (GPX4), and reducing iron-related lipid peroxidation in C57BL/6 N mice. Additionally, MGZ exhibited excellent protection against I/R-induced mitochondrial dysfunction by restoring ATP production, mitochondrial DNA copy numbers, and mitochondrial morphology in kidney tissues. Mechanistically, molecular docking and surface plasmon resonance experiments demonstrated that MGZ exhibited a high binding affinity with the mitochondrial outer membrane protein mitoNEET. Collectively, our findings indicated the renal protective effect of MGZ was closely linked to regulating the mitoNEET-mediated ferroptosis pathway, thus offering potential therapeutic strategies for ameliorating I/R injuries.


Subject(s)
Ferroptosis , Reperfusion Injury , Mice , Animals , Humans , HEK293 Cells , Molecular Docking Simulation , Mice, Inbred C57BL , Kidney/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Membrane Proteins/metabolism , Iron-Binding Proteins/metabolism , Iron-Binding Proteins/pharmacology
3.
Haematologica ; 104(4): 669-677, 2019 04.
Article in English | MEDLINE | ID: mdl-30442724

ABSTRACT

Myelosuppression is a major and frequently dose-limiting side effect of anticancer therapy and is responsible for most treatment-related morbidity and mortality. In addition, repeated cycles of DNA damage and cell death of hematopoietic stem and progenitor cells, followed by compensatory proliferation and selection pressure, lead to genomic instability and pave the way for therapy-related myelodysplastic syndromes and secondary acute myeloid leukemia. Protection of hematopoietic stem and progenitor cells from chemo- and radiotherapy in patients with solid tumors would reduce both immediate complications and long-term sequelae. Epidermal growth factor (EGF) and prostaglandin E2 (PGE2) were reported to prevent chemo- or radiotherapy-induced myelosuppression in mice. We tested both molecules for potentially protective effects on human CD34+ cells in vitro and established a xenograft mouse model to analyze stress resistance and regeneration of human hematopoiesis in vivo EGF was neither able to protect human stem and progenitor cells in vitro nor to promote hematopoietic regeneration following sublethal irradiation in vivo PGE2 significantly reduced in vitro apoptotic susceptibility of human CD34+ cells to taxol and etoposide. This could, however, be ascribed to reduced proliferation rather than to a change in apoptosis signaling and BCL-2 protein regulation. Accordingly, 16,16-dimethyl-PGE2 (dmPGE2) did not accelerate regeneration of the human hematopoietic system in vivo Repeated treatment of sublethally irradiated xenograft mice with known antiapoptotic substances, such as human FLT3L and thrombopoietin (TPO), which suppress transcription of the proapoptotic BCL-2 proteins BIM and BMF, also only marginally promoted human hematopoietic regeneration in vivo.


Subject(s)
Autoantigens/pharmacology , Dinoprostone/pharmacology , Epidermal Growth Factor/pharmacology , Hematopoiesis/drug effects , Iodide Peroxidase/pharmacology , Iron-Binding Proteins/pharmacology , Membrane Proteins/pharmacology , Animals , Drug Evaluation , Humans , Mice , Mice, Knockout
4.
Biometals ; 31(3): 301-312, 2018 06.
Article in English | MEDLINE | ID: mdl-29516297

ABSTRACT

Human and bovine lactoferrin (hLf and bLf) are multifunctional iron-binding glycoprotein constitutively synthesized and secreted by glandular epithelial cells and by neutrophils following induction. HLf and bLf possess very high similarity of sequence. Therefore, most of the in vitro and in vivo studies are carried out with commercial bLf (cbLf), available in large quantities and recognized by Food and Drug Administration (FDA, USA) as a safe substance. Physico-chemical heterogeneity of different cbLf preparations influences their effectiveness. CbLf iron-saturation affects thermal stability and resistance to proteolysis. Moreover, other metal ions such as Al(III), Cu(II), Mg(II), Mn(II), Zn(II) are chelated by cbLf, even if at lower affinity than Fe(III). Ca(II) is also sequestered by the carboxylate groups of sialic acid present on glycan chains of cbLf thus provoking the release of LPS, contributing to bactericidal activity. Similarly to more than 50% of eukaryotic proteins, cbLf possesses five N-glycosylation sites, also contributing to the resistance to proteolysis and, putatively, to the protection of intestinal mucosa from pathogens. CbLfs possess several functions as anti-microbial, anti-biofilm, anti-adhesive, anti-invasive and anti-inflammatory activities. They are also relevant modulators of iron and inflammatory homeostasis. However, the efficacy of cbLfs in exerting several functions can be erratic mainly depending from integrity, degree of iron and other metal ions saturation, N-glycosylation sites and chains, desialylated forms, Ca(II) sequestration, presence of contaminants and finally the ability to enter inside nucleus.


Subject(s)
Chelating Agents/chemistry , Glycoproteins/chemistry , Iron-Binding Proteins/chemistry , Lactoferrin/chemistry , Animals , Cattle , Chelating Agents/pharmacology , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Glycoproteins/pharmacology , Humans , Iron/chemistry , Iron-Binding Proteins/pharmacology , Lactoferrin/pharmacology , Metals/chemistry , Protein Binding , United States , United States Food and Drug Administration
5.
Clin Sci (Lond) ; 130(11): 853-70, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27129098

ABSTRACT

The mitochondrion is a major site for the metabolism of the transition metal, iron, which is necessary for metabolic processes critical for cell vitality. The enigmatic mitochondrial protein, frataxin, is known to play a significant role in both cellular and mitochondrial iron metabolism due to its iron-binding properties and its involvement in iron-sulfur cluster (ISC) and heme synthesis. The inherited neuro- and cardio-degenerative disease, Friedreich's ataxia (FA), is caused by the deficient expression of frataxin that leads to deleterious alterations in iron metabolism. These changes lead to the accumulation of inorganic iron aggregates in the mitochondrial matrix that are presumed to play a key role in the oxidative damage and subsequent degenerative features of this disease. Furthermore, the concurrent dys-regulation of cellular antioxidant defense, which coincides with frataxin deficiency, exacerbates oxidative stress. Hence, the pathogenesis of FA underscores the importance of the integrated homeostasis of cellular iron metabolism and the cytoplasmic and mitochondrial redox environments. This review focuses on describing the pathogenesis of the disease, the molecular mechanisms involved in mitochondrial iron-loading and the dys-regulation of cellular antioxidant defense due to frataxin deficiency. In turn, current and emerging therapeutic strategies are also discussed.


Subject(s)
Friedreich Ataxia/drug therapy , Homeostasis/drug effects , Iron-Binding Proteins/pharmacology , Iron/metabolism , Mitochondria/drug effects , Oxidative Stress/drug effects , Animals , Friedreich Ataxia/metabolism , Humans , Mitochondria/metabolism , Frataxin
6.
J Thromb Haemost ; 13(8): 1479-93, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26039631

ABSTRACT

BACKGROUND: Aspirin and P2Y12 antagonists are antiplatelet compounds that are used clinically in patients with thrombosis. However, some patients are 'resistant' to antiplatelet therapy, which increases their risk of developing acute coronary syndromes. These patients often present with an underlying condition that is associated with altered levels of circulating platelet primers and platelet hyperactivity. Platelet primers cannot stimulate platelet activation, but, in combination with physiologic stimuli, significantly enhance platelet function. OBJECTIVES: To explore the role of platelet primers in resistance to antiplatelet therapy, and to evaluate whether phosphoinositide 3-kinase (PI3K) contributes to this process. METHODS AND RESULTS: We used platelet aggregation, thromboxane A2 production and ex vivo thrombus formation as functional readouts of platelet activity. Platelets were treated with the potent P2Y12 inhibitor AR-C66096, aspirin, or a combination of both, in the presence or absence of the platelet primers insulin-like growth factor-1 (IGF-1) and thrombopoietin (TPO), or the Gz-coupled receptor ligand epinephrine. We found that platelet primers largely overcame the inhibitory effects of antiplatelet compounds on platelet functional responses. IGF-1-mediated and TPO-mediated, but not epinephrine-mediated, enhancements in the presence of antiplatelet drugs were blocked by the PI3K inhibitors wortmannin and LY294002. CONCLUSIONS: These results demonstrate that platelet primers can contribute to antiplatelet resistance. Furthermore, our data demonstrate that there are PI3K-dependent and PI3K-independent mechanisms driving primer-mediated resistance to antiplatelet therapy.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Aspirin/pharmacology , Blood Platelets/drug effects , Drug Resistance , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Adenosine Triphosphate/pharmacology , Autoantigens/pharmacology , Biomarkers/metabolism , Blood Coagulation/drug effects , Blood Platelets/metabolism , CD36 Antigens/metabolism , Epinephrine/pharmacology , Humans , Insulin-Like Growth Factor I/pharmacology , Iodide Peroxidase/pharmacology , Iron-Binding Proteins/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Platelet Aggregation/drug effects , Protein Kinase Inhibitors/pharmacology , Receptor, PAR-1/metabolism , Thromboxane A2/metabolism
7.
Clin Exp Immunol ; 180(1): 58-69, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25412700

ABSTRACT

T helper type 17 (Th17) cells play a pathogenic role in autoimmune disease, while interleukin (IL)-10-producing Th10 cells serve a protective role. The balance between the two subsets is regulated by the local cytokine milieu and by the relative expression of intact forkhead box protein 3 (FoxP3) compared to FoxP3Δ2, missing exon 2. Th17 and Th10 cell differentiation has usually been studied using polyclonal stimuli, and little is known about the ability of physiologically relevant self-antigens to induce Th17 or Th10 cell differentiation in autoimmune thyroid disease. We subjected mononuclear cells from healthy donors and patients with Hashimoto's thyroiditis (HT) or Graves' disease (GD) to polyclonal stimulation, or stimulation with human thyroglobulin (TG), human thyroid peroxidase (TPO), or Esherichia coli lipopolysaccharide (LPS). TPO and LPS induced increased differentiation of naive CD4(+) CD45RA(+) CD45R0(-) T cells from HT patients into Th17 cells. Th10 cell proportions were decreased in HT after polyclonal stimulation, but were comparable to those of healthy donors after antigen-specific stimulation. Taken together, our data show that an increased Th17 : Th10 ratio was found in HT patients after stimulation with thyroid-specific self-antigens. We also observed an elevated baseline production of IL-6 and transforming growth factor (TGF)-ß1 and of mRNA encoding FoxP3Δ2 rather than intact FoxP3. This may contribute to the skewing towards Th17 cell responses in HT.


Subject(s)
Alternative Splicing/immunology , Cell Differentiation/immunology , Forkhead Transcription Factors/immunology , Graves Disease/immunology , Hashimoto Disease/immunology , Th17 Cells/immunology , Adult , Aged , Alternative Splicing/drug effects , Antigens, CD/immunology , Autoantigens/immunology , Autoantigens/pharmacology , Cell Differentiation/drug effects , Escherichia coli/chemistry , Female , Graves Disease/pathology , Hashimoto Disease/pathology , Humans , Interleukin-10/immunology , Interleukin-6/immunology , Iodide Peroxidase/immunology , Iodide Peroxidase/pharmacology , Iron-Binding Proteins/immunology , Iron-Binding Proteins/pharmacology , Lipopolysaccharides/chemistry , Lipopolysaccharides/pharmacology , Male , Middle Aged , Protein Isoforms/immunology , Th17 Cells/pathology , Thyroglobulin/immunology , Thyroglobulin/pharmacology , Transforming Growth Factor beta1/immunology
8.
Neurochem Res ; 36(12): 2452-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21882036

ABSTRACT

Frataxin plays important roles in the mitochondrial respiratory chain and in the differentiation of neurons during early development. In this study, we observed the effects of frataxin on cell proliferation and neuroblast differentiation in the mouse hippocampal dentate gyrus. For this, we constructed an expression vector, PEP-1, that was fused with frataxin to create a PEP-1-frataxin fusion protein that easily penetrated frataxin into the blood-brain barrier. Three mg/kg PEP-1-frataxin was intraperitoneally administered to mice once a day for 2 weeks. The administration of PEP-1 alone did not result in any significant changes in the number of Ki67-positive cells and doublecortin (DCX)-immunoreactive neuroblasts in the mouse dentate gyrus. However, the administration of PEP-1-frataxin significantly increased the number of Ki67-positive cells and DCX-immunoreactive neuroblasts in the mouse dentate gyrus. In addition, PEP-1-frataxin significantly reduced 4-hydroxynonenal protein levels and malondialdehyde formation, while Cu, Zn-superoxide dismutase protein levels were maintained. These results suggest that frataxin effectively increased cell proliferation and neuroblast differentiation by decreasing lipid peroxidation in the dentate gyrus.


Subject(s)
Cell Proliferation/drug effects , Cysteamine/analogs & derivatives , Dentate Gyrus/physiology , Iron-Binding Proteins/pharmacology , Lipid Peroxidation/drug effects , Peptides/pharmacology , Recombinant Fusion Proteins/pharmacology , Aldehydes/metabolism , Animals , Cell Differentiation/drug effects , Cysteamine/pharmacology , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Doublecortin Domain Proteins , Doublecortin Protein , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Neural Stem Cells , Neurogenesis , Neuropeptides/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Frataxin
9.
Int J Biol Macromol ; 49(3): 311-6, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21600915

ABSTRACT

Artemin acts as a molecular chaperone by protecting Artemia embryos undergoing encystment from damage, caused by heat or other forms of stress. According to the amino acid sequence alignment, although artemin shows a fair amount of homology with ferritin, it also contains an extra C-terminal. Analysis of the C-terminal extension of artemin model in previous studies has shown that there are some favorable interactions between this region and its surrounding cleft. In the current study we tried to investigate the role of this C-terminal in chaperone activity of artemin. This extra C-terminal (39 residues) was deleted and the truncated gene was cloned and expressed in Escherichia coli. According to in vivo chaperone-like activity studies, both full-length and C-terminal truncated artemin conferred thermotolerance on transfected E. coli cells. However, bacteria expressing truncated derivative of artemin was less resistant than those producing native artemin against heat. Moreover, the activity recovery on carbonic anhydrase (CA), as protein substrate, was less in the presence of truncated artemin than that of full-length artemin. The results demonstrated that C-terminal deletion decreases the ability of artemin for chaperone-like activity. Theoretical investigations showed that deletion of artemin C-terminal extension makes substantial structural alterations in a way that structural stability and overall integrity of artemin decrease.


Subject(s)
Artemia , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/metabolism , Peptide Fragments/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Sequence Deletion , Amino Acid Sequence , Animals , Artemia/chemistry , Artemia/genetics , Arthropod Proteins , Carbonic Anhydrases/chemistry , Cloning, Molecular , Escherichia coli/cytology , Escherichia coli/genetics , Escherichia coli/physiology , Heat-Shock Response/genetics , Humans , Iron-Binding Proteins/genetics , Iron-Binding Proteins/pharmacology , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Refolding/drug effects , RNA-Binding Proteins/genetics , RNA-Binding Proteins/pharmacology
10.
J Neurol Sci ; 298(1-2): 64-9, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20817181

ABSTRACT

Reactive oxygen species (ROS) actively contribute to the development of a number of human diseases including ischemia. In response to oxidative stress, frataxin has a significant ability to improve cell survival though its biological function is unclear in relation to ischemia. To explore frataxin's role in protecting against ischemic cell death, we constructed PEP-1-Frataxin cell-permeable fusion protein. In a dose- and time-dependent manner PEP-1-Frataxin rapidly transduced into astrocyte cells and protected them against oxidative stress-induced cell death. Further, using an animal model, immunohistochemical analysis revealed that PEP-1-Frataxin prevented neuronal cell death in the CA1 region of the hippocampus induced by transient forebrain ischemia. These results demonstrate that transduced PEP-1-Frataxin protects against cell death in vitro and in vivo, suggesting that transduction of PEP-1-Frataxin could be useful as a therapeutic agent for various human diseases related to oxidative stress.


Subject(s)
Cysteamine/analogs & derivatives , Iron-Binding Proteins/pharmacology , Neurons/drug effects , Neuroprotective Agents , Oxidative Stress/drug effects , Peptides/pharmacology , Animals , Astrocytes/drug effects , Blood-Brain Barrier/metabolism , CA1 Region, Hippocampal/pathology , Cell Death/drug effects , Cell Line, Tumor , Coloring Agents , Cysteamine/pharmacology , Dose-Response Relationship, Drug , Genetic Vectors , Gerbillinae , Humans , Iron-Binding Proteins/biosynthesis , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/pathology , Microscopy, Fluorescence , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/pharmacology , Tetrazolium Salts , Thiazoles , Transduction, Genetic , Frataxin
11.
Cancer Lett ; 293(1): 58-64, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20083341

ABSTRACT

Oxidative DNA damage is the most critical factor implicated in carcinogenesis and other disorders. However, the protective effects of lunasin against oxidative DNA damage have not yet reported. In this study, we report here the protective effect of lunasin purified from Solanum nigrum L. against oxidative DNA. Lunasin protected DNA from the oxidative damage induced by Fe(2+) ion and hydroxyl radical. To better understand the mechanism for the protective effect of lunasin against DNA damage, the abilities to chelate Fe(2+), scavenge the generated hydroxyl radical and block the generation of hydroxyl radical were evaluated. Although it did not scavenge generated hydroxyl radical, lunasin blocked the generation of hydroxyl radical by chelating Fe(2+) ion. We conclude that lunasin protects DNA from oxidation by blocking fenton reaction between Fe(2+) and H(2)O(2) by chelating Fe(2+) and that consumption of lunasin may play an important role in the chemoprevention for the oxidative carcinogenesis.


Subject(s)
Anticarcinogenic Agents/pharmacology , DNA Damage , DNA/metabolism , Iron-Binding Proteins/pharmacology , Plant Proteins/pharmacology , Solanum nigrum/chemistry , Animals , Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/isolation & purification , Antioxidants/pharmacology , Ferrous Compounds/chemistry , Ferrous Compounds/metabolism , Free Radical Scavengers/pharmacology , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , Hydroxyl Radical/antagonists & inhibitors , Hydroxyl Radical/metabolism , Iron/antagonists & inhibitors , Iron/pharmacology , Iron Chelating Agents/pharmacology , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/isolation & purification , Mice , NIH 3T3 Cells , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Plant Proteins/chemistry , Plant Proteins/isolation & purification , Seeds/chemistry
12.
Hum Mol Genet ; 19(7): 1221-9, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20053667

ABSTRACT

The inability to produce normal levels of the mitochondrial protein frataxin causes the hereditary degenerative disorder Friedreich's Ataxia (FRDA), a syndrome characterized by progressive gait instability, cardiomyopathy and high incidence of diabetes. Frataxin is an iron-binding protein involved in the biogenesis of iron-sulfur clusters (ISC), prosthetic groups allowing essential cellular functions such as oxidative phosphorylation, enzyme catalysis and gene regulation. Although several evidence suggest that frataxin acts as an iron-chaperone within the mitochondrial compartment, we have recently demonstrated the existence of a functional extramitochondrial pool of mature frataxin in various human cell types. Here, we show that a similar proteolytic process generates both mature mitochondrial and extramitochondrial frataxin. To address the physiological function of human extramitochondrial frataxin, we searched for ISC-dependent interaction partners. We demonstrate that the extramitochondrial form of frataxin directly interacts with cytosolic aconitase/iron regulatory protein-1 (IRP1), a bifunctional protein alternating between an enzymatic and a RNA-binding function through the 'iron-sulfur switch' mechanism. Importantly, we found that the cytosolic aconitase defect and consequent IRP1 activation occurring in FRDA cells are reversed by the action of extramitochondrial frataxin. These results provide new insight into the control of cytosolic aconitase/IRP1 switch and expand current knowledge about the molecular pathogenesis of FRDA.


Subject(s)
Aconitate Hydratase/metabolism , Cytosol/metabolism , Iron Regulatory Protein 1/metabolism , Iron-Binding Proteins/pharmacology , Aconitate Hydratase/genetics , Cells, Cultured , Friedreich Ataxia/genetics , Gene Expression Regulation , Humans , Iron Regulatory Protein 1/genetics , Frataxin
13.
Blood ; 111(7): 3415-23, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18202223

ABSTRACT

Hematopoietic stem cells (HSCs) are the basis of bone marrow transplantation and are attractive target cells for hematopoietic gene therapy, but these important clinical applications have been severely hampered by difficulties in ex vivo expansion of HSCs. In particular, the use of cord blood for adult transplantation is greatly limited by the number of HSCs. Previously we identified angiopoietin-like proteins and IGF-binding protein 2 (IGFBP2) as new hormones that, together with other factors, can expand mouse bone marrow HSCs in culture. Here, we measure the activity of multipotent human severe combined immunodeficient (SCID)-repopulating cells (SRCs) by transplantation into the nonobese diabetic SCID (NOD/SCID) mice; secondary transplantation was performed to evaluate the self-renewal potential of SRCs. A serum-free medium containing SCF, TPO, and FGF-1 or Flt3-L cannot significantly support expansion of the SRCs present in human cord blood CD133+ cells. Addition of either angiopoietin-like 5 or IGF-binding protein 2 to the cultures led to a sizable expansion of HSC numbers, as assayed by NOD/SCID transplantation. A serum-free culture containing SCF, TPO, FGF-1, angiopoietin-like 5, and IGFBP2 supports an approximately 20-fold net expansion of repopulating human cord blood HSCs, a number potentially applicable to several clinical processes including HSC transplantation.


Subject(s)
Cell Culture Techniques , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Insulin-Like Growth Factor Binding Protein 2/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , AC133 Antigen , Angiopoietin-like Proteins , Angiopoietins , Animals , Antigens, CD , Autoantigens/pharmacology , Culture Media, Conditioned/pharmacology , Culture Media, Serum-Free/pharmacology , Fetal Blood/metabolism , Fibroblast Growth Factor 1/pharmacology , Genetic Therapy , Glycoproteins , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Iodide Peroxidase/pharmacology , Iron-Binding Proteins/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Peptides , Transplantation, Heterologous
14.
J Food Sci ; 72(1): S019-29, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17995893

ABSTRACT

BACKGROUND: The enhancing effect of meat on nonheme iron bioavailability in humans is thought to be due to the release of low-molecular-weight (LMW) iron-binding peptides during digestion. OBJECTIVE: To better characterize the LMW iron-binding peptides from meat digests. METHODS: Cooked beef, chicken, cod, lamb, and pork myofibrillar or sarcoplasmic protein extracts, casein, and egg albumin were digested in vitro with pepsin or pepsin/pancreatin. Ultrafiltrates were analyzed for N and iron and further characterized by gel filtration with added 59Fe, amino acid analysis, and LC-MS. RESULTS: 84% to 98% of total iron in enzymic digests was associated with soluble LMW peptides (< 10 kDa) of the myofibrillar proteins compared to only 2% to 20% in the corresponding sarcoplasmic protein digests. Pepsin digestion alone of the myobrillar proteins generated > 80% soluble LMW iron, compared to < 5% with casein and egg albumin. Iron-binding peptides from myofibrillar protein with an estimated 2 kDa molecular mass were separated by gel filtration. Peptides in this fraction were enriched in aspartic and glutamic acid residues and included potential peptide fragments of myosin. CONCLUSION: LMW (< 10 kDa) peptides in enzyme digests of myofibrillar proteins were the major facilitators of iron solubility. Unlike with casein, egg albumin, and most sarcoplasmic proteins, these LMW peptides were generated on pepsin digestion. One group of iron-binding peptides had a mass of approximately 2 kDa and was enriched in glutamic and aspartic acids. Such early generation of a multitude of LMW iron-binding peptides could explain the enhancing effect of muscle tissue on iron absorption.


Subject(s)
Amino Acids/analysis , Iron, Dietary/pharmacokinetics , Iron-Binding Proteins/pharmacology , Meat/analysis , Muscle Proteins/metabolism , Animals , Biological Availability , Cattle , Chickens , Digestion , Filtration , Gadus morhua , Humans , Intestinal Absorption , Molecular Weight , Muscle, Skeletal/metabolism , Sheep , Solubility , Species Specificity , Swine
15.
Biosci Biotechnol Biochem ; 71(6): 1505-13, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17587682

ABSTRACT

In a previous study of ours, the superoxide scavenging activity of aqueous extracts from dinophycean red tide flagellates was detected by an electron spin resonance (ESR)-spin trapping method, but not by an L-012 (luminol analog)-dependent chemiluminescence (CL) method. To investigate the discrepancy between the two methods, the effect of ferric-protein complexes on superoxide scavenging activity was examined. The reduced signal intensity of 5,5-dimethyl-1-pyrroline-N-oxide (DMPO)-OOH due to superoxide dismutase (SOD) did not change with the addition of horseradish peroxidase (HRP), while the reduced CL response due to SOD was restored by the addition of different concentrations of HRP. Since HRP is a ferric-protein complex, the effects of other ferric-protein complexes, catalase and hemoglobin, on the reduced CL response due to SOD were examined, and similar results were obtained. As is the case with SOD, the reduced CL response activity due to an aqueous extract from a raphidophycean red tide flagellate, Chattonella ovata, was also enhanced by HRP, catalase, and hemoglobin. ESR spectra analyzed at 77 K indicated that aqueous extracts of Gymnodinium impudicum and Alexandrium affine, both of which are dinophycean red tide flagellates, contained a ferric-protein complex, and that an extract of C. ovata did not. These results suggest that the presence of such a ferric-protein complex is a causative factor in the discrepancy between the ESR and luminol CL methods when determining superoxide scavenging activity.


Subject(s)
Electron Spin Resonance Spectroscopy/methods , Free Radical Scavengers/metabolism , Iron-Binding Proteins/pharmacology , Luminescent Measurements/methods , Superoxides/metabolism , Electron Spin Resonance Spectroscopy/standards , Luminescent Measurements/standards , Luminol , Methods , Reproducibility of Results , Spin Labels
16.
Cancer Gene Ther ; 13(1): 74-81, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16052231

ABSTRACT

While external ionizing radiation has been used for treating non-small cell lung cancer (NSCLC), improved efficacy of this modality would be an important advance. Ectopic expression of the sodium iodide symporter (NIS) and thyroperoxidase (TPO) genes in NSCLC cells facilitated concentration of iodide in NSCLC cells, which markedly induced apoptosis in vitro and in vivo. Pre-incubation of the NIS/TPO-modified NSCLC cells in iodide followed by ionizing radiation generates bystander tumoricidal effects and potently enhances tumor cell killing. This iodide-induced bystander effect is associated with enhanced gap junction intercellular communication (GJIC) activity and increased connexin-43 (Cx43) expression. Thus, iodide may serve as an enhancer to markedly improve the efficacy of radiation therapy in combined therapeutic modalities.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cell Death/radiation effects , Iodides/pharmacology , Lung Neoplasms/metabolism , Apoptosis/drug effects , Autoantigens/metabolism , Autoantigens/pharmacology , Bystander Effect , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Death/drug effects , Cell Line, Tumor , Connexin 43/metabolism , Genetic Therapy , Humans , Iodide Peroxidase/metabolism , Iodide Peroxidase/pharmacology , Iodides/metabolism , Iron-Binding Proteins/metabolism , Iron-Binding Proteins/pharmacology , Lung Neoplasms/genetics , Radiation, Ionizing , Sensitivity and Specificity , Symporters/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...