Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Blood ; 130(10): 1243-1246, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28739636

RESUMEN

Erythroferrone (ERFE) is a glycoprotein hormone secreted by erythroblasts in response to stimulation by erythropoietin (EPO). We previously demonstrated that ERFE messenger RNA expression and serum protein concentration increase in mice subjected to hemorrhage or EPO therapy, that ERFE acts on hepatocytes to suppress hepcidin, and that the resulting decrease in hepcidin augments iron delivery for intensified erythropoiesis. We also showed that ERFE contributes to pathological hepcidin suppression and iron overload in mice with nontransfused ß-thalassemia. We now report the development and technical validation of a rabbit monoclonal antibody-based sandwich immunoassay for human ERFE. We use this assay to show that blood loss or EPO administration increases serum ERFE concentrations in humans, and that patients with both nontransfused and transfused ß-thalassemia have very high serum ERFE levels, which decrease after blood transfusion. The assay should be useful for human studies of normal and disordered erythropoiesis and its effect on iron homeostasis.


Asunto(s)
Inmunoensayo/métodos , Hormonas Peptídicas/sangre , Adulto , Anciano , Donantes de Sangre , Eritropoyesis , Hepcidinas/sangre , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Talasemia beta/sangre
2.
Br J Haematol ; 167(5): 692-6, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25209728

RESUMEN

In transfusional iron overload, extra-hepatic iron distribution differs, depending on the underlying condition. Relative mechanisms of plasma non-transferrin bound iron (NTBI) generation may account for these differences. Markers of iron metabolism (plasma NTBI, labile iron, hepcidin, transferrin, monocyte SLC40A1 [ferroportin]), erythropoiesis (growth differentiation factor 15, soluble transferrin receptor) and tissue hypoxia (erythropoietin) were compared in patients with Thalassaemia Major (TM), Sickle Cell Disease and Diamond-Blackfan Anaemia (DBA), with matched transfusion histories. The most striking differences between these conditions were relationships of NTBI to erythropoietic markers, leading us to propose three mechanisms of NTBI generation: iron overload (all), ineffective erythropoiesis (predominantly TM) and low transferrin-iron utilization (DBA).


Asunto(s)
Anemia de Diamond-Blackfan/sangre , Anemia de Células Falciformes/sangre , Hierro/sangre , Talasemia/sangre , Transferrina , Adolescente , Adulto , Anemia de Diamond-Blackfan/terapia , Anemia de Células Falciformes/terapia , Biomarcadores/sangre , Proteínas Sanguíneas/metabolismo , Transfusión Sanguínea , Eritropoyesis , Femenino , Humanos , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/etiología , Masculino , Talasemia/terapia
3.
Br J Haematol ; 160(3): 399-403, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23216540

RESUMEN

This exploratory study assessed apoptosis in peripheral blood leucocytes (PBL) from ß-thalassaemia patients receiving chronic transfusions and chelation therapy (deferasirox or deferoxamine) at baseline, 1, 6, and 12 months. At baseline, thalassaemic PBLs presented 50% greater levels of Bax (BAX), 75% higher caspase-3/7, 48% higher caspase-8 and 88% higher caspase-9 activities and 428% more nucleosomal DNA fragmentation than control subjects. Only neutrophils correlated significantly with apoptotic markers. Previously, we showed that over the treatment year, hepatic iron declined; we now show that the ratio of Bax/Bcl-2 (BCL2), (-27·3%/year), and caspase-9 activity (-13·3%/year) declined in both treatment groups, suggesting that chelation decreases body iron and indicators of PBL apoptosis.


Asunto(s)
Apoptosis , Leucocitos/metabolismo , Talasemia beta/metabolismo , Adolescente , Adulto , Transfusión Sanguínea , Caspasas/metabolismo , Terapia por Quelación , Niño , Preescolar , Fragmentación del ADN , Femenino , Humanos , Masculino , Adulto Joven , Proteína X Asociada a bcl-2/metabolismo , Talasemia beta/terapia
4.
Complement Ther Med ; 70: 102856, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35843474

RESUMEN

OBJECTIVES: L-Glutamine was FDA-approved for sickle cell disease (SCD) in 2017, yet the mechanism(s)-of-action are poorly understood. This study investigates the potential activation of autophagy as a previously unexplored mechanism-of-benefit. DESIGN: Prospective, open-label, 8-week, phase-2 trial of oral L-glutamine (10 g TID) in patients with SCD at risk for pulmonary hypertension identified by Doppler-echocardiography by an elevated tricuspid-regurgitant-jet-velocity (TRV)≥ 2.5 m/s. Peripheral blood mononuclear cells (PBMCs) were isolated from blood samples taken from SCD patients at baseline, two, four, six and eight weeks of glutamine therapy, and from controls at baseline; BAX (pro-apoptotic marker) and LC3-II/LC3-I (autophagy marker) were measured via western blot analysis to assess apoptosis and autophagy respectively. SETTING: Comprehensive SCD Center in Oakland, California. RESULTS: Patients with SCD (n = 8) had a mean age of 44 ± 16, 50% were male; 63% Hb-SS, and mean TRV= 3.1 ± 0.7 m/s. Controls' mean age (n = 5) was 32 ± 12% and 57% were male; all were Hb-AA with a mean TRV= 1.8 ± 0.6. At baseline, SCD-PBMCs had 2-times higher levels of BAX and LC3-I versus controls (both p = 0.03). Levels of BAX expression increased by 300% after 8-weeks of glutamine supplementation (p = 0.005); LC3-I protein levels decreased while LC3-II levels increased by 70%, giving a significant increase in the LC3-II/LC3-I ratio (p = 0.02). CONCLUSION: PBMCs from glutamine-supplemented SCD patients have upregulated apoptotic and autophagy proteins. The parallel increase in BAX and the LC3-II / LC3-I ratio with glutamine supplementation suggest a possible role of autophagic cell death. The increase in apoptotic markers provide insight into a possible mechanism used by peripheral PBMCs during glutamine supplementation in patients with SCD.


Asunto(s)
Anemia de Células Falciformes , Suplementos Dietéticos , Glutamina/uso terapéutico , Leucocitos Mononucleares/fisiología , Insuficiencia de la Válvula Tricúspide , Adulto , Apoptosis , Autofagia , Biomarcadores , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Proteína X Asociada a bcl-2
5.
Brain Res ; 1748: 147066, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32818532

RESUMEN

Phospholipase A2G6-associated neurodegeneration (PLAN) is a rare early-onset monogenic neurodegenerative movement disorder which targets the basal ganglia and other regions in the central and peripheral nervous system; presenting as a series of heterogenous subtypes in patients. We describe here a B6.C3-Pla2g6m1J/CxRwb mouse model of PLAN which presents with early-onset neurodegeneration at 90 days which is analogous of the disease progression that is observed in PLAN patients. Homozygous mice had a progressively worsening motor deficit, which presented as tremors starting at 65 days and progressed to severe motor dysfunction and increased falls on the wire hang test at 90 days. This motor deficit positively correlated with a reduction in tyrosine hydroxylase (TH) protein expression in dopaminergic neurons of the substantia nigra (SN) without any neuronal loss. Fluorescence imaging of Thy1-YFP revealed spheroid formation in the SN. The spheroids in homozygous mice strongly mirrors those observed in patients and were demonstrated to correlate strongly with the motor deficits as measured by the wire hang test. The appearance of spheroids preceded TH loss and increased spheroid numbers negatively correlated with TH expression. Perls/DAB staining revealed the presence of iron accumulation within the SN of mice. This mouse model captures many of the major hallmarks of PLAN including severe-early onset neurodegeneration, a motor deficit that correlates directly to TH levels, spheroid formation and iron accumulation within the basal ganglia. Thus, this mouse line is a useful tool for further research efforts to improve understanding of how these disease mechanisms give rise to the disease presentations seen in PLAN patients as well as to test novel therapies.


Asunto(s)
Ganglios Basales/metabolismo , Hierro/metabolismo , Destreza Motora/fisiología , Distrofias Neuroaxonales/metabolismo , Sustancia Negra/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Animales , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Ratones
6.
Acta Haematol ; 122(2-3): 174-83, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19907155

RESUMEN

This review highlights recent advances in iron metabolism that are relevant to sickle cell disease (SCD). SCD is a common hemoglobinopathy that results in chronic inflammation. Improved understanding of how iron metabolism is controlled by proteins such as hepcidin, ferroportin, hypoxia-inducible factor 1, and growth differentiation factor 15 have revealed how they are involved in the organ toxicity of SCD. SCD patients have lower levels of non-transferrin-bound iron (NTBI) relative to other hemoglobinopathies, such as thalassemia. Care for SCD now commonly uses transfusion that results in iron overload and necessitates the need for chelation. New oral chelation therapy using deferasirox (Exjade/ICL670) appears to be safe and may even lower the amount of toxic free NTBI and enhance patient compliance. Finally, we suggest that iron metabolism and trafficking is different in SCD compared to other hemoglobinopathies. The high levels of inflammatory cytokines in SCD may enhance macrophage/reticuloendothelial cell iron and/or renal cell iron retention. This makes the tissues that retain iron different in SCD, and thus the organs that fail in SCD are different from those of other hemoglobinopathies, such as the cardiomyopathy or endocrinopathies of thalassemia.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Quelantes del Hierro/uso terapéutico , Hierro/metabolismo , Anemia de Células Falciformes/fisiopatología , Humanos
7.
Cell Stem Cell ; 25(4): 531-541.e6, 2019 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-31585094

RESUMEN

Pelizaeus-Merzbacher disease (PMD) is an X-linked leukodystrophy caused by mutations in Proteolipid Protein 1 (PLP1), encoding a major myelin protein, resulting in profound developmental delay and early lethality. Previous work showed involvement of unfolded protein response (UPR) and endoplasmic reticulum (ER) stress pathways, but poor PLP1 genotype-phenotype associations suggest additional pathogenetic mechanisms. Using induced pluripotent stem cell (iPSC) and gene-correction, we show that patient-derived oligodendrocytes can develop to the pre-myelinating stage, but subsequently undergo cell death. Mutant oligodendrocytes demonstrated key hallmarks of ferroptosis including lipid peroxidation, abnormal iron metabolism, and hypersensitivity to free iron. Iron chelation rescued mutant oligodendrocyte apoptosis, survival, and differentiationin vitro, and post-transplantation in vivo. Finally, systemic treatment of Plp1 mutant Jimpy mice with deferiprone, a small molecule iron chelator, reduced oligodendrocyte apoptosis and enabled myelin formation. Thus, oligodendrocyte iron-induced cell death and myelination is rescued by iron chelation in PMD pre-clinical models.


Asunto(s)
Deferiprona/uso terapéutico , Células Madre Pluripotentes Inducidas/fisiología , Quelantes del Hierro/uso terapéutico , Hierro/metabolismo , Proteína Proteolipídica de la Mielina/metabolismo , Oligodendroglía/fisiología , Enfermedad de Pelizaeus-Merzbacher/terapia , Animales , Diferenciación Celular , Células Cultivadas , Ferroptosis , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/trasplante , Peroxidación de Lípido , Ratones , Ratones Mutantes , Mutación/genética , Proteína Proteolipídica de la Mielina/genética , Oligodendroglía/efectos de los fármacos , Oligodendroglía/trasplante , Enfermedad de Pelizaeus-Merzbacher/genética , Enfermedad de Pelizaeus-Merzbacher/patología , Trasplante de Células Madre , Reparación del Gen Blanco
8.
Haematologica ; 93(6): 817-25, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18469351

RESUMEN

BACKGROUND: We assessed whether oxidant-stress and inflammation in beta-thalassemia could be controlled by the novel oral iron chelator deferasirox as effectively as by deferoxamine. DESIGN AND METHODS: Forty-nine subjects were enrolled from seven sites and studied at baseline, and after 1, 6, and 12 months of therapy. Malondialdehyde, protein carbonyls, vitamins E and C, total non-transferrin bound iron, transferrin saturation, C-reactive protein, cytokines, serum ferritin concentration and liver iron concentration were measured. RESULTS: Liver iron concentration and ferritin declined significantly in both treatment groups during the study. This paralleled a significant decline in the oxidative-stress marker malondialdehyde (deferasirox -22%/year, deferoxamine -28%/year, average decline p=0.006). The rates of decline did not differ between treatment groups. Malondialdehyde was higher in both treatment groups than in a group of 30 non-thalassemic controls (p < 0.001). The inflammatory marker high-sensitivity C-reactive protein decreased significantly only in the group receiving deferasirox (deferasirox -51%/year, deferoxamine +8.5%/year, p = 0.02). This result was confounded by a chance difference in the level of high-sensitivity C-reactive protein between the two groups at baseline, but analyses controlling for this difference suggested an equally large treatment effect. CONCLUSIONS: Iron chelation therapy with deferoxamine or with deferasirox was equally effective in decreasing iron burden and malondialdehyde. The possible differential effect of the two chelators on inflammation warrants further investigation.


Asunto(s)
Benzoatos/farmacología , Deferoxamina/farmacología , Quelantes del Hierro/farmacología , Oxidantes/metabolismo , Triazoles/farmacología , Talasemia beta/tratamiento farmacológico , Adolescente , Adulto , Ácido Ascórbico/metabolismo , Benzoatos/efectos adversos , Proteína C-Reactiva/metabolismo , Niño , Deferasirox , Deferoxamina/efectos adversos , Femenino , Humanos , Quelantes del Hierro/efectos adversos , Peroxidación de Lípido , Masculino , Estrés Oxidativo , Triazoles/efectos adversos , Vitamina E/metabolismo , Talasemia beta/patología
10.
Mutat Res ; 578(1-2): 15-22, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16202794

RESUMEN

We have measured the inhibition of the mutagenicity of the mycotoxin aflatoxin-B(1) in the liver of the rat by plant material of Scutellaria baicalensis, or Huang-qin. The addition of one percent dried Huang-qin to the feed of the animals reduced the mutant frequency of a subsequent administration of aflatoxin-B1 by approximately 60 and 77%, respectively, for two different batches of the plant material. The addition of Huang-qin also increased the expression of the gene for glutathione S-transferase A5 subunit by 2.5-3.0-fold, and decreased expression of P450 cytochrome 3A2 by 1.8-2.0-fold. The greater increase of the expression of the GST gene may result in the protection shown by Huang-qin. The sensitivity of the hepatic mitochondria to swelling, a measure of the mitochondrial permeability transition, is increased significantly in animals that are on a diet containing Huang-qin. This may lead to increased sensitivity to apoptosis on treatment with toxic compounds. The two batches of Huang-qin material show differences in both chemical composition and preventive potential. This study demonstrates how a combination of generating and analysis of plant varieties together with a mammalian assay for efficacy may improve the search for better plant-based prevention of cancer initiation.


Asunto(s)
Aflatoxina B1/toxicidad , Antimutagênicos/farmacología , Mutágenos/toxicidad , Scutellaria baicalensis/química , Scutellaria baicalensis/genética , Animales , Animales Modificados Genéticamente , Sistema Enzimático del Citocromo P-450/metabolismo , Inducción Enzimática/efectos de los fármacos , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Mitocondrias Hepáticas/efectos de los fármacos , Distribución Aleatoria , Ratas , Scutellaria baicalensis/metabolismo
11.
Br J Haematol ; 135(2): 254-63, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17010049

RESUMEN

Blood transfusion therapy is life-saving for patients with beta-thalassaemia and sickle cell disease (SCD), but often results in severe iron overload. This pilot study examined whether the biomarkers of tissue injury or inflammation differ in these two diseases. Plasma malondialdehyde (MDA) was significantly increased 1.8-fold in thalassaemia relative to control patients. In contrast, MDA in SCD was not significantly different from controls. In multivariate analysis, the strongest predictors of elevated MDA were liver iron concentration (P < 0.001) and specific diagnosis (P = 0.019). A significant 2-fold elevation of non-transferrin bound iron (NTBI) was observed in thalassaemia relative to SCD. NTBI was not a significant predictor of high MDA in multivariate analysis. SCD patients showed a significant 2.2-fold elevation of the inflammatory marker interleukin (IL)-6 relative to controls, and a 3.6- and 1.7-fold increase in IL-5 and IL-10 relative to thalassaemia. Although alpha-tocopherol was significantly decreased by at least 32% in both thalassaemia and SCD, indicating ongoing oxidant stress and antioxidant consumption, gamma-tocopherol, a nitric oxide-selective antioxidant, was increased 36% in SCD relative to thalassaemia. These results demonstrate that thalassaemia patients have increased MDA and circulating NTBI relative to SCD patients and lower levels of some cytokines and gamma-tocopherol. This supports the hypothesis that the biology of SCD may show increased inflammation and increased levels of protective antioxidants compared with thalassaemia.


Asunto(s)
Anemia de Células Falciformes/metabolismo , Inflamación/etiología , Sobrecarga de Hierro/metabolismo , Estrés Oxidativo , Talasemia beta/metabolismo , Adolescente , Adulto , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/terapia , Antioxidantes/metabolismo , Biomarcadores/sangre , Proteína C-Reactiva/metabolismo , Niño , Femenino , Ferritinas/metabolismo , Humanos , Interleucinas/sangre , Hierro/metabolismo , Sobrecarga de Hierro/complicaciones , Hígado/metabolismo , Masculino , Malondialdehído/sangre , Reacción a la Transfusión , Talasemia beta/complicaciones , Talasemia beta/terapia , gamma-Tocoferol/sangre
12.
Arch Biochem Biophys ; 414(2): 255-60, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12781777

RESUMEN

Iron and iron complexes stimulate lipid peroxidation and formation of malondialdehyde (MDA). We have studied the effects of Fe2+ and ascorbate on mitochondrial permeability transition induced by phosphate and Ca2+. Iron is necessary for detectable MDA formation, but only Ca2+ and phosphate are necessary for the induction of membrane potential loss (Deltapsi) and Ca2+ release. Keeping the iron at a constant concentration and varying the Ca2+ level changed the mitochondrial Ca2+ retention times, but not the amount of MDA formation. The antioxidant butylated hydroxytoluene at low concentrations prevented MDA formation, but not mitochondrial Ca2+ release. Preincubation of mitochondria with Fe2+ decreased Ca2+ retention time in a concentration-dependent manner and facilitated Ca2+-stimulated MDA accumulation. Thus, Ca2+ phosphate-induced mitochondrial permeability transition (MPT) can be separated mechanistically from MDA accumulation. Lipid peroxidation products do not appear to participate in the initial phase of the permeability transition, but sensitize mitochondria toward MPT.


Asunto(s)
Hierro/metabolismo , Peroxidación de Lípido , Mitocondrias/metabolismo , Animales , Antioxidantes/farmacología , Hidroxitolueno Butilado/farmacología , Calcio/metabolismo , Calcio/farmacología , Membranas Intracelulares/metabolismo , Hierro/farmacología , Cinética , Masculino , Malondialdehído/metabolismo , Potenciales de la Membrana , Mitocondrias Hepáticas/patología , Permeabilidad , Ratas , Factores de Tiempo
13.
Arch Biochem Biophys ; 397(2): 345-53, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11795893

RESUMEN

Mitochondria decay with age from oxidative damage and loss of protective mechanisms. Resistance, repair, and replacement mechanisms are essential for mitochondrial preservation and maintenance. Iron plays an essential role in the maintenance of mitochondria, through its two major functional forms: heme and iron-sulfur clusters. Both iron-based cofactors are formed and utilized in the mitochondria and then distributed throughout the cell. This is an important function of mitochondria that is not directly related to the production of ATP. Heme and iron-sulfur clusters are important for the normal assembly and for the optimal activity of the electron transfer complexes. Loss of mitochondrial cytochrome c oxidase (complex IV), integrity of mtDNA, and function can result from abnormal homeostasis of iron. We review the physiological role of iron-sulfur clusters and heme in the integrity of the mitochondria and the generation of oxidants.


Asunto(s)
Hemo/metabolismo , Proteínas Hierro-Azufre/metabolismo , Hierro/metabolismo , Mitocondrias/fisiología
14.
Arch Biochem Biophys ; 398(2): 198-202, 2002 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11831850

RESUMEN

Isolated mitochondria loaded with Ca(2+) and then exposed to Fe(2+) show a transient release of Ca(2+). The magnitude of this response depends on the Ca(2+) loading and the kinetics of the response depends on the concentration of added Fe(2+). We investigated the Fe(2+)-induced Ca(2+) release mechanism by measuring mitochondrial Ca(2+) uptake in the presence of Fe(2+). The presence of Fe(2+) inhibits Ca(2+) uptake two times. Since mitochondria can cycle Ca(2+) across their inner membrane, the suppression of Ca(2+) uptake, but not release, results in an elevation of the extramitochondrial Ca(2+), thereby varying the steady state. The transient release of Ca(2+) initially observed from mitochondria appears to occur via the electroneutral 2H(+)/Ca(2+)-exchange mechanism, since it can be markedly decreased by cyclosporin A and does not involve lipid peroxidation. When Fe(2+) accumulation is completed, reuptake of released Ca(2+) into mitochondria resumes. Finally, we propose that Fe(2+) either inhibits Ca(2+) entry at the uniporter or is transported by it into the matrix.


Asunto(s)
Calcio/metabolismo , Hierro/metabolismo , Mitocondrias Hepáticas/metabolismo , Animales , Transporte Iónico/efectos de los fármacos , Hierro/farmacología , Ratas
15.
Proc Natl Acad Sci U S A ; 99(4): 2264-9, 2002 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-11854522

RESUMEN

Approximately two billion people, mainly women and children, are iron deficient. Two studies examined the effects of iron deficiency and supplementation on rats. In study 1, mitochondrial functional parameters and mitochondrial DNA (mtDNA) damage were assayed in iron-deficient (< or =5 microg/day) and iron-normal (800 microg/day) rats and in both groups after daily high-iron supplementation (8,000 microg/day) for 34 days. This dose is equivalent to the daily dose commonly given to iron-deficient humans. Iron-deficient rats had lower liver mitochondrial respiratory control ratios and increased levels of oxidants in polymorphonuclear-leukocytes, as assayed by dichlorofluorescein (P < 0.05). Rhodamine 123 fluorescence of polymorphonuclear-leukocytes also increased (P < 0.05). Lowered respiratory control ratios were found in daily high-iron-supplemented rats regardless of the previous iron status (P < 0.05). mtDNA damage was observed in both iron-deficient rats and rats receiving daily high-iron supplementation, compared with iron-normal rats (P < 0.05). Study 2 compared iron-deficient rats given high doses of iron (8,000 microg) either daily or every third day and found that rats given iron supplements every third day had less mtDNA damage on the second and third day after the last dose compared to daily high iron doses. Both inadequate and excessive iron (10 x nutritional need) cause significant mitochondrial malfunction. Although excess iron has been known to cause oxidative damage, the observation of oxidant-induced damage to mitochondria from iron deficiency has been unrecognized previously. Untreated iron deficiency, as well as excessive-iron supplementation, are deleterious and emphasize the importance of maintaining optimal iron intake.


Asunto(s)
ADN Mitocondrial/genética , Deficiencias de Hierro , Hierro/farmacología , Mitocondrias/metabolismo , Animales , ADN Mitocondrial/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Colorantes Fluorescentes/farmacología , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias Hepáticas/efectos de los fármacos , Neutrófilos/metabolismo , Ratas , Ratas Sprague-Dawley , Respiración , Rodamina 123/farmacología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA