Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Psychiatry ; 29(4): 1139-1152, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38212377

RESUMO

Iron is an essential element for the development and functionality of the brain, and anomalies in its distribution and concentration in brain tissue have been found to be associated with the most frequent neurodegenerative diseases. When magnetic resonance techniques allowed iron quantification in vivo, it was confirmed that the alteration of brain iron homeostasis is a common feature of many neurodegenerative diseases. However, whether iron is the main actor in the neurodegenerative process, or its alteration is a consequence of the degenerative process is still an open question. Because the different iron-related pathogenic mechanisms are specific for distinctive diseases, identifying the molecular mechanisms common to the various pathologies could represent a way to clarify this complex topic. Indeed, both iron overload and iron deficiency have profound consequences on cellular functioning, and both contribute to neuronal death processes in different manners, such as promoting oxidative damage, a loss of membrane integrity, a loss of proteostasis, and mitochondrial dysfunction. In this review, with the attempt to elucidate the consequences of iron dyshomeostasis for brain health, we summarize the main pathological molecular mechanisms that couple iron and neuronal death.


Assuntos
Encéfalo , Ferro , Doenças Neurodegenerativas , Animais , Humanos , Encéfalo/metabolismo , Morte Celular , Homeostase , Ferro/metabolismo , Sobrecarga de Ferro/metabolismo , Mitocôndrias/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Estresse Oxidativo
2.
Artigo em Inglês | MEDLINE | ID: mdl-38955859

RESUMO

OBJECTIVE: The purpose of this study was to assess how well ChatGPT, an AI-powered chatbot, performed in helping to manage pediatric sialadenitis and identify when sialendoscopy was necessary. METHODS: 49 clinical cases of pediatric sialadenitis were retrospectively reviewed. ChatGPT was given patient data, and it offered differential diagnoses, proposed further tests, and suggested treatments. The decisions made by the treating otolaryngologists were contrasted with the answers provided by ChatGPT. Analysis was done on ChatGPT response consistency and interrater reliability. RESULTS: ChatGPT showed 78.57% accuracy in primary diagnosis, and 17.35% of cases were considered likely. On the other hand, otolaryngologists recommended fewer further examinations than ChatGPT (111 vs. 60, p < 0.001). For additional exams, poor agreement was found between ChatGPT and otolaryngologists. Only 28.57% of cases received a pertinent and essential treatment plan via ChatGPT, indicating that the platform's treatment recommendations were frequently lacking. For treatment ratings, judges' interrater reliability was greatest (Kendall's tau = 0.824, p < 0.001). For the most part, ChatGPT's response constancy was high. CONCLUSIONS: Although ChatGPT has the potential to correctly diagnose pediatric sialadenitis, there are a number of noteworthy limitations with regard to its ability to suggest further testing and treatment regimens. Before widespread clinical use, more research and confirmation are required. To guarantee that chatbots are utilized properly and effectively to supplement human expertise rather than to replace it, a critical viewpoint is required.

3.
Cell Mol Life Sci ; 78(7): 3355-3367, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33439270

RESUMO

Neuroferritinopathy is a rare autosomal dominant inherited movement disorder caused by alteration of the L-ferritin gene that results in the production of a ferritin molecule that is unable to properly manage iron, leading to the presence of free redox-active iron in the cytosol. This form of iron has detrimental effects on cells, particularly severe for neuronal cells, which are highly sensitive to oxidative stress. Although very rare, the disorder is notable for two reasons. First, neuroferritinopathy displays features also found in a larger group of disorders named Neurodegeneration with Brain Iron Accumulation (NBIA), such as iron deposition in the basal ganglia and extrapyramidal symptoms; thus, the elucidation of its pathogenic mechanism may contribute to clarifying the incompletely understood aspects of NBIA. Second, neuroferritinopathy shows the characteristic signs of an accelerated process of aging; thus, it can be considered an interesting model to study the progress of aging. Here, we will review the clinical and neurological features of neuroferritinopathy and summarize biochemical studies and data from cellular and animal models to propose a pathogenic mechanism of the disorder.


Assuntos
Apoferritinas/metabolismo , Distúrbios do Metabolismo do Ferro/patologia , Ferro/metabolismo , Distrofias Neuroaxonais/patologia , Animais , Humanos , Distúrbios do Metabolismo do Ferro/metabolismo , Distrofias Neuroaxonais/metabolismo
4.
Int J Mol Sci ; 21(10)2020 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-32456086

RESUMO

Pantothenate Kinase-associated Neurodegeneration (PKAN) belongs to a wide spectrum of diseases characterized by brain iron accumulation and extrapyramidal motor signs. PKAN is caused by mutations in PANK2, encoding the mitochondrial pantothenate kinase 2, which is the first enzyme of the biosynthesis of Coenzyme A. We established and characterized glutamatergic neurons starting from previously developed PKAN Induced Pluripotent Stem Cells (iPSCs). Results obtained by inductively coupled plasma mass spectrometry indicated a higher amount of total cellular iron in PKAN glutamatergic neurons with respect to controls. PKAN glutamatergic neurons, analyzed by electron microscopy, exhibited electron dense aggregates in mitochondria that were identified as granules containing calcium phosphate. Calcium homeostasis resulted compromised in neurons, as verified by monitoring the activity of calcium-dependent enzyme calpain1, calcium imaging and voltage dependent calcium currents. Notably, the presence of calcification in the internal globus pallidus was confirmed in seven out of 15 genetically defined PKAN patients for whom brain CT scan was available. Moreover, we observed a higher prevalence of brain calcification in females. Our data prove that high amount of iron coexists with an impairment of cytosolic calcium in PKAN glutamatergic neurons, indicating both, iron and calcium dys-homeostasis, as actors in pathogenesis of the disease.


Assuntos
Cálcio/metabolismo , Ferro/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Adolescente , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Cálcio/efeitos adversos , Calpaína/metabolismo , Criança , Pré-Escolar , Estudos de Coortes , Citoplasma/fisiologia , Feminino , Homeostase , Humanos , Células-Tronco Pluripotentes Induzidas , Lactente , Ferro/efeitos adversos , Imageamento por Ressonância Magnética , Masculino , Espectrometria de Massas , Microscopia Eletrônica , Mitocôndrias/enzimologia , Mitocôndrias/ultraestrutura , Neurônios/fisiologia , Neurônios/ultraestrutura , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Fosfotransferases (Aceptor do Grupo Álcool) , Tomografia Computadorizada por Raios X , Adulto Jovem
5.
Adv Exp Med Biol ; 1173: 153-177, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31456210

RESUMO

Neurodegeneration with brain iron accumulation (NBIA) is a group of seriously devastating and life-threatening rare monogenic diseases characterized by focal iron accumulation in the brain. The main symptoms of NBIA comprise progressive movement disorder, often including painful dystonia, parkinsonism, mental disability, and early death. Currently, a single established therapy is not available to reverse the progression of these debilitating disorders. The complexity of NBIA emerged from the identification of various causative genes, and up to 15 genes have been identified to date. Although the NBIA genes are involved in different cellular biochemical pathways, they show the common characteristic of generating severe iron accumulation in the basal ganglia of the patients' brains. Thus, the molecular events that lead to brain iron overload and their important roles in the pathophysiology of the diseases are not easy to identify and are poorly understood. This review summarizes the current knowledge on NBIA disorders, with a particular focus on the data describing the role of iron in the pathogenic mechanisms.


Assuntos
Encéfalo/fisiopatologia , Ferro/metabolismo , Gânglios da Base , Encéfalo/metabolismo , Humanos , Sobrecarga de Ferro , Doenças Neurodegenerativas
6.
Neurobiol Dis ; 81: 119-33, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25447222

RESUMO

Neuroferritinopathy is a rare genetic disease with a dominant autosomal transmission caused by mutations of the ferritin light chain gene (FTL). It belongs to Neurodegeneration with Brain Iron Accumulation, a group of disorders where iron dysregulation is tightly associated with neurodegeneration. We studied the 498-499InsTC mutation which causes the substitution of the last 9 amino acids and an elongation of extra 16 amino acids at the C-terminus of L-ferritin peptide. An analysis with cyclic voltammetry on the purified protein showed that this structural modification severely reduces the ability of the protein to store iron. In order to analyze the impact of the mutation in vivo, we generated mouse models for the some pathogenic human FTL gene in FVB and C57BL/6J strains. Transgenic mice in the FVB background showed high accumulation of the mutated ferritin in brain where it correlated with increased iron deposition with age, as scored by magnetic resonance imaging. Notably, the accumulation of iron-ferritin bodies was accompanied by signs of oxidative damage. In the C57BL/6 background, both the expression of the mutant ferritin and the iron levels were lower than in the FVB strain. Nevertheless, also these mice showed oxidative alterations in the brain. Furthermore, post-natal hippocampal neurons obtained from these mice experienced a marked increased cell death in response to chronic iron overload and/or acute oxidative stress, in comparison to wild-type neurons. Ultrastructural analyses revealed an accumulation of lipofuscin granules associated with iron deposits, particularly enriched in the cerebellum and striatum of our transgenic mice. Finally, experimental subjects were tested throughout development and aging at 2-, 8- and 18-months for behavioral phenotype. Rotarod test revealed a progressive impaired motor coordination building up with age, FTL mutant old mice showing a shorter latency to fall from the apparatus, according to higher accumulation of iron aggregates in the striatum. Our data show that our 498-499InsTC mouse models recapitulate early pathological and clinical traits of the human neuroferritinopathy, thus providing a valuable model for the study of the disease. Finally, we propose a mechanistic model of lipofuscine formation that can account for the etiopathogenesis of human neuroferritinopathy.


Assuntos
Apoferritinas/genética , Encéfalo/patologia , Distúrbios do Metabolismo do Ferro/etiologia , Distrofias Neuroaxonais , Doenças Neurodegenerativas/etiologia , Transtornos Psicomotores/etiologia , Fatores Etários , Animais , Apoferritinas/metabolismo , Encéfalo/metabolismo , Morte Celular/genética , Células Cultivadas , Dano ao DNA/genética , Modelos Animais de Doenças , Progressão da Doença , Feminino , Hipocampo/citologia , Humanos , Distúrbios do Metabolismo do Ferro/complicações , Distúrbios do Metabolismo do Ferro/genética , Distúrbios do Metabolismo do Ferro/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Moleculares , Distrofias Neuroaxonais/complicações , Distrofias Neuroaxonais/genética , Distrofias Neuroaxonais/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo
7.
Hum Mol Genet ; 21(18): 4049-59, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22692681

RESUMO

Pantothenate kinase-associated neurodegeneration (PKAN) is a neurodegenerative disease belonging to the group of neurodegeneration with brain iron accumulation disorders. It is characterized by progressive impairments in movement, speech and cognition. The disease is inherited in a recessive manner due to mutations in the Pantothenate Kinase-2 (PANK2) gene that encodes a mitochondrial protein involved in Coenzyme A synthesis. To investigate the link between a PANK2 gene defect and iron accumulation, we analyzed primary skin fibroblasts from three PKAN patients and three unaffected subjects. The oxidative status of the cells and their ability to respond to iron were analyzed in both basal and iron supplementation conditions. In basal conditions, PKAN fibroblasts show an increase in carbonylated proteins and altered expression of antioxidant enzymes with respect to the controls. After iron supplementation, the PKAN fibroblasts had a defective response to the additional iron. Under these conditions, ferritins were up-regulated and Transferrin Receptor 1 (TfR1) was down-regulated to a minor extent in patients compared with the controls. Analysis of iron regulatory proteins (IRPs) reveals that, with respect to the controls, PKAN fibroblasts have a reduced amount of membrane-associated mRNA-bound IRP1, which responds imperfectly to iron. This accounts for the defective expression of ferritin and TfR1 in patients' cells. The inaccurate quantity of these proteins produced a higher bioactive labile iron pool and consequently increased iron-dependent reactive oxygen species formation. Our results suggest that Pank2 deficiency promotes an increased oxidative status that is further enhanced by the addition of iron, potentially causing damage in cells.


Assuntos
Fibroblastos/metabolismo , Ferro/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Pele/patologia , Estudos de Casos e Controles , Catalase/metabolismo , Células Cultivadas , Ferritinas/metabolismo , Fibroblastos/enzimologia , Humanos , Proteínas Reguladoras de Ferro/metabolismo , Mutação de Sentido Incorreto , Oxirredução , Estresse Oxidativo , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Ligação Proteica , Carbonilação Proteica , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
8.
Hum Mol Genet ; 21(24): 5294-305, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22983956

RESUMO

Neurodegeneration with brain iron accumulation (NBIA) comprises a group of neurodegenerative disorders characterized by high brain content of iron and presence of axonal spheroids. Mutations in the PANK2 gene, which encodes pantothenate kinase 2, underlie an autosomal recessive inborn error of coenzyme A metabolism, called pantothenate kinase-associated neurodegeneration (PKAN). PKAN is characterized by dystonia, dysarthria, rigidity and pigmentary retinal degeneration. The pathogenesis of this disorder is poorly understood and, although PANK2 is a mitochondrial protein, perturbations in mitochondrial bioenergetics have not been reported. A knock-out (KO) mouse model of PKAN exhibits retinal degeneration and azoospermia, but lacks any neurological phenotype. The absence of a clinical phenotype has partially been explained by the different cellular localization of the human and murine PANK2 proteins. Here we demonstrate that the mouse Pank2 protein localizes to mitochondria, similar to its human orthologue. Moreover, we show that Pank2-defective neurons derived from KO mice have an altered mitochondrial membrane potential, a defect further corroborated by the observations of swollen mitochondria at the ultra-structural level and by the presence of defective respiration.


Assuntos
Sistema Nervoso Central/enzimologia , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Doenças Neurodegenerativas/enzimologia , Estresse Oxidativo/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Sistema Nervoso Central/metabolismo , Humanos , Potenciais da Membrana/genética , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Doenças Neurodegenerativas/genética , Estresse Oxidativo/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética
9.
Cell Death Dis ; 15(5): 361, 2024 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-38796462

RESUMO

Disease models of neurodegeneration with brain iron accumulation (NBIA) offer the possibility to explore the relationship between iron dyshomeostasis and neurodegeneration. We analyzed hiPS-derived astrocytes from PANK2-associated neurodegeneration (PKAN), an NBIA disease characterized by progressive neurodegeneration and high iron accumulation in the globus pallidus. Previous data indicated that PKAN astrocytes exhibit alterations in iron metabolism, general impairment of constitutive endosomal trafficking, mitochondrial dysfunction and acquired neurotoxic features. Here, we performed a more in-depth analysis of the interactions between endocytic vesicles and mitochondria via superresolution microscopy experiments. A significantly lower number of transferrin-enriched vesicles were in contact with mitochondria in PKAN cells than in control cells, confirming the impaired intracellular fate of cargo endosomes. The investigation of cytosolic and mitochondrial iron parameters indicated that mitochondrial iron availability was substantially lower in PKAN cells compared to that in the controls. In addition, PKAN astrocytes exhibited defects in tubulin acetylation/phosphorylation, which might be responsible for unregulated vesicular dynamics and inappropriate iron delivery to mitochondria. Thus, the impairment of iron incorporation into these organelles seems to be the cause of cell iron delocalization, resulting in cytosolic iron overload and mitochondrial iron deficiency, triggering mitochondrial dysfunction. Overall, the data elucidate the mechanism of iron accumulation in CoA deficiency, highlighting the importance of mitochondrial iron deficiency in the pathogenesis of disease.


Assuntos
Astrócitos , Citosol , Sobrecarga de Ferro , Ferro , Mitocôndrias , Astrócitos/metabolismo , Astrócitos/patologia , Humanos , Mitocôndrias/metabolismo , Citosol/metabolismo , Ferro/metabolismo , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/patologia , Tubulina (Proteína)/metabolismo , Fosforilação , Deficiências de Ferro , Acetilação
10.
Front Clin Diabetes Healthc ; 4: 1326239, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38264059

RESUMO

Introduction: Diabetes is a major cause of disease burden with considerable public health significance. While the pancreas plays a significant role in glucose homeostasis, the association between pancreatitis and new onset diabetes is not well understood. The purpose of this study was to examine that association using large real-world data. Materials and methods: Utilizing the IBM® MarketScan® commercial claims database from 2016 to 2019, pancreatitis and diabetes regardless of diagnostic category, were identified using International Classification of Diseases, Tenth Revision [ICD-10] codes. We then performed descriptive analyses characterizing non-pancreatitis (NP), acute pancreatitis (AP), and chronic pancreatitis (CP) cohort subjects. Stratified Cox proportional hazards regression models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CI) of diabetes across the three clinical categories. Results: In total, 310,962 individuals were included in the analysis. During 503,274 person-years of follow-up, we identified 15,951 incident diabetes cases. While men and women had higher incidence rates of CP and AP-related diabetes, the rates were significantly greater in men and highest among individuals with CP (91.6 per 1000 persons-years (PY)) followed by AP (75.9 per 1000-PY) as compared to those with NP (27.8 per 1000-PY). After adjustment for diabetes risk factors, relative to the NP group, the HR for future diabetes was 2.59 (95% CI: 2.45-2.74) (P<0.001) for the CP group, and 2.39 (95% CI: 2.30-2.48) (P<0.001) for the AP group. Conclusion: Pancreatitis was associated with a high risk of diabetes independent of demographic, lifestyle, and comorbid conditions.

11.
Pharmaceutics ; 15(1)2023 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-36678831

RESUMO

The novel brain-penetrant peroxisome proliferator-activated receptor gamma agonist leriglitazone, previously validated for other rare neurodegenerative diseases, is a small molecule that acts as a regulator of mitochondrial function and exerts neuroprotective, anti-oxidative and anti-inflammatory effects. Herein, we tested whether leriglitazone can be effective in ameliorating the mitochondrial defects that characterize an hiPS-derived model of Pantothenate kinase-2 associated Neurodegeneration (PKAN). PKAN is caused by a genetic alteration in the mitochondrial enzyme pantothenate kinase-2, whose function is to catalyze the first reaction of the CoA biosynthetic pathway, and for which no effective cure is available. The PKAN hiPS-derived astrocytes are characterized by mitochondrial dysfunction, cytosolic iron deposition, oxidative stress and neurotoxicity. We monitored the effect of leriglitazone in comparison with CoA on hiPS-derived astrocytes from three healthy subjects and three PKAN patients. The treatment with leriglitazone did not affect the differentiation of the neuronal precursor cells into astrocytes, and it improved the viability of PKAN cells and their respiratory activity, while diminishing the iron accumulation similarly or even better than CoA. The data suggest that leriglitazone is well tolerated in this cellular model and could be considered a beneficial therapeutic approach in the treatment of PKAN.

12.
J Pers Med ; 13(10)2023 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-37888040

RESUMO

BACKGROUND: While intraoperative neuromonitoring (IONM) helps the early identification of recurrent laryngeal nerve (RLN) damage, IONM's role in RLN damage prevention is not defined, given the lack of large studies on the subject. METHODS: In a PRISMA-compliant framework, all original thyroid surgery prospective studies providing early postoperative endoscopic data for all patients were pooled in a random-effects meta-analysis. We compared the temporary (and definitive where available) RLN damage rates according to IONM use and IONM type (intermittent, I-IONM, or continuous, C-IONM). RESULTS: We identified 2358 temporary and 257 definitive RLN injuries in, respectively, 73,325 and 66,476 nerves at risk. The pooled temporary and definitive RLN injury rates were, respectively, 3.15% and 0.422% considering all procedures, 3.29% and 0.409% in cases using IONM, and 3.16% and 0.463 in cases not using IONM. I-IONM and C-IONM, respectively, showed a pooled temporary RLN injury rate of 2.48% and 2.913% and a pooled definitive injury rate of 0.395% and 0.4%. All pooled rates had largely overlapping 95% confidence intervals. CONCLUSIONS: Our data suggest that IONM does not affect the temporary or definitive RLN injury rate following thyroidectomy, though its use can be advised in selected cases and for bilateral palsy prevention.

13.
Front Cell Neurosci ; 16: 878103, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35783094

RESUMO

PKAN disease is caused by mutations in the PANK2 gene, encoding the mitochondrial enzyme pantothenate kinase 2, catalyzing the first and key reaction in Coenzyme A (CoA) biosynthetic process. This disorder is characterized by progressive neurodegeneration and excessive iron deposition in the brain. The pathogenic mechanisms of PKAN are still unclear, and the available therapies are only symptomatic. Although iron accumulation is a hallmark of PKAN, its relationship with CoA dysfunction is not clear. We have previously developed hiPS-derived astrocytes from PKAN patients showing iron overload, thus recapitulating the human phenotype. In this work, we demonstrated that PKAN astrocytes presented an increase in transferrin uptake, a key route for cellular iron intake via transferrin receptor-mediated endocytosis of transferrin-bound iron. Investigation of constitutive exo-endocytosis and vesicular dynamics, exploiting the activity-enriching biosensor SynaptoZip, led to the finding of a general impairment in the constitutive endosomal trafficking in PKAN astrocytes. CoA and 4-phenylbutyric acid treatments were found to be effective in partially rescuing the aberrant vesicular behavior and iron intake. Our results demonstrate that the impairment of CoA biosynthesis could interfere with pivotal intracellular mechanisms involved in membrane fusions and vesicular trafficking, leading to an aberrant transferrin receptor-mediated iron uptake.

14.
Cell Death Dis ; 13(2): 185, 2022 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-35217637

RESUMO

Neurodegeneration associated with defective pantothenate kinase-2 (PKAN) is an early-onset monogenic autosomal-recessive disorder. The hallmark of the disease is the massive accumulation of iron in the globus pallidus brain region of patients. PKAN is caused by mutations in the PANK2 gene encoding the mitochondrial enzyme pantothenate kinase-2, whose function is to catalyze the first reaction of the CoA biosynthetic pathway. To date, the way in which this alteration leads to brain iron accumulation has not been elucidated. Starting from previously obtained hiPS clones, we set up a differentiation protocol able to generate inhibitory neurons. We obtained striatal-like medium spiny neurons composed of approximately 70-80% GABAergic neurons and 10-20% glial cells. Within this mixed population, we detected iron deposition in both PKAN cell types, however, the viability of PKAN GABAergic neurons was strongly affected. CoA treatment was able to reduce cell death and, notably, iron overload. Further differentiation of hiPS clones in a pure population of astrocytes showed particularly evident iron accumulation, with approximately 50% of cells positive for Perls staining. The analysis of these PKAN astrocytes indicated alterations in iron metabolism, mitochondrial morphology, respiratory activity, and oxidative status. Moreover, PKAN astrocytes showed signs of ferroptosis and were prone to developing a stellate phenotype, thus gaining neurotoxic features. This characteristic was confirmed in iPS-derived astrocyte and glutamatergic neuron cocultures, in which PKAN glutamatergic neurons were less viable in the presence of PKAN astrocytes. This newly generated astrocyte model is the first in vitro disease model recapitulating the human phenotype and can be exploited to deeply clarify the pathogenetic mechanisms underlying the disease.


Assuntos
Astrócitos , Neurodegeneração Associada a Pantotenato-Quinase , Astrócitos/metabolismo , Coenzima A/genética , Coenzima A/metabolismo , Humanos , Ferro/metabolismo , Neurônios/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/genética , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Fenótipo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo
15.
J Biol Chem ; 285(16): 11948-57, 2010 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-20159981

RESUMO

Nucleotide insertions that modify the C terminus of ferritin light chain (FTL) cause neurodegenerative movement disorders named neuroferritinopathies, which are inherited with dominant transmission. The disorders are characterized by abnormal brain iron accumulation. Here we describe the biochemical and crystallographic characterization of pathogenic FTL mutant p.Phe167SerfsX26 showing that it is a functional ferritin with an altered conformation of the C terminus. Moreover we analyze functional and stability properties of ferritin heteropolymers made of 20-23 H-chains and 1-4 L-chains with representative pathogenic mutations or the last 10-28 residues truncated. All the heteropolymers containing the pathogenic or truncated mutants had a strongly reduced capacity to incorporate iron, both when expressed in Escherichia coli, and in vitro when iron was supplied as Fe(III) in the presence of ascorbate. The mutations also reduced the physical stability of the heteropolymers. The data indicate that even a few mutated L-chains are sufficient to alter the permeability of 1-2 of the 6 hydrophobic channels and modify ferritin capacity to incorporate iron. The dominant-negative action of the mutations explains the dominant transmission of the disorder. The data support the hypothesis that hereditary ferritinopathies are due to alterations of ferritin functionality and provide new input on the mechanism of the function of isoferritins.


Assuntos
Apoferritinas/genética , Apoferritinas/metabolismo , Ferro/metabolismo , Mutação , Degeneração Neural/genética , Degeneração Neural/metabolismo , Sequência de Aminoácidos , Apoferritinas/química , Cristalografia por Raios X , Genes Dominantes , Humanos , Interações Hidrofóbicas e Hidrofílicas , Técnicas In Vitro , Modelos Moleculares , Dados de Sequência Molecular , Degeneração Neural/etiologia , Conformação Proteica , Estabilidade Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Eletricidade Estática
16.
Hum Mol Genet ; 18(1): 1-11, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18815198

RESUMO

Mitochondrial ferritin (FtMt) is a nuclear-encoded iron-sequestering protein that specifically localizes in mitochondria. In mice it is highly expressed in cells characterized by high-energy consumption, while is undetectable in iron storage tissues like liver and spleen. FtMt expression in mammalian cells was shown to cause a shift of iron from cytosol to mitochondria, and in yeast it rescued the defects associated with frataxin deficiency. To study the role of FtMt in oxidative damage, we analyzed the effect of its expression in HeLa cells after incubation with H(2)O(2) and Antimycin A, and after a long-term growth in glucose-free media that enhances mitochondrial respiratory activity. FtMt reduced the level of reactive oxygen species (ROS), increased the level of adenosine 5'triphosphate and the activity of mitochondrial Fe-S enzymes, and had a positive effect on cell viability. Furthermore, FtMt expression reduces the size of cytosolic and mitochondrial labile iron pools. In cells grown in glucose-free media, FtMt level was reduced owing to faster degradation rate, however it still protected the activity of mitochondrial Fe-S enzymes without affecting the cytosolic iron status. In addition, FtMt expression in fibroblasts from Friedreich ataxia (FRDA) patients prevented the formation of ROS and partially rescued the impaired activity of mitochondrial Fe-S enzymes, caused by frataxin deficiency. These results indicate that the primary function of FtMt involves the control of ROS formation through the regulation of mitochondrial iron availability. They are consistent with the expression pattern of FtMt observed in mouse tissues, suggesting a FtMt protective role in cells characterized by defective iron homeostasis and respiration, such as in FRDA.


Assuntos
Ferritinas/metabolismo , Ataxia de Friedreich/metabolismo , Regulação da Expressão Gênica , Ferro/metabolismo , Proteínas Mitocondriais/metabolismo , Animais , Células Cultivadas , Ferritinas/genética , Fibroblastos/metabolismo , Ataxia de Friedreich/genética , Expressão Gênica , Células HeLa , Humanos , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Oxirredução , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
17.
Haematologica ; 96(10): 1424-32, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21712541

RESUMO

BACKGROUND: Mitochondrial ferritin is a nuclear encoded iron-storage protein localized in mitochondria. It has anti-oxidant properties related to its ferroxidase activity, and it is able to sequester iron avidly into the organelle. The protein has a tissue-specific pattern of expression and is also highly expressed in sideroblasts of patients affected by hereditary sideroblastic anemia and by refractory anemia with ringed sideroblasts. The present study examined whether mitochondrial ferritin has a role in the pathogenesis of these diseases. DESIGN AND METHODS: We analyzed the effect of mitochondrial ferritin over-expression on the JAK2/STAT5 pathway, on iron metabolism and on heme synthesis in erythroleukemic cell lines. Furthermore its effect on apoptosis was evaluated on human erythroid progenitors. RESULTS: Data revealed that a high level of mitochondrial ferritin reduced reactive oxygen species and Stat5 phosphorylation while promoting mitochondrial iron loading and cytosolic iron starvation. The decline of Stat5 phosphorylation induced a decrease of the level of anti-apoptotic Bcl-xL transcript compared to that in control cells; however, transferrin receptor 1 transcript increased due to the activation of the iron responsive element/iron regulatory protein machinery. Also, high expression of mitochondrial ferritin increased apoptosis, limited heme synthesis and promoted the formation of Perls-positive granules, identified by electron microscopy as iron granules in mitochondria. CONCLUSIONS: Our results provide evidence suggesting that Stat5-dependent transcriptional regulation is displaced by strong cytosolic iron starvation status induced by mitochondrial ferritin. The protein interferes with JAK2/STAT5 pathways and with the mechanism of mitochondrial iron accumulation.


Assuntos
Ferritinas/genética , Ferro/metabolismo , Janus Quinase 2/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Animais , Apoptose , Linhagem Celular Tumoral , Ferritinas/biossíntese , Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Heme/metabolismo , Humanos , Células K562 , Camundongos , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT5/biossíntese , Transcrição Gênica
18.
Cells ; 10(8)2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34440737

RESUMO

In 2001, a new type of human ferritin was identified by searching for homologous sequences to H-ferritin in the human genome. After the demonstration that this ferritin is located specifically in the mitochondrion, it was called mitochondrial ferritin. Studies on the properties of this new type of ferritin have been limited by its very high homology with the cytosolic H-ferritin, which is expressed at higher levels in cells. This great similarity made it difficult to obtain specific antibodies against the mitochondrial ferritin devoid of cross-reactivity with cytosolic ferritin. Thus, the knowledge of the physiological role of mitochondrial ferritin is still incomplete despite 20 years of research. In this review, we summarize the literature on mitochondrial ferritin expression regulation and its physical and biochemical properties, with particular attention paid to the differences with cytosolic ferritin and its role in physiological condition. Until now, there has been no evidence that the alteration of the mitochondrial ferritin gene is causative of any disorder; however, the identified association of the mitochondrial ferritin with some disorders is discussed.


Assuntos
Ferritinas/metabolismo , Mitocôndrias/metabolismo , Anemia Sideroblástica/metabolismo , Anemia Sideroblástica/patologia , Citosol/metabolismo , Ferritinas/química , Ataxia de Friedreich/metabolismo , Ataxia de Friedreich/patologia , Humanos , Ferro/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Conformação Proteica , Síndrome das Pernas Inquietas/metabolismo , Síndrome das Pernas Inquietas/patologia
19.
Clin Case Rep ; 9(10): e04947, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34667612

RESUMO

Erythema multiforme major, an immune-mediated skin reaction to infections or medications with oral involvement, should be taken into account as a potential side effect of several vaccines, including SARS-CoV-2. Correct patient history collection allows prompt recognition and subsequent successful medical management with oral corticosteroids.

20.
Neurobiol Dis ; 37(1): 77-85, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19781644

RESUMO

Neuroferritinopathies are dominantly inherited movement disorders associated with nucleotide insertions in the L-ferritin gene that modify the protein's C-terminus. The insertions alter physical and functional properties of the ferritins, causing an imbalance in brain iron homeostasis. We describe the effects produced by the over-expression in HeLa and neuroblastoma SH-SY5Y cells of two pathogenic L-ferritin variants, 460InsA and 498InsTC. Both peptides co-assembled with endogenous ferritins, producing molecules with reduced iron incorporation capacity, acting in a dominant negative manner. The cells showed an increase in cell death and a decrease in proteasomal activity. The formation of iron-ferritin aggregates became evident after 10 days of variant expression and was not associated with increased cell death. The addition of iron chelators or antioxidants restored proteasomal activity and reduced aggregate formation. The data indicate that cellular iron imbalance and oxidative damage are primary causes of cell death, while aggregate formation is a secondary effect.


Assuntos
Apoferritinas/genética , Apoferritinas/metabolismo , Transtornos dos Movimentos/genética , Estresse Oxidativo/fisiologia , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/fisiologia , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Morte Celular/fisiologia , Linhagem Celular Tumoral , Variação Genética , Células HeLa , Humanos , Ferro/metabolismo , Quelantes de Ferro/farmacologia , Mutação , Neuroblastoma/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/fisiologia , Multimerização Proteica , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA