Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
bioRxiv ; 2023 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-37745522

RESUMO

Beta-Propeller Protein-Associated Neurodegeneration (BPAN) is one of the commonest forms of Neurodegeneration with Brain Iron Accumulation, caused by mutations in the gene encoding the autophagy-related protein, WDR45. The mechanisms linking autophagy, iron overload and neurodegeneration in BPAN are poorly understood and, as a result, there are currently no disease-modifying treatments for this progressive disorder. We have developed a patient-derived, induced pluripotent stem cell (iPSC)-based midbrain dopaminergic neuronal cell model of BPAN (3 patient, 2 age-matched controls and 2 isogenic control lines) which shows defective autophagy and aberrant gene expression in key neurodegenerative, neurodevelopmental and collagen pathways. A high content imaging-based medium-throughput blinded drug screen using the FDA-approved Prestwick library identified 5 cardiac glycosides that both corrected disease-related defective autophagosome formation and restored BPAN-specific gene expression profiles. Our findings have clear translational potential and emphasise the utility of iPSC-based modelling in elucidating disease pathophysiology and identifying targeted therapeutics for early-onset monogenic disorders.

2.
Mol Genet Metab ; 137(3): 283-291, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36240582

RESUMO

Studies aimed at supporting different treatment approaches for pantothenate kinase-associated neurodegeneration (PKAN) have revealed the complexity of coenzyme A (CoA) metabolism and the limits of our current knowledge about disease pathogenesis. Here we offer a foundation for critically evaluating the myriad approaches, argue for the importance of unbiased disease models, and highlight some of the outstanding questions that are central to our understanding and treating PKAN.


Assuntos
Neurodegeneração Associada a Pantotenato-Quinase , Humanos , Coenzima A/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo
3.
Antioxid Redox Signal ; 37(1-3): 150-170, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34569265

RESUMO

Significance: Iron accumulation occurs in the central nervous system (CNS) in a variety of neurological conditions as diverse as spinal cord injury, stroke, multiple sclerosis, Parkinson's disease, and others. Iron is a redox-active metal that gives rise to damaging free radicals if its intracellular levels are not controlled or if it is not properly sequestered within cells. The accumulation of iron occurs due to dysregulation of mechanisms that control cellular iron homeostasis. Recent Advances: The molecular mechanisms that regulate cellular iron homeostasis have been revealed in much detail in the past three decades, and new advances continue to be made. Understanding which aspects of iron homeostasis are dysregulated in different conditions will provide insights into the causes of iron accumulation and iron-mediated tissue damage. Recent advances in iron-dependent lipid peroxidation leading to cell death, called ferroptosis, has provided useful insights that are highly relevant for the lipid-rich environment of the CNS. Critical Issues: This review examines the mechanisms that control normal cellular iron homeostasis, the dysregulation of these mechanisms in neurological disorders, and more recent work on how iron can induce tissue damage via ferroptosis. Future Directions: Quick and reliable tests are needed to determine if and when ferroptosis contributes to the pathogenesis of neurological disorders. In addition, there is need to develop better druggable agents to scavenge lipid radicals and reduce CNS damage for neurological conditions for which there are currently few effective treatments. Antioxid. Redox Signal. 37, 150-170.


Assuntos
Ferroptose , Doenças Neurodegenerativas , Sistema Nervoso Central/metabolismo , Homeostase , Humanos , Ferro/metabolismo , Peroxidação de Lipídeos , Lipídeos
4.
EMBO Mol Med ; 11(12): e10489, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31660701

RESUMO

Pantothenate kinase-associated neurodegeneration (PKAN) is an inborn error of CoA metabolism causing dystonia, parkinsonism, and brain iron accumulation. Lack of a good mammalian model has impeded studies of pathogenesis and development of rational therapeutics. We took a new approach to investigating an existing mouse mutant of Pank2 and found that isolating the disease-vulnerable brain revealed regional perturbations in CoA metabolism, iron homeostasis, and dopamine metabolism and functional defects in complex I and pyruvate dehydrogenase. Feeding mice a CoA pathway intermediate, 4'-phosphopantetheine, normalized levels of the CoA-, iron-, and dopamine-related biomarkers as well as activities of mitochondrial enzymes. Human cell changes also were recovered by 4'-phosphopantetheine. We can mechanistically link a defect in CoA metabolism to these secondary effects via the activation of mitochondrial acyl carrier protein, which is essential to oxidative phosphorylation, iron-sulfur cluster biogenesis, and mitochondrial fatty acid synthesis. We demonstrate the fidelity of our model in recapitulating features of the human disease. Moreover, we identify pharmacodynamic biomarkers, provide insights into disease pathogenesis, and offer evidence for 4'-phosphopantetheine as a candidate therapeutic for PKAN.


Assuntos
Coenzima A/metabolismo , Dopamina/metabolismo , Ferro/metabolismo , Panteteína/análogos & derivados , Neurodegeneração Associada a Pantotenato-Quinase/tratamento farmacológico , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Animais , Biomarcadores/metabolismo , Genótipo , Camundongos , Panteteína/farmacologia , Panteteína/uso terapêutico , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo
5.
Mol Genet Genomic Med ; 7(7): e00736, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31087512

RESUMO

BACKGROUND: Mitochondrial membrane protein-associated neurodegeneration (MPAN) is caused by pathogenic sequence variants in C19orf12. Autosomal recessive inheritance has been demonstrated. We present evidence of autosomal dominant MPAN and propose a mechanism to explain these cases. METHODS: Two large families with apparently dominant MPAN were investigated; additional singleton cases of MPAN were identified. Gene sequencing and multiplex ligation-dependent probe amplification were used to characterize the causative sequence variants in C19orf12. Post-mortem brain from affected subjects was examined. RESULTS: In two multi-generation non-consanguineous families, we identified different nonsense sequence variations in C19orf12 that segregate with the MPAN phenotype. Brain pathology was similar to that of autosomal recessive MPAN. We additionally identified a preponderance of cases with single heterozygous pathogenic sequence variants, including two with de novo changes. CONCLUSIONS: We present three lines of clinical evidence to demonstrate that MPAN can manifest as a result of only one pathogenic C19orf12 sequence variant. We propose that truncated C19orf12 proteins, resulting from nonsense variants in the final exon in our autosomal dominant cohort, impair function of the normal protein produced from the non-mutated allele via a dominant negative mechanism and cause loss of function. These findings impact the clinical diagnostic evaluation and counseling.


Assuntos
Distúrbios do Metabolismo do Ferro/genética , Proteínas de Membrana/genética , Proteínas Mitocondriais/genética , Distrofias Neuroaxonais/genética , Adulto , Encéfalo , Códon sem Sentido/genética , Estudos de Coortes , Família , Feminino , Genes Dominantes/genética , Heterozigoto , Humanos , Distúrbios do Metabolismo do Ferro/metabolismo , Masculino , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/metabolismo , Mutação , Distrofias Neuroaxonais/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Linhagem
6.
Mol Genet Metab ; 116(4): 289-97, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26547561

RESUMO

Pantothenate kinase-associated neurodegeneration (PKAN) is a progressive movement disorder that is due to mutations in PANK2. Pathologically, it is a member of a class of diseases known as neurodegeneration with brain iron accumulation (NBIA) and features increased tissue iron and ubiquitinated proteinaceous aggregates in the globus pallidus. We have previously determined that these aggregates represent condensed residue derived from degenerated pallidal neurons. However, the protein content, other than ubiquitin, of these aggregates remains unknown. In the present study, we performed biochemical and immunohistochemical studies to characterize these aggregates and found them to be enriched in apolipoprotein E that is poorly soluble in detergent solutions. However, we did not determine a significant association between APOE genotype and the clinical phenotype of disease in our database of 81 cases. Rather, we frequently identified similar ubiquitin- and apolipoprotein E-enriched lesions in these neurons in non-PKAN patients in the penumbrae of remote infarcts that involve the globus pallidus, and occasionally in other brain sites that contain large γ-aminobutyric acid (GABA)ergic neurons. Our findings, taken together, suggest that tissue or cellular hypoxic/ischemic injury within the globus pallidus may underlie the pathogenesis of PKAN.


Assuntos
Apolipoproteínas E/química , Isquemia Encefálica/genética , Neurônios GABAérgicos/química , Neurodegeneração Associada a Pantotenato-Quinase/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Agregação Patológica de Proteínas/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Isquemia Encefálica/complicações , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Estudos de Casos e Controles , Criança , Feminino , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/patologia , Expressão Gênica , Globo Pálido/metabolismo , Globo Pálido/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Neurodegeneração Associada a Pantotenato-Quinase/complicações , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Agregação Patológica de Proteínas/complicações , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Ubiquitina/química , Ubiquitina/genética , Ubiquitina/metabolismo
7.
Hum Mol Genet ; 23(1): 24-39, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23943793

RESUMO

Iron-sulfur (Fe-S) clusters are ancient enzyme cofactors found in virtually all life forms. We evaluated the physiological effects of chronic Fe-S cluster deficiency in human skeletal muscle, a tissue that relies heavily on Fe-S cluster-mediated aerobic energy metabolism. Despite greatly decreased oxidative capacity, muscle tissue from patients deficient in the Fe-S cluster scaffold protein ISCU showed a predominance of type I oxidative muscle fibers and higher capillary density, enhanced expression of transcriptional co-activator PGC-1α and increased mitochondrial fatty acid oxidation genes. These Fe-S cluster-deficient muscles showed a dramatic up-regulation of the ketogenic enzyme HMGCS2 and the secreted protein FGF21 (fibroblast growth factor 21). Enhanced muscle FGF21 expression was reflected by elevated circulating FGF21 levels in the patients, and robust FGF21 secretion could be recapitulated by respiratory chain inhibition in cultured myotubes. Our findings reveal that mitochondrial energy starvation elicits a coordinated response in Fe-S-deficient skeletal muscle that is reflected systemically by increased plasma FGF21 levels.


Assuntos
Acidose Láctica/congênito , Fatores de Crescimento de Fibroblastos/metabolismo , Hidroximetilglutaril-CoA Sintase/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Músculo Esquelético/metabolismo , Doenças Musculares/congênito , Fatores de Transcrição/genética , Acidose Láctica/genética , Acidose Láctica/metabolismo , Acidose Láctica/patologia , Adulto , Idoso , Estudos de Casos e Controles , Células Cultivadas , Metabolismo Energético , Feminino , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Humanos , Hidroximetilglutaril-CoA Sintase/genética , Proteínas Ferro-Enxofre/genética , Masculino , Pessoa de Meia-Idade , Mitocôndrias Musculares/metabolismo , Mitocôndrias Musculares/patologia , Doenças Musculares/genética , Doenças Musculares/metabolismo , Doenças Musculares/patologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/metabolismo
8.
Cell Metab ; 17(2): 271-81, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23395173

RESUMO

Iron regulatory proteins (Irps) 1 and 2 posttranscriptionally control the expression of transcripts that contain iron-responsive element (IRE) sequences, including ferritin, ferroportin, transferrin receptor, and hypoxia-inducible factor 2α (HIF2α). We report here that mice with targeted deletion of Irp1 developed pulmonary hypertension and polycythemia that was exacerbated by a low-iron diet. Hematocrits increased to 65% in iron-starved mice, and many polycythemic mice died of abdominal hemorrhages. Irp1 deletion enhanced HIF2α protein expression in kidneys of Irp1(-/-) mice, which led to increased erythropoietin (EPO) expression, polycythemia, and concomitant tissue iron deficiency. Increased HIF2α expression in pulmonary endothelial cells induced high expression of endothelin-1, likely contributing to the pulmonary hypertension of Irp1(-/-) mice. Our results reveal why anemia is an early physiological consequence of iron deficiency, highlight the physiological significance of Irp1 in regulating erythropoiesis and iron distribution, and provide important insights into the molecular pathogenesis of pulmonary hypertension.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Deleção de Genes , Hipertensão Pulmonar/complicações , Proteína 1 Reguladora do Ferro/metabolismo , Policitemia/complicações , Biossíntese de Proteínas , Animais , Dieta , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotelina-1/genética , Endotelina-1/metabolismo , Eritropoetina/sangue , Hemorragia Gastrointestinal/sangue , Hemorragia Gastrointestinal/complicações , Hemorragia Gastrointestinal/patologia , Hematopoese Extramedular/efeitos dos fármacos , Hipertensão Pulmonar/sangue , Hipertensão Pulmonar/patologia , Ferro/farmacologia , Proteína 1 Reguladora do Ferro/deficiência , Proteína 2 Reguladora do Ferro/metabolismo , Longevidade , Camundongos , Modelos Biológicos , Degeneração Neural/sangue , Degeneração Neural/complicações , Degeneração Neural/patologia , Especificidade de Órgãos/efeitos dos fármacos , Policitemia/sangue , Policitemia/patologia , Biossíntese de Proteínas/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética
9.
J Biol Chem ; 287(48): 40119-30, 2012 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-23035118

RESUMO

BACKGROUND: ISCU myopathy is a disease caused by muscle-specific deficiency of the Fe-S cluster scaffold protein ISCU. RESULTS: MyoD expression enhanced ISCU mRNA mis-splicing, and oxidative stress exacerbated ISCU depletion in patient cells. CONCLUSION: ISCU protein deficiency in patients results from muscle-specific mis-splicing as well as oxidative stress. SIGNIFICANCE: Oxidative stress negatively influences the mammalian Fe-S cluster assembly machinery by destabilization of ISCU. Iron-sulfur (Fe-S) cluster cofactors are formed on the scaffold protein ISCU. ISCU myopathy is a disease caused by an intronic mutation that leads to abnormally spliced ISCU mRNA. We found that two predominant mis-spliced ISCU mRNAs produce a truncated and short-lived ISCU protein product in multiple patient cell types. Expression of the muscle-specific transcription factor MyoD further diminished normal splicing of ISCU mRNA in patient myoblasts, demonstrating that the process of muscle differentiation enhances the loss of normal ISCU mRNA splicing. ISCU protein was nearly undetectable in patient skeletal muscle, but was higher in patient myoblasts, fibroblasts, and lymphoblasts. We next treated patient cells with pro-oxidants to mimic the oxidative stress associated with muscle activity. Brief hydrogen peroxide treatment or incubation in an enriched oxygen atmosphere led to a marked further reduction of ISCU protein levels, which could be prevented by pretreatment with the antioxidant ascorbate. Thus, we conclude that skeletal muscle differentiation of patient cells causes a higher degree of abnormal ISCU splicing and that oxidative stress resulting from skeletal muscle work destabilizes the small amounts of normal ISCU protein generated in patient skeletal muscles.


Assuntos
Diferenciação Celular , Proteínas Ferro-Enxofre/genética , Doenças Mitocondriais/metabolismo , Músculo Esquelético/citologia , Estresse Oxidativo , Splicing de RNA , Adulto , Idoso , Animais , Feminino , Humanos , Proteínas Ferro-Enxofre/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Doenças Mitocondriais/genética , Doenças Mitocondriais/fisiopatologia , Músculo Esquelético/metabolismo , Proteína MyoD/genética , Proteína MyoD/metabolismo , Especificidade de Órgãos , Adulto Jovem
10.
PLoS One ; 7(4): e35241, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22529995

RESUMO

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by cortical and spinal motor neuron dysfunction. Routine magnetic resonance imaging (MRI) studies have previously shown hypointense signal in the motor cortex on T(2)-weighted images in some ALS patients, however, the cause of this finding is unknown. To investigate the utility of this MR signal change as a marker of cortical motor neuron degeneration, signal abnormalities on 3T and 7T MR images of the brain were compared, and pathology was obtained in two ALS patients to determine the origin of the motor cortex hypointensity. Nineteen patients with clinically probable or definite ALS by El Escorial criteria and 19 healthy controls underwent 3T MRI. A 7T MRI scan was carried out on five ALS patients who had motor cortex hypointensity on the 3T FLAIR sequence and on three healthy controls. Postmortem 7T MRI of the brain was performed in one ALS patient and histological studies of the brains and spinal cords were obtained post-mortem in two patients. The motor cortex hypointensity on 3T FLAIR images was present in greater frequency in ALS patients. Increased hypointensity correlated with greater severity of upper motor neuron impairment. Analysis of 7T T(2)(*)-weighted gradient echo imaging localized the signal alteration to the deeper layers of the motor cortex in both ALS patients. Pathological studies showed increased iron accumulation in microglial cells in areas corresponding to the location of the signal changes on the 3T and 7T MRI of the motor cortex. These findings indicate that the motor cortex hypointensity on 3T MRI FLAIR images in ALS is due to increased iron accumulation by microglia.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Córtex Cerebral/patologia , Ferro/metabolismo , Imageamento por Ressonância Magnética , Adulto , Córtex Cerebral/metabolismo , Feminino , Humanos , Corpos de Inclusão/patologia , Masculino , Microglia/metabolismo , Microglia/patologia , Pessoa de Meia-Idade , Córtex Motor/metabolismo , Córtex Motor/patologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Neuroimagem , Medula Espinal/patologia
11.
Cilia ; 1(1): 3, 2012 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23351752

RESUMO

BACKGROUND: The central tenet of cilia function is sensing and transmitting information. The capacity to directly contact extracellular surfaces would empower primary cilia to probe the environment for information about the nature and location of nearby surfaces. It has been well established that flagella and other motile cilia perform diverse cellular functions through adhesion. We hypothesized that mammalian primary cilia also interact with the extracellular environment through direct physical contact. METHODS: We identified cilia in rod photoreceptors and cholangiocytes in fixed mouse tissues and examined the structures that these cilia contact in vivo. We then utilized an MDCK cell culture model to characterize the nature of the contacts we observed. RESULTS: In retina and liver tissue, we observed that cilia from nearby cells touch one another. Using MDCK cells, we found compelling evidence that these contacts are stable adhesions that form bridges between two cells, or networks between many cells. We examined the nature and duration of the cilia-cilia contacts and discovered primary cilia movements that facilitate cilia-cilia encounters. Stable adhesions form as the area of contact expands from a single point to a stretch of tightly bound, adjacent cilia membranes. The cilia-cilia contacts persisted for hours and were resistant to several harsh treatments such as proteases and DTT. Unlike many other cell adhesion mechanisms, calcium was not required for the formation or maintenance of cilia adhesion. However, swainsonine, which blocks maturation of N-linked glycoproteins, reduced contact formation. We propose that cellular control of adhesion maintenance is active because cilia adhesion did not prevent cell division; rather, contacts dissolved during mitosis as cilia were resorbed. CONCLUSIONS: The demonstration that mammalian primary cilia formed prolonged, direct, physical contacts supports a novel paradigm: that mammalian primary cilia detect features of the extracellular space, not just as passive antennae, but also through direct physical contact. We present a model for the cycle of glycoprotein-dependent contact formation, maintenance, and termination, and discuss the implications for potential physiological functions of cilia-cilia contacts.

12.
PLoS One ; 6(10): e25404, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22003390

RESUMO

Genetic ablation of Iron Regulatory Protein 2 (Irp2, Ireb2), which post-transcriptionally regulates iron metabolism genes, causes a gait disorder in mice that progresses to hind-limb paralysis. Here we have demonstrated that misregulation of iron metabolism from loss of Irp2 causes lower motor neuronal degeneration with significant spinal cord axonopathy. Mitochondria in the lumbar spinal cord showed significantly decreased Complex I and II activities, and abnormal morphology. Lower motor neurons appeared to be the most adversely affected neurons, and we show that functional iron starvation due to misregulation of iron import and storage proteins, including transferrin receptor 1 and ferritin, may have a causal role in disease. We demonstrated that two therapeutic approaches were beneficial for motor neuron survival. First, we activated a homologous protein, IRP1, by oral Tempol treatment and found that axons were partially spared from degeneration. Secondly, we genetically decreased expression of the iron storage protein, ferritin, to diminish functional iron starvation. These data suggest that functional iron deficiency may constitute a previously unrecognized molecular basis for degeneration of motor neurons in mice.


Assuntos
Deleção de Genes , Deficiências de Ferro , Proteína 2 Reguladora do Ferro/deficiência , Proteína 2 Reguladora do Ferro/genética , Mitocôndrias/metabolismo , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Animais , Apoferritinas/biossíntese , Atrofia/metabolismo , Axônios/efeitos dos fármacos , Axônios/metabolismo , Axônios/patologia , Biomarcadores/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Óxidos N-Cíclicos/farmacologia , Homeostase/efeitos dos fármacos , Homeostase/genética , Ferro/metabolismo , Proteína 1 Reguladora do Ferro/deficiência , Proteína 1 Reguladora do Ferro/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Mitocôndrias/patologia , Neurônios Motores/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Marcadores de Spin , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia
13.
Cancer Cell ; 20(3): 315-27, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21907923

RESUMO

Inactivation of the TCA cycle enzyme, fumarate hydratase (FH), drives a metabolic shift to aerobic glycolysis in FH-deficient kidney tumors and cell lines from patients with hereditary leiomyomatosis renal cell cancer (HLRCC), resulting in decreased levels of AMP-activated kinase (AMPK) and p53 tumor suppressor, and activation of the anabolic factors, acetyl-CoA carboxylase and ribosomal protein S6. Reduced AMPK levels lead to diminished expression of the DMT1 iron transporter, and the resulting cytosolic iron deficiency activates the iron regulatory proteins, IRP1 and IRP2, and increases expression of the hypoxia inducible factor HIF-1α, but not HIF-2α. Silencing of HIF-1α or activation of AMPK diminishes invasive activities, indicating that alterations of HIF-1α and AMPK contribute to the oncogenic growth of FH-deficient cells.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Fumarato Hidratase/deficiência , Deficiências de Ferro , Neoplasias Renais/metabolismo , Leiomiomatose/congênito , Acetilcoenzima A/biossíntese , Acetil-CoA Carboxilase/biossíntese , Acetil-CoA Carboxilase/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Proteínas de Transporte de Cátions/biossíntese , Linhagem Celular Tumoral , Fumarato Hidratase/metabolismo , Glicólise/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Proteína 1 Reguladora do Ferro/biossíntese , Proteína 1 Reguladora do Ferro/metabolismo , Proteína 2 Reguladora do Ferro/biossíntese , Proteína 2 Reguladora do Ferro/metabolismo , Neoplasias Renais/enzimologia , Neoplasias Renais/patologia , Leiomiomatose/metabolismo , Leiomiomatose/patologia , Camundongos , NADP/biossíntese , Síndromes Neoplásicas Hereditárias , Ribose/biossíntese , Proteína S6 Ribossômica/biossíntese , Proteína S6 Ribossômica/metabolismo , Neoplasias Cutâneas , Tenoiltrifluoracetona/farmacologia , Proteína Supressora de Tumor p53/biossíntese , Neoplasias Uterinas
14.
J Clin Invest ; 120(5): 1749-61, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20364084

RESUMO

Glutaredoxin 5 (GLRX5) deficiency has previously been identified as a cause of anemia in a zebrafish model and of sideroblastic anemia in a human patient. Here we report that GLRX5 is essential for iron-sulfur cluster biosynthesis and the maintenance of normal mitochondrial and cytosolic iron homeostasis in human cells. GLRX5, a mitochondrial protein that is highly expressed in erythroid cells, can homodimerize and assemble [2Fe-2S] in vitro. In GLRX5-deficient cells, [Fe-S] cluster biosynthesis was impaired, the iron-responsive element-binding (IRE-binding) activity of iron regulatory protein 1 (IRP1) was activated, and increased IRP2 levels, indicative of relative cytosolic iron depletion, were observed together with mitochondrial iron overload. Rescue of patient fibroblasts with the WT GLRX5 gene by transfection or viral transduction reversed a slow growth phenotype, reversed the mitochondrial iron overload, and increased aconitase activity. Decreased aminolevulinate delta, synthase 2 (ALAS2) levels attributable to IRP-mediated translational repression were observed in erythroid cells in which GLRX5 expression had been downregulated using siRNA along with marked reduction in ferrochelatase levels and increased ferroportin expression. Erythroblasts express both IRP-repressible ALAS2 and non-IRP-repressible ferroportin 1b. The unique combination of IRP targets likely accounts for the tissue-specific phenotype of human GLRX5 deficiency.


Assuntos
Anemia Sideroblástica/metabolismo , Citosol/metabolismo , Eritroblastos/metabolismo , Glutarredoxinas/deficiência , Glutarredoxinas/fisiologia , Heme/metabolismo , Ferro/metabolismo , 5-Aminolevulinato Sintetase/metabolismo , Sequência de Aminoácidos , Células HeLa , Humanos , Proteínas Ferro-Enxofre/metabolismo , Dados de Sequência Molecular , Fenótipo , RNA Interferente Pequeno/metabolismo , Homologia de Sequência de Aminoácidos
15.
Metab Brain Dis ; 24(4): 673-84, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19851851

RESUMO

Maintenance of appropriate iron homeostasis in the brain is important, but the mechanisms involved in brain iron uptake are incompletely understood. Here, we have analyzed where messenger RNAs that encode iron transport proteins are expressed in the brain, using the Allen Brain atlas, and we conclude that several important iron transporters are highly expressed in the choroid plexus. Based on recent estimates of the surface area of the choroid plexus and on MRI imaging studies of manganese uptake in the brain, we propose that the choroid plexus may have a much greater role than has been previously appreciated in brain iron transport.


Assuntos
Encéfalo/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Plexo Corióideo/metabolismo , Homeostase/fisiologia , Ferro/metabolismo , Animais , Atlas como Assunto , Encéfalo/anatomia & histologia , Química Encefálica/fisiologia , Mapeamento Encefálico , Capilares/citologia , Capilares/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Ceruloplasmina/metabolismo , Plexo Corióideo/citologia , Células Epiteliais/metabolismo , Ferritinas/metabolismo , Expressão Gênica/fisiologia , Imageamento por Ressonância Magnética , Camundongos , RNA Mensageiro/metabolismo
16.
J Neurosci ; 29(3): 610-9, 2009 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-19158288

RESUMO

Amyotrophic lateral sclerosis (ALS), characterized by degeneration of spinal motor neurons, consists of sporadic and familial forms. One cause of familial ALS is missense mutations in the superoxide dismutase 1 (SOD1) gene. Iron accumulation occurs in the CNS of both forms of ALS; however, its contribution to the pathogenesis of ALS is not known. We examined the role of iron in a transgenic mouse line overexpressing the human SOD1(G37R) mutant. We show that multiple mechanisms may underlie the iron accumulation in neurons and glia in SOD1(G37R) transgenic mice. These include dysregulation of proteins involved in iron influx and sensing of intracellular iron; iron accumulation in ventral motor neurons secondary to blockage of anterograde axonal transport; and increased mitochondrial iron load in neurons and glia. We also show that treatment of SOD1(G37R) mice with an iron chelator extends life span by 5 weeks, accompanied by increased survival of spinal motor neurons and improved locomotor function. These data suggest that iron chelator therapy might be useful for the treatment of ALS.


Assuntos
Esclerose Lateral Amiotrófica/complicações , Esclerose Lateral Amiotrófica/patologia , Sistema Nervoso Central/metabolismo , Ferro/metabolismo , Fatores Etários , Aldeídos/uso terapêutico , Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/genética , Animais , Peso Corporal/efeitos dos fármacos , Peso Corporal/genética , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Ferrozina , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Homeostase , Hidrazonas/uso terapêutico , Indóis , Quelantes de Ferro/uso terapêutico , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/metabolismo , Mutação , Fosfopiruvato Hidratase/metabolismo , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Neuropatia Ciática/metabolismo , Superóxido Dismutase/genética
17.
J Neurosci ; 28(48): 12736-47, 2008 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-19036966

RESUMO

CNS injury-induced hemorrhage and tissue damage leads to excess iron, which can cause secondary degeneration. The mechanisms that handle this excess iron are not fully understood. We report that spinal cord contusion injury (SCI) in mice induces an "iron homeostatic response" that partially limits iron-catalyzed oxidative damage. We show that ceruloplasmin (Cp), a ferroxidase that oxidizes toxic ferrous iron, is important for this process. SCI in Cp-deficient mice demonstrates that Cp detoxifies and mobilizes iron and reduces secondary tissue degeneration and functional loss. Our results provide new insights into how astrocytes and macrophages handle iron after SCI. Importantly, we show that iron chelator treatment has a delayed effect in improving locomotor recovery between 3 and 6 weeks after SCI. These data reveal important aspects of the molecular control of CNS iron homeostasis after SCI and suggest that iron chelator therapy may improve functional recovery after CNS trauma and hemorrhagic stroke.


Assuntos
Ceruloplasmina/farmacologia , Hemorragia/tratamento farmacológico , Distúrbios do Metabolismo do Ferro/tratamento farmacológico , Ferro/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Ceruloplasmina/uso terapêutico , Modelos Animais de Doenças , Hemorragia/complicações , Hemorragia/fisiopatologia , Ferro/metabolismo , Distúrbios do Metabolismo do Ferro/etiologia , Distúrbios do Metabolismo do Ferro/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/fisiologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/fisiologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/fisiopatologia , Resultado do Tratamento
18.
Glia ; 56(4): 436-48, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18205174

RESUMO

Secondary tissue damage that occurs within days after spinal cord injury contributes significantly to permanent paralysis, sensory loss, and other functional disabilities. The acute inflammatory response is thought to contribute largely to this secondary damage. We show here that 15-deoxy-delta-12,14-prostaglandin J2 (15d-PGJ2), a metabolite of prostaglandin D2 (PGD2) that has anti-inflammatory actions, given daily for the first 2 weeks after spinal cord contusion injury in mice, results in significant improvement of sensory and locomotor function. 15d-PGJ2-treated mice also show diminished signs of microglia/macrophage activation, increased neuronal survival, greater serotonergic innervation, and reduced demyelination in the injured spinal cord. These changes are accompanied by a reduction in chemokine and pro-inflammatory cytokine expression. Our results also indicate that 15d-PGJ2 is likely to reduce inflammation in the injured spinal cord by attenuating multiple signaling pathways: reducing activation of NF-kappa B; enhancing expression of suppressor of cytokine signaling1 and reducing the activation of Janus activated Kinase 2.


Assuntos
Fatores Imunológicos/uso terapêutico , Prostaglandina D2/análogos & derivados , Traumatismos da Medula Espinal/tratamento farmacológico , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Doenças Desmielinizantes/tratamento farmacológico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Microglia/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Prostaglandina D2/uso terapêutico , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Fatores de Tempo
19.
EMBO J ; 26(12): 2823-31, 2007 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-17541408

RESUMO

Ferroportin (Fpn), a ferrous iron Fe(II) transporter responsible for the entry of iron into plasma, is regulated post-translationally through internalization and degradation following binding of the hormone hepcidin. Cellular iron export is impaired in mice and humans with aceruloplasminemia, an iron overload disease due to mutations in the ferroxidase ceruloplasmin (Cp). In the absence of Cp Fpn is rapidly internalized and degraded. Depletion of extracellular Fe(II) by the yeast ferroxidase Fet3p or iron chelators can maintain cell surface Fpn in the absence of Cp. Iron remains bound to Fpn in the absence of multicopper oxidases. Fpn with bound iron is recognized by a ubiquitin ligase, which ubiquitinates Fpn on lysine 253. Mutation of lysine 253 to alanine prevents ubiquitination and maintains Fpn-iron on cell surface in the absence of ferroxidase activity. The requirement for a ferroxidase to maintain iron transport activity represents a new mechanism of regulating cellular iron export, a new function for Cp and an explanation for brain iron overload in patients with aceruloplasminemia.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Ceruloplasmina/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Animais , Endocitose , Humanos , Camundongos , RNA Interferente Pequeno , Ratos , Células Tumorais Cultivadas
20.
Glia ; 55(3): 294-302, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17096403

RESUMO

We have previously shown that intraspinal microinjection of lysophosphatidylcholine (LPC), a potent demyelinating agent, results in a rapid but brief influx of T cells (between 6 and 12 h). This is accompanied by a robust activation of macrophages/microglia that leads to demyelination by 48 h. In the present study, we examined whether this brief influx of T cells contributes to the activation of macrophages/microglia and demyelination by injecting LPC into the dorsal column white matter of athymic Nude mice that lack T cells. We show that there is a significant reduction in macrophage/microglial activation and myelin clearance after LPC injection in Nude mice as compared with wildtype controls. We also show that there is no difference in the recruitment of hematogenous macrophages into the spinal cord after LPC injection in the two mouse strains. Of the T cell cytokines assessed, there was a marked reduction in the mRNA expression of interleukin-2 (IL-2) in Nude mice compared with wildtype animals. Neutralizing IL-2 with function-blocking antibodies in wildtype animals resulted in a significant decrease in the number of phagocytic macrophages/microglia and a reduction in demyelination induced by LPC. While there may be other defects in Nude mice that might contribute to the effects shown here, these data suggest that the brief influx of T cells in this model of chemically-induced demyelination could play a role in macrophage/microglial activation and demyelination. These results may also have implications for remyelination in this and other types of CNS damage.


Assuntos
Quimiotaxia de Leucócito/imunologia , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/imunologia , Ativação Linfocitária/imunologia , Lisofosfatidilcolinas/farmacologia , Macrófagos/imunologia , Linfócitos T/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos/farmacologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/induzido quimicamente , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/fisiopatologia , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Feminino , Interleucina-2/antagonistas & inibidores , Interleucina-2/genética , Interleucina-2/imunologia , Ativação Linfocitária/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/imunologia , Bainha de Mielina/patologia , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Fibras Nervosas Mielinizadas/imunologia , Fibras Nervosas Mielinizadas/patologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/imunologia , Medula Espinal/patologia , Linfócitos T/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA