Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Free Radic Biol Med ; 221: 273-282, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-38740102

RESUMO

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


Assuntos
Fibroblastos , Ferro , Lipofuscina , Lisossomos , Estresse Oxidativo , Espécies Reativas de Oxigênio , Serina , Humanos , Lisossomos/metabolismo , Lisossomos/efeitos dos fármacos , Lipofuscina/metabolismo , Ferro/metabolismo , Fibroblastos/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Serina/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Distrofias Neuroaxonais/metabolismo , Distrofias Neuroaxonais/patologia , Distrofias Neuroaxonais/tratamento farmacológico , Distrofias Neuroaxonais/genética , 2,2'-Dipiridil/farmacologia , 2,2'-Dipiridil/análogos & derivados , Quelantes de Ferro/farmacologia
2.
Ann Clin Transl Neurol ; 11(5): 1359-1364, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38561955

RESUMO

Neuroferritinopathy is a disorder of neurodegeneration with brain iron accumulation that has no proven disease-modifying treatments. Clinical trials require biomarkers of iron deposition. We examined brain iron accumulation in one presymptomatic FTL mutation carrier, two individuals with neuroferritinopathy and one healthy control using ultra-high-field 7T MRI. There was increased magnetic susceptibility, suggestive of iron deposition, in superficial and deep gray matter in both presymptomatic and symptomatic neuroferritinopathy. Cavitation of the putamen and globus pallidus increased with disease stage and at follow up. The widespread brain iron deposition in presymptomatic and early disease provides an opportunity for monitoring disease-modifying intervention.


Assuntos
Distúrbios do Metabolismo do Ferro , Ferro , Imageamento por Ressonância Magnética , Distrofias Neuroaxonais , Humanos , Distrofias Neuroaxonais/diagnóstico por imagem , Distrofias Neuroaxonais/genética , Distrofias Neuroaxonais/metabolismo , Distrofias Neuroaxonais/patologia , Distúrbios do Metabolismo do Ferro/diagnóstico por imagem , Distúrbios do Metabolismo do Ferro/metabolismo , Distúrbios do Metabolismo do Ferro/genética , Ferro/metabolismo , Adulto , Masculino , Feminino , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Pessoa de Meia-Idade , Apoferritinas/metabolismo , Apoferritinas/genética
3.
Elife ; 122023 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-36645408

RESUMO

Infantile neuroaxonal dystrophy (INAD) is caused by recessive variants in PLA2G6 and is a lethal pediatric neurodegenerative disorder. Loss of the Drosophila homolog of PLA2G6, leads to ceramide accumulation, lysosome expansion, and mitochondrial defects. Here, we report that retromer function, ceramide metabolism, the endolysosomal pathway, and mitochondrial morphology are affected in INAD patient-derived neurons. We show that in INAD mouse models, the same features are affected in Purkinje cells, arguing that the neuropathological mechanisms are evolutionary conserved and that these features can be used as biomarkers. We tested 20 drugs that target these pathways and found that Ambroxol, Desipramine, Azoramide, and Genistein alleviate neurodegenerative phenotypes in INAD flies and INAD patient-derived neural progenitor cells. We also develop an AAV-based gene therapy approach that delays neurodegeneration and prolongs lifespan in an INAD mouse model.


Assuntos
Proteínas de Drosophila , Distrofias Neuroaxonais , Transtornos Parkinsonianos , Camundongos , Animais , Neurônios/metabolismo , Transtornos Parkinsonianos/metabolismo , Drosophila/metabolismo , Ceramidas/metabolismo , Distrofias Neuroaxonais/genética , Distrofias Neuroaxonais/metabolismo , Distrofias Neuroaxonais/patologia , Fosfolipases A2 do Grupo VI/metabolismo , Proteínas do Olho/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
4.
Neurobiol Dis ; 165: 105649, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35122944

RESUMO

BACKGROUND: PLA2G6-Associated Neurodegeneration (PLAN) is a rare neurodegenerative disease with autosomal recessive inheritance, which belongs to the NBIA (Neurodegeneration with Brain Iron Accumulation) group. Although the pathogenesis of the disease remains largely unclear, lipid peroxidation seems to play a central role in the pathogenesis. Currently, there is no cure for the disease. OBJECTIVE: In this work, we examined the presence of lipid peroxidation, iron accumulation and mitochondrial dysfunction in two cellular models of PLAN, patients-derived fibroblasts and induced neurons, and assessed the effects of α-tocopherol (vitamin E) in correcting the pathophysiological alterations in PLAN cell cultures. METHODS: Pathophysiological alterations were examined in fibroblasts and induced neurons generated by direct reprograming. Iron and lipofuscin accumulation were assessed using light and electron microscopy, as well as biochemical analysis techniques. Reactive Oxygen species production, lipid peroxidation and mitochondrial dysfunction were measured using specific fluorescent probes analysed by fluorescence microscopy and flow cytometry. RESULTS: PLAN fibroblasts and induced neurons clearly showed increased lipid peroxidation, iron accumulation and altered mitochondrial membrane potential. All these pathological features were reverted with vitamin E treatment. CONCLUSIONS: PLAN fibroblasts and induced neurons reproduce the main pathological alterations of the disease and provide useful tools for disease modelling. The main pathological alterations were corrected by Vitamin E supplementation in both models, suggesting that blocking lipid peroxidation progression is a critical therapeutic target.


Assuntos
Distrofias Neuroaxonais , Doenças Neurodegenerativas , Fosfolipases A2 do Grupo VI/metabolismo , Humanos , Ferro/metabolismo , Peroxidação de Lipídeos , Mitocôndrias/metabolismo , Distrofias Neuroaxonais/metabolismo , Distrofias Neuroaxonais/patologia , Doenças Neurodegenerativas/metabolismo , Vitamina E/metabolismo , Vitamina E/farmacologia
5.
Oxid Med Cell Longev ; 2022: 8096009, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35116093

RESUMO

The loss of transient receptor potential mucolipin 1 (TRPML1), an endosomal and lysosomal Ca2+-releasing channel, has been implicated in neurodegenerative disorders. Mounting evidence have shown that TRPML1 could clear intraneuronal amyloid-ß (Aß), which triggers a hypothesis that TRPML1 activation may be beneficial for axonal transport in Alzheimer's disease (AD). In this work, the functional roles of TRPML1 were studied in the APP/PS1 transgenic mice and Aß1-42-stimulated hippocampal neurons HT22. We found that lentivirus-mediated overexpression of TRPML1 was shown to promote an accumulation of autolysosomes and increase brain-derived neurotrophic factor (BDNF) transportation to the nucleus, suggesting an axon-protective function. More importantly, we found that TRPML1 also increased p62 that interacted with dynein. Lentivirus-mediated knockdown of p62 or inhibition of dynein by ciliobrevin D stimulation was found to reduce autolysosome formation and nuclear accumulation of BDNF in HT22 cells with Aß1-42 stimulation. Inhibition of p62 by XRK3F2 stimulation was observed to promote the death of hippocampal neurons of the APP/PS1 transgenic mice. TRPML1 recruited dynein by interacting with p62 to promote the autophagosome-lysosome fusion to mediate BDNF nuclear translocation to impede axon dystrophy in mice with Alzheimer-like phenotypes. In summary, these results demonstrate the presence of a TRPML1/p62/dynein regulatory network in AD, and activation of TRPML1 is required for axon protection to prevent neuroaxonal dystrophy.


Assuntos
Doença de Alzheimer/patologia , Autofagossomos/metabolismo , Lisossomos/metabolismo , Proteína Sequestossoma-1/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/farmacologia , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Dineínas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Distrofias Neuroaxonais/metabolismo , Distrofias Neuroaxonais/patologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fragmentos de Peptídeos/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteína Sequestossoma-1/antagonistas & inibidores , Proteína Sequestossoma-1/genética , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Canais de Potencial de Receptor Transitório/genética
6.
Brain Res ; 1748: 147066, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32818532

RESUMO

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.


Assuntos
Gânglios da Base/metabolismo , Ferro/metabolismo , Destreza Motora/fisiologia , Distrofias Neuroaxonais/metabolismo , Substância Negra/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Animais , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Camundongos
7.
Stem Cell Reports ; 13(5): 832-846, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31587993

RESUMO

Neuroferritinopathy (NF) is a movement disorder caused by alterations in the L-ferritin gene that generate cytosolic free iron. NF is a unique pathophysiological model for determining the direct consequences of cell iron dysregulation. We established lines of induced pluripotent stem cells from fibroblasts from two NF patients and one isogenic control obtained by CRISPR/Cas9 technology. NF fibroblasts, neural progenitors, and neurons exhibited the presence of increased cytosolic iron, which was also detectable as: ferritin aggregates, alterations in the iron parameters, oxidative damage, and the onset of a senescence phenotype, particularly severe in the neurons. In this spontaneous senescence model, NF cells had impaired survival and died by ferroptosis. Thus, non-ferritin-bound iron is sufficient per se to cause both cell senescence and ferroptotic cell death in human fibroblasts and neurons. These results provide strong evidence supporting the primary role of iron in neuronal aging and degeneration.


Assuntos
Ferroptose , Distúrbios do Metabolismo do Ferro/patologia , Ferro/metabolismo , Distrofias Neuroaxonais/patologia , Neurônios/patologia , Células Cultivadas , Senescência Celular , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Distúrbios do Metabolismo do Ferro/metabolismo , Pessoa de Meia-Idade , Distrofias Neuroaxonais/metabolismo , Neurônios/metabolismo
8.
Metallomics ; 11(10): 1635-1647, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31513212

RESUMO

In mammals, the iron storage and detoxification protein ferritin is composed of two functionally and genetically distinct subunit types, H (heavy) and L (light). The two subunits co-assemble in various ratios, with a tissue specific distribution, to form shell-like protein structures of 24 subunits within which a mineralized iron core is stored. The H-subunits possess ferroxidase centers that catalyze the rapid oxidation of ferrous ions, whereas the L-subunit does not have such centers and is believed to play an important role in electron transfer reactions that occur during the uptake and release of iron. Pathogenic mutations on the L-chain lead to neuroferritinopathy, a neurodegenerative disease characterized by abnormal accumulation of ferritin inclusion bodies and iron in the central nervous system. Here, we have characterized the thermal stability, iron loading capacity, iron uptake, and iron release properties of ferritin heteropolymers carrying the three pathogenic L-ferritin mutants (L154fs, L167fs, and L148fs, which for simplicity we named Ln1, Ln2 and Ln3, respectively), and a non-pathogenic variant (L135P) bearing a single substitution on the 3-fold axes of L-subunits. The UV-Vis data show a similar iron loading capacity (ranging between 1800 to 2400 Fe(iii)/shell) for all ferritin samples examined in this study, with Ln2 holding the least amount of iron (i.e. 1800 Fe(iii)/shell). The three pathogenic L-ferritin mutants revealed higher rates of iron oxidation and iron release, suggesting that a few mutated L-chains on the heteropolymer have a significant effect on iron permeability through the ferritin shell. DSC thermograms showed a strong destabilization effect, the severity of which depends on the location of the frameshift mutations (i.e. wt heteropolymer ferritin ≅ homopolymer H-chain > L135P > Ln2 > Ln1 > Ln3). Variant L135P had only minor effects on the protein functionality and stability, suggesting that local melting of the 3-fold axes in this variant may not be responsible for neuroferritinopathy-like disorders. The data support the hypothesis that hereditary neuroferritinopathies are due to alterations of ferritin functionality and lower physical stability which correlate with the frameshifts introduced at the C-terminal sequence and explain the dominant transmission of the disorder.


Assuntos
Apoferritinas/genética , Apoferritinas/metabolismo , Distúrbios do Metabolismo do Ferro/genética , Ferro/metabolismo , Distrofias Neuroaxonais/genética , Apoferritinas/química , Humanos , Distúrbios do Metabolismo do Ferro/metabolismo , Modelos Moleculares , Distrofias Neuroaxonais/metabolismo , Oxirredução , Mutação Puntual , Estabilidade Proteica , Desdobramento de Proteína
9.
Biomed Pharmacother ; 118: 109068, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31404774

RESUMO

NBIA (Neurodegeneration with brain iron accumulation) is a group of inherited neurologic disorders characterized by marked genetic heterogeneity, in which iron atypical accumulates in basal ganglia resulting in brain magnetic resonance imaging changes, histopathological abnormalities, and neuropsychiatric clinical symptoms. With the rapid development of high-throughput sequencing technologies, ten candidate genes have been identified, including PANK2, PLA2G6, C19orf12, WDR45, FA2H, ATP13A2, FTL, CP, C2orf37, and COASY. They are involved in seemingly unrelated cellular pathways, such as iron homeostasis (FTL, CP), lipid metabolism (PLA2G6, C19orf12, FA2H), Coenzyme A synthesis (PANK2, COASY), and autophagy (WDR45, ATP13A2). In particular, PANK2, COASY, PLA2G6, and C19orf12 are located on mitochondria, which associate with certain subtypes of NBIA showing mitochondria dysregulation. However, the relationships among those four genes are still unclear. Therefore, this review is specifically focused on dysregulation of mitochondria in NBIA and afore-mentioned four genes, with summaries of both pathological and clinical findings.


Assuntos
Fosfolipases A2 do Grupo VI/genética , Distúrbios do Metabolismo do Ferro/genética , Mitocôndrias/patologia , Proteínas Mitocondriais/genética , Distrofias Neuroaxonais/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Transferases/genética , Humanos , Distúrbios do Metabolismo do Ferro/metabolismo , Distúrbios do Metabolismo do Ferro/patologia , Metabolismo dos Lipídeos/genética , Potencial da Membrana Mitocondrial/genética , Distrofias Neuroaxonais/metabolismo , Distrofias Neuroaxonais/patologia
10.
Brain Res ; 1712: 25-33, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30707893

RESUMO

PLA2G6-associated neurodegeneration (PLAN, NBIA2) is the second most common type of neurodegeneration with brain iron accumulation (NBIA), caused by recessive mutations of PLA2G6 gene, which encodes Ca2+-independent phospholipase A2ß (iPLA2ß). In most PLAN cases, decreased iPLA2ß activity and iron deposition was observed meanwhile, and researchers also identified a PLA2G6 mutation family without iron deposition shown by MRI images. This brought us the question of whether decreased iPLA2ß activity was the cause of iron deposition in PLAN. In this study, we used S-BEL as the antagonist of iPLA2ß to block its activity and used SH-SY5Y cells as the expression system. We incubated SH-SY5Y cells with different concentrations of S-BEL. The results showed that decreased iPLA2ß activity led no obvious iron accumulation, while changes of cells state and activation of apoptosis were observed. To further investigate the cause of unchanged iron level, we examined the cellular iron regulatory proteins involved in iron uptake, storage and export. The results were as follows: TfR1 (iron uptake protein) expression was decreased, the expression of ferritin heavy chain and light chain (iron storage protein) was increased. There was no alteration of the expression of DMT1 (iron uptake protein) and FPN1 (iron export protein). Under the condition of decreased iPLA2ß activity, there was no obvious iron accumulation but iron uptake activity decreased and iron storage activity increased. Therefore, we speculate that the decreased iPLA2ß activity may not be the main reason for iron deposition in PLAN.


Assuntos
Fosfolipases A2 do Grupo VI/metabolismo , Ferro/metabolismo , Distrofias Neuroaxonais/metabolismo , Antígenos CD/metabolismo , Apoferritinas/metabolismo , Fenômenos Bioquímicos , Transporte Biológico , Encéfalo/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular Tumoral , Fosfolipases A2 do Grupo VI/genética , Humanos , Imageamento por Ressonância Magnética , Naftalenos/farmacologia , Distrofias Neuroaxonais/genética , Pironas/farmacologia , Receptores da Transferrina/metabolismo , Fatores de Transcrição/metabolismo
12.
J Cell Physiol ; 233(12): 9179-9190, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30076709

RESUMO

Nuclear receptor coactivator 4 mediated ferritinophagy is an autophagic phenomenon that specifically involves ferritin to release intracellular free iron. Ferritinophagy is implicated in maintaining efficient erythropoiesis. Notably, ferritinophagy also plays a central role in driving some pathological processes, including Parkinson's disease (PD) and urinary tract infections. Some evidence has demonstrated that ferritinophagy is critical to induce ferroptosis. Ferroptosis is a newly nonapoptotic form of cell death, characterized by the accumulation of iron-based lipid reactive oxygen species. Ferroptosis plays an important role in inhibiting some types of cancers, such as hepatocellular carcinoma, pancreatic carcinoma, prostate cancer, and breast cancer. Conversely, the activation of ferroptosis accelerates neurodegeneration diseases, including PD and Alzheimer's disease. Therefore, in this review, we summarize the regulatory mechanisms related to ferritinophagy and ferroptosis. Moreover, the distinctive effects of ferritinophagy in human erythropoiesis and some pathologies, coupled with the promotive or inhibitory role of tumorous and neurodegenerative diseases mediated by ferroptosis, are elucidated. Obviously, activating or inhibiting ferroptosis could be exploited to achieve desirable therapeutic effects on diverse cancers and neurodegeneration diseases. Interrupting ferritinophagy to control iron level might provide a potentially therapeutic avenue to suppress urinary tract infections.


Assuntos
Ferritinas/metabolismo , Distúrbios do Metabolismo do Ferro/metabolismo , Ferro/metabolismo , Neoplasias/metabolismo , Distrofias Neuroaxonais/metabolismo , Autofagia/genética , Eritropoese/genética , Ferritinas/efeitos adversos , Humanos , Distúrbios do Metabolismo do Ferro/genética , Distúrbios do Metabolismo do Ferro/patologia , Neoplasias/classificação , Neoplasias/etiologia , Neoplasias/patologia , Distrofias Neuroaxonais/genética , Distrofias Neuroaxonais/patologia , Coativadores de Receptor Nuclear/genética , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Infecções Urinárias/genética , Infecções Urinárias/metabolismo
13.
Handb Clin Neurol ; 145: 157-166, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28987166

RESUMO

Neurodegeneration with brain iron accumulation (NBIA) describes a heterogeneous group of inherited rare clinical and genetic entities. Clinical core symptoms comprise a combination of early-onset dystonia, pyramidal and extrapyramidal signs with ataxia, cognitive decline, behavioral abnormalities, and retinal and axonal neuropathy variably accompanying these core features. Increased nonphysiologic, nonaging-associated brain iron, most pronounced in the basal ganglia, is often termed the unifying characteristic of these clinically variable disorders, though occurrence and extent can be fluctuating or even absent. Neuropathologically, NBIA disorders usually are associated with widespread axonal spheroids and local iron accumulation in the basal ganglia. Postmortem, Lewy body, TDP-43, or tau pathology has been observed. Genetics have fostered ongoing progress in elucidating underlying pathophysiologic mechanisms of NBIA disorders. Ten associated genes have been established, with many more being suggested as new technologies and data emerge. Clinically, certain symptom combinations can suggest a specific genetic defect. Genetic tests, combined with postmortem neuropathology, usually make for the final disease confirmation. Despite these advances, treatment to date remains mainly symptomatic. This chapter reviews the established genetic defects leading to different NBIA subtypes, highlights phenotypic presentations to direct genetic testing, and briefly discusses the scarce available treatment options and upcoming challenges and future hopes of the field.


Assuntos
Encéfalo/metabolismo , Distúrbios do Metabolismo do Ferro , Ferro/metabolismo , Mutação/genética , Distrofias Neuroaxonais , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Humanos , Distúrbios do Metabolismo do Ferro/genética , Distúrbios do Metabolismo do Ferro/metabolismo , Distúrbios do Metabolismo do Ferro/patologia , Distrofias Neuroaxonais/genética , Distrofias Neuroaxonais/metabolismo , Distrofias Neuroaxonais/patologia
14.
IUBMB Life ; 69(6): 423-434, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28276141

RESUMO

Ferroptosis is a recently described form of regulated necrotic cell death, which appears to contribute to a number of diseases, such as tissue ischemia/reperfusion injury, acute renal failure, and neurodegeneration. A hallmark of ferroptosis is iron-dependent lipid peroxidation, which can be inhibited by the key ferroptosis regulator glutathione peroxidase 4(Gpx4), radical trapping antioxidants and ferroptosis-specific inhibitors, such as ferrostatins and liproxstatins, as well as iron chelation. Although great strides have been made towards a better understanding of the proximate signals of distinctive lipid peroxides in ferroptosis, still little is known about the mechanistic implication of iron in the ferroptotic process. Hence, this review aims at summarizing recent advances in our understanding to what is known about enzymatic and nonenzymatic routes of lipid peroxidation, the involvement of iron in this process and the identification of novel players in ferroptotic cell death. Additionally, we review early works carried out long time before the term "ferroptosis" was actually introduced but which were instrumental in a better understanding of the role of ferroptosis in physiological and pathophysiological contexts. © 2017 IUBMB Life, 69(6):423-434, 2017.


Assuntos
Antioxidantes/farmacologia , Glutationa Peroxidase/antagonistas & inibidores , Distúrbios do Metabolismo do Ferro/metabolismo , Ferro/metabolismo , Distrofias Neuroaxonais/metabolismo , Insuficiência Renal/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Cicloexilaminas/farmacologia , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Humanos , Quelantes de Ferro/uso terapêutico , Distúrbios do Metabolismo do Ferro/tratamento farmacológico , Distúrbios do Metabolismo do Ferro/genética , Distúrbios do Metabolismo do Ferro/patologia , Peroxidação de Lipídeos/efeitos dos fármacos , Necrose/genética , Necrose/metabolismo , Necrose/patologia , Distrofias Neuroaxonais/tratamento farmacológico , Distrofias Neuroaxonais/genética , Distrofias Neuroaxonais/patologia , Fenilenodiaminas/farmacologia , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Quinoxalinas/farmacologia , Insuficiência Renal/tratamento farmacológico , Insuficiência Renal/genética , Insuficiência Renal/patologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/prevenção & controle , Compostos de Espiro/farmacologia
15.
PLoS One ; 11(8): e0161341, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27574973

RESUMO

Mutations in the ferritin light chain (FTL) gene cause the neurodegenerative disease neuroferritinopathy or hereditary ferritinopathy (HF). HF is characterized by a severe movement disorder and by the presence of nuclear and cytoplasmic iron-containing ferritin inclusion bodies (IBs) in glia and neurons throughout the central nervous system (CNS) and in tissues of multiple organ systems. Herein, using primary mouse embryonic fibroblasts from a mouse model of HF, we show significant intracellular accumulation of ferritin and an increase in susceptibility to oxidative damage when cells are exposed to iron. Treatment of the cells with the iron chelator deferiprone (DFP) led to a significant improvement in cell viability and a decrease in iron content. In vivo, iron overload and DFP treatment of the mouse model had remarkable effects on systemic iron homeostasis and ferritin deposition, without significantly affecting CNS pathology. Our study highlights the role of iron in modulating ferritin aggregation in vivo in the disease HF. It also puts emphasis on the potential usefulness of a therapy based on chelators that can target the CNS to remove and redistribute iron and to resolubilize or prevent ferritin aggregation while maintaining normal systemic iron stores.


Assuntos
Apoferritinas/metabolismo , Fibroblastos/efeitos dos fármacos , Quelantes de Ferro/administração & dosagem , Distúrbios do Metabolismo do Ferro/tratamento farmacológico , Sobrecarga de Ferro/tratamento farmacológico , Distrofias Neuroaxonais/tratamento farmacológico , Piridonas/administração & dosagem , Animais , Sobrevivência Celular , Células Cultivadas , Terapia por Quelação , Deferiprona , Modelos Animais de Doenças , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Ferro/efeitos adversos , Quelantes de Ferro/farmacologia , Distúrbios do Metabolismo do Ferro/metabolismo , Sobrecarga de Ferro/metabolismo , Masculino , Camundongos , Distrofias Neuroaxonais/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Piridonas/farmacologia
16.
Ideggyogy Sz ; 69(5-6): 157-66, 2016 Mar 30.
Artigo em Húngaro | MEDLINE | ID: mdl-27468605

RESUMO

The rare, genetically determined group of diseases characterized by pathological accumulation of iron in the central nervous system and progressive, typically movement disorder's symptoms are called NBIA (neurodegeneration with brain iron accumulation). By the rapid development of molecular genetics, it has become apparent that different mutations in numerous genes can lead to pathological cerebral iron accumulation. Simultaneously, it has also been recognized that the age of onset, the symptoms and the prognosis of NBIA disorders are much more diverse than it was previously perceived. To our knowledge, a review article on the most recent clinical data of NBIA has not been published in Hungarian. In the first part of this publication, we survey the general clinical characteristics and the diagnostic algorithm of NBIA diseases and address some considerations for differential diagnostics. In the second part of this review, the particular NBIA disorders are presented in details. The purpose of this article is to provide a clinical overview that may be useful for neurologists, pediatricians and any other medical practitioners interested in this field.


Assuntos
Encéfalo/metabolismo , Transtornos Heredodegenerativos do Sistema Nervoso/diagnóstico , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Distúrbios do Metabolismo do Ferro/diagnóstico , Distúrbios do Metabolismo do Ferro/genética , Ferro/metabolismo , Mutação , Distrofias Neuroaxonais/diagnóstico , Distrofias Neuroaxonais/genética , Alopecia/diagnóstico , Alopecia/genética , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/genética , Doenças dos Gânglios da Base/diagnóstico , Doenças dos Gânglios da Base/genética , Encéfalo/patologia , Ceruloplasmina/genética , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/genética , Fosfolipases A2 do Grupo VI/genética , Transtornos Heredodegenerativos do Sistema Nervoso/metabolismo , Transtornos Heredodegenerativos do Sistema Nervoso/terapia , Humanos , Hipogonadismo/diagnóstico , Hipogonadismo/genética , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/genética , Distúrbios do Metabolismo do Ferro/metabolismo , Distúrbios do Metabolismo do Ferro/terapia , Oxigenases de Função Mista/genética , Distrofias Neuroaxonais/metabolismo , Distrofias Neuroaxonais/terapia , Transtornos Parkinsonianos/diagnóstico , Transtornos Parkinsonianos/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Doenças Raras/diagnóstico , Doenças Raras/genética , Transferases/genética
17.
Neuropediatrics ; 47(2): 123-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26859818

RESUMO

Neurodegeneration with brain iron accumulation (NBIA) is a heterogeneous group of single gene disorders with distinguished clinical phenotypes and definitive imaging findings. Beta propeller protein-associated neurodegeneration (BPAN) is a subentity of NBIA with X linked dominant inheritance. In this report, we describe a girl with autistic regression, seizures, intracranial calcification, iron accumulation in substantia nigra, and globi pallidi, and diagnosis of BPAN was established based on the identification of previously described disease causing variant in WD repeat domain 45 (WDR45) gene encoding for ß propeller protein. This is the first genetically proven case from India. BPAN is an underrecognized disorder and must be considered as a differential diagnosis in children with atypical Rett features and should be enlisted among the causes for autistic regression and intracranial calcification. Pediatricians must be aware of this rare entity for establishing early diagnosis, prognostication, and genetic counseling. Treatment is usually supportive. More research is needed to explore drugs in the management of BPAN that can facilitate the autophagy and promotes cytoprotection.


Assuntos
Transtorno Autístico/etiologia , Doenças dos Gânglios da Base/genética , Doenças dos Gânglios da Base/patologia , Calcinose/etiologia , Proteínas de Transporte/genética , Distúrbios do Metabolismo do Ferro/genética , Distúrbios do Metabolismo do Ferro/patologia , Distrofias Neuroaxonais/genética , Distrofias Neuroaxonais/patologia , Gânglios da Base/metabolismo , Gânglios da Base/patologia , Doenças dos Gânglios da Base/complicações , Doenças dos Gânglios da Base/metabolismo , Pré-Escolar , Feminino , Humanos , Distúrbios do Metabolismo do Ferro/complicações , Distúrbios do Metabolismo do Ferro/metabolismo , Distrofias Neuroaxonais/complicações , Distrofias Neuroaxonais/metabolismo , Substância Negra/metabolismo , Substância Negra/patologia
18.
Mol Psychiatry ; 21(11): 1599-1607, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26728570

RESUMO

The 'neurodegeneration with brain iron accumulation' (NBIA) disease family entails movement or cognitive impairment, often with psychiatric features. To understand how iron loading affects the brain, we studied mice with disruption of two iron regulatory genes, hemochromatosis (Hfe) and transferrin receptor 2 (Tfr2). Inductively coupled plasma atomic emission spectroscopy demonstrated increased iron in the Hfe-/- × Tfr2mut brain (P=0.002, n ≥5/group), primarily localized by Perls' staining to myelinated structures. Western immunoblotting showed increases of the iron storage protein ferritin light polypeptide and microarray and real-time reverse transcription-PCR revealed decreased transcript levels (P<0.04, n ≥5/group) for five other NBIA genes, phospholipase A2 group VI, fatty acid 2-hydroxylase, ceruloplasmin, chromosome 19 open reading frame 12 and ATPase type 13A2. Apart from the ferroxidase ceruloplasmin, all are involved in myelin homeostasis; 16 other myelin-related genes also showed reduced expression (P<0.05), although gross myelin structure and integrity appear unaffected (P>0.05). Overlap (P<0.0001) of differentially expressed genes in Hfe-/- × Tfr2mut brain with human gene co-expression networks suggests iron loading influences expression of NBIA-related and myelin-related genes co-expressed in normal human basal ganglia. There was overlap (P<0.0001) of genes differentially expressed in Hfe-/- × Tfr2mut brain and post-mortem NBIA basal ganglia. Hfe-/- × Tfr2mut mice were hyperactive (P<0.0112) without apparent cognitive impairment by IntelliCage testing (P>0.05). These results implicate myelin-related systems involved in NBIA neuropathogenesis in early responses to iron loading. This may contribute to behavioral symptoms in NBIA and hemochromatosis and is relevant to patients with abnormal iron status and psychiatric disorders involving myelin abnormalities or resistant to conventional treatments.


Assuntos
Distúrbios do Metabolismo do Ferro/metabolismo , Distúrbios do Metabolismo do Ferro/fisiopatologia , Ferro/efeitos adversos , Distrofias Neuroaxonais/metabolismo , Distrofias Neuroaxonais/fisiopatologia , Animais , Encéfalo/metabolismo , Expressão Gênica , Hemocromatose/genética , Hemocromatose/metabolismo , Proteína da Hemocromatose/metabolismo , Proteína da Hemocromatose/fisiologia , Ferro/sangue , Ferro/metabolismo , Distúrbios do Metabolismo do Ferro/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos AKR , Bainha de Mielina/metabolismo , Distrofias Neuroaxonais/genética , Linhagem , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo
19.
Neurobiol Dis ; 87: 59-68, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26707700

RESUMO

Aberrant brain iron deposition is observed in both common and rare neurodegenerative disorders, including those categorized as Neurodegeneration with Brain Iron Accumulation (NBIA), which are characterized by focal iron accumulation in the basal ganglia. Two NBIA genes are directly involved in iron metabolism, but whether other NBIA-related genes also regulate iron homeostasis in the human brain, and whether aberrant iron deposition contributes to neurodegenerative processes remains largely unknown. This study aims to expand our understanding of these iron overload diseases and identify relationships between known NBIA genes and their main interacting partners by using a systems biology approach. We used whole-transcriptome gene expression data from human brain samples originating from 101 neuropathologically normal individuals (10 brain regions) to generate weighted gene co-expression networks and cluster the 10 known NBIA genes in an unsupervised manner. We investigated NBIA-enriched networks for relevant cell types and pathways, and whether they are disrupted by iron loading in NBIA diseased tissue and in an in vivo mouse model. We identified two basal ganglia gene co-expression modules significantly enriched for NBIA genes, which resemble neuronal and oligodendrocytic signatures. These NBIA gene networks are enriched for iron-related genes, and implicate synapse and lipid metabolism related pathways. Our data also indicates that these networks are disrupted by excessive brain iron loading. We identified multiple cell types in the origin of NBIA disorders. We also found unforeseen links between NBIA networks and iron-related processes, and demonstrate convergent pathways connecting NBIAs and phenotypically overlapping diseases. Our results are of further relevance for these diseases by providing candidates for new causative genes and possible points for therapeutic intervention.


Assuntos
Gânglios da Base/metabolismo , Perfilação da Expressão Gênica/métodos , Distúrbios do Metabolismo do Ferro/metabolismo , Distrofias Neuroaxonais/metabolismo , Transcriptoma , Adolescente , Idoso , Idoso de 80 Anos ou mais , Animais , Gânglios da Base/patologia , Criança , Pré-Escolar , Modelos Animais de Doenças , Feminino , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Lactente , Ferro/metabolismo , Distúrbios do Metabolismo do Ferro/patologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Transgênicos , Distrofias Neuroaxonais/patologia , Neurônios/metabolismo , Neurônios/patologia , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Substância Branca/metabolismo , Substância Branca/patologia , Adulto Jovem
20.
Annu Rev Genomics Hum Genet ; 16: 257-79, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25973518

RESUMO

Neurodegeneration with brain iron accumulation (NBIA) comprises a heterogeneous group of progressive disorders with the common feature of excessive iron deposition in the brain. Over the last decade, advances in sequencing technologies have greatly facilitated rapid gene discovery, and several single-gene disorders are now included in this group. Identification of the genetic bases of the NBIA disorders has advanced our understanding of the disease processes caused by reduced coenzyme A synthesis, impaired lipid metabolism, mitochondrial dysfunction, and defective autophagy. The contribution of iron to disease pathophysiology remains uncertain, as does the identity of a putative final common pathway by which the iron accumulates. Ongoing elucidation of the pathogenesis of each NBIA disorder will have significant implications for the identification and design of novel therapies to treat patients with these disorders.


Assuntos
Encéfalo/metabolismo , Variação Genética , Distúrbios do Metabolismo do Ferro/genética , Distúrbios do Metabolismo do Ferro/fisiopatologia , Ferro/metabolismo , Doenças Neurodegenerativas/genética , Animais , Autofagia/genética , Encéfalo/fisiopatologia , Ceruloplasmina/deficiência , Ceruloplasmina/genética , Ceruloplasmina/metabolismo , Coenzima A/biossíntese , Fosfolipases A2 do Grupo VI/genética , Fosfolipases A2 do Grupo VI/metabolismo , Humanos , Distúrbios do Metabolismo do Ferro/metabolismo , Metabolismo dos Lipídeos/genética , Camundongos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Distrofias Neuroaxonais/genética , Distrofias Neuroaxonais/metabolismo , Distrofias Neuroaxonais/fisiopatologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/fisiopatologia , Transtornos Parkinsonianos/genética , Transtornos Parkinsonianos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA