Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 81(1): 12, 2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38129330

RESUMO

Friedreich ataxia (FA) is a rare, recessive neuro-cardiodegenerative disease caused by deficiency of the mitochondrial protein frataxin. Mitochondrial dysfunction, a reduction in the activity of iron-sulfur enzymes, iron accumulation, and increased oxidative stress have been described. Dorsal root ganglion (DRG) sensory neurons are among the cellular types most affected in the early stages of this disease. However, its effect on mitochondrial function remains to be elucidated. In the present study, we found that in primary cultures of DRG neurons as well as in DRGs from the FXNI151F mouse model, frataxin deficiency resulted in lower activity and levels of the electron transport complexes, mainly complexes I and II. In addition, altered mitochondrial morphology, indicative of degeneration was observed in DRGs from FXNI151F mice. Moreover, the NAD+/NADH ratio was reduced and sirtuin activity was impaired. We identified alpha tubulin as the major acetylated protein from DRG homogenates whose levels were increased in FXNI151F mice compared to WT mice. In the mitochondria, superoxide dismutase (SOD2), a SirT3 substrate, displayed increased acetylation in frataxin-deficient DRG neurons. Since SOD2 acetylation inactivates the enzyme, and higher levels of mitochondrial superoxide anion were detected, oxidative stress markers were analyzed. Elevated levels of hydroxynonenal bound to proteins and mitochondrial Fe2+ accumulation was detected when frataxin decreased. Honokiol, a SirT3 activator, restores mitochondrial respiration, decreases SOD2 acetylation and reduces mitochondrial superoxide levels. Altogether, these results provide data at the molecular level of the consequences of electron transport chain dysfunction, which starts negative feedback, contributing to neuron lethality. This is especially important in sensory neurons which have greater susceptibility to frataxin deficiency compared to other tissues.


Assuntos
Ataxia de Friedreich , Sirtuína 3 , Sirtuínas , Camundongos , Animais , Sirtuína 3/metabolismo , Gânglios Espinais/metabolismo , Sirtuínas/metabolismo , Acetilação , Proteínas de Ligação ao Ferro/genética , Frataxina , Mitocôndrias/metabolismo , Superóxido Dismutase/metabolismo , Ferro/metabolismo
2.
Cell Mol Life Sci ; 79(2): 74, 2022 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-35038030

RESUMO

Friedreich Ataxia (FA) is a rare neuro-cardiodegenerative disease caused by mutations in the frataxin (FXN) gene. The most prevalent mutation is a GAA expansion in the first intron of the gene causing decreased frataxin expression. Some patients present the GAA expansion in one allele and a missense mutation in the other allele. One of these mutations, FXNI154F, was reported to result in decreased content of mature frataxin and increased presence of an insoluble intermediate proteoform in cellular models. By introducing this mutation into the murine Fxn gene (I151F, equivalent to human I154F) we have now analyzed the consequences of this pathological point mutation in vivo. We have observed that FXNI151F homozygous mice present low frataxin levels in all tissues, with no evidence of insoluble proteoforms. Moreover, they display neurological deficits resembling those observed in FA patients. Biochemical analysis of heart, cerebrum and cerebellum have revealed decreased content of components from OXPHOS complexes I and II, decreased aconitase activity, and alterations in antioxidant defenses. These mitochondrial alterations are more marked in the nervous system than in heart, precede the appearance of neurological symptoms, and are similar to those observed in other FA models. We conclude that the primary pathological mechanism underlying the I151F mutation is frataxin deficiency, like in patients carrying GAA expansions. Therefore, patients carrying the I154F mutation would benefit from frataxin replacement therapies. Furthermore, our results also show that the FXNI151F mouse is an excellent tool for analyzing tissue-specific consequences of frataxin deficiency and for testing new therapies.


Assuntos
Ataxia de Friedreich/genética , Proteínas de Ligação ao Ferro/genética , Mitocôndrias/metabolismo , Doenças Mitocondriais/genética , Mutação Puntual , Alelos , Animais , Comportamento Animal , Biomarcadores/metabolismo , Códon , Modelos Animais de Doenças , Feminino , Ataxia de Friedreich/fisiopatologia , Células HEK293 , Humanos , Íntrons , Proteínas de Ligação ao Ferro/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Mitocondriais/fisiopatologia , Mutação , Mutação de Sentido Incorreto , Fenótipo , Proteômica , Aumento de Peso , Frataxina
3.
Biochem J ; 478(1): 1-20, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33305808

RESUMO

Friedreich ataxia (FA) is a neurodegenerative disease caused by the deficiency of frataxin, a mitochondrial protein. In primary cultures of dorsal root ganglia neurons, we showed that frataxin depletion resulted in decreased levels of the mitochondrial calcium exchanger NCLX, neurite degeneration and apoptotic cell death. Here, we describe that frataxin-deficient dorsal root ganglia neurons display low levels of ferredoxin 1 (FDX1), a mitochondrial Fe/S cluster-containing protein that interacts with frataxin and, interestingly, is essential for the synthesis of calcitriol, the active form of vitamin D. We provide data that calcitriol supplementation, used at nanomolar concentrations, is able to reverse the molecular and cellular markers altered in DRG neurons. Calcitriol is able to recover both FDX1 and NCLX levels and restores mitochondrial membrane potential indicating an overall mitochondrial function improvement. Accordingly, reduction in apoptotic markers and neurite degeneration was observed and, as a result, cell survival was also recovered. All these beneficial effects would be explained by the finding that calcitriol is able to increase the mature frataxin levels in both, frataxin-deficient DRG neurons and cardiomyocytes; remarkably, this increase also occurs in lymphoblastoid cell lines derived from FA patients. In conclusion, these results provide molecular bases to consider calcitriol for an easy and affordable therapeutic approach for FA patients.


Assuntos
Calcitriol/farmacologia , Ferredoxinas/metabolismo , Ataxia de Friedreich/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Neurônios/metabolismo , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/metabolismo , Apoptose/efeitos dos fármacos , Calcitriol/biossíntese , Calcitriol/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Humanos , Proteínas dos Microfilamentos/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Vitamina D/metabolismo , Frataxina
4.
Neurobiol Dis ; 148: 105162, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33171227

RESUMO

Friedreich ataxia (FRDA), the most common autosomal recessive ataxia, is characterized by degeneration of the large sensory neurons and spinocerebellar tracts, cardiomyopathy, and increased incidence in diabetes. The underlying pathophysiological mechanism of FRDA, driven by a significantly decreased expression of frataxin (FXN), involves increased oxidative stress, reduced activity of enzymes containing iron­sulfur clusters (ISC), defective energy production, calcium dyshomeostasis, and impaired mitochondrial biogenesis, leading to mitochondrial dysfunction. The peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-activated transcriptional factor playing a key role in mitochondrial function and biogenesis, fatty acid storage, energy metabolism, and antioxidant defence. It has been previously shown that the PPARγ/PPARγ coactivator 1 alpha (PGC-1α) pathway is dysregulated when there is frataxin deficiency, thus contributing to FRDA pathogenesis and supporting the PPARγ pathway as a potential therapeutic target. Here we assess whether MIN-102 (INN: leriglitazone), a novel brain penetrant and orally bioavailable PPARγ agonist with an improved profile for central nervous system (CNS) diseases, rescues phenotypic features in cellular and animal models of FRDA. In frataxin-deficient dorsal root ganglia (DRG) neurons, leriglitazone increased frataxin protein levels, reduced neurite degeneration and α-fodrin cleavage mediated by calpain and caspase 3, and increased survival. Leriglitazone also restored mitochondrial membrane potential and partially reversed decreased levels of mitochondrial Na+/Ca2+ exchanger (NCLX), resulting in an improvement of mitochondrial functions and calcium homeostasis. In frataxin-deficient primary neonatal cardiomyocytes, leriglitazone prevented lipid droplet accumulation without increases in frataxin levels. Furthermore, leriglitazone improved motor function deficit in YG8sR mice, a FRDA mouse model. In agreement with the role of PPARγ in mitochondrial biogenesis, leriglitazone significantly increased markers of mitochondrial biogenesis in FRDA patient cells. Overall, these results suggest that targeting the PPARγ pathway by leriglitazone may provide an efficacious therapy for FRDA increasing the mitochondrial function and biogenesis that could increase frataxin levels in compromised frataxin-deficient DRG neurons. Alternately, leriglitazone improved the energy metabolism by increasing the fatty acid ß-oxidation in frataxin-deficient cardiomyocytes without elevation of frataxin levels. This could be linked to a lack of significant mitochondrial biogenesis and cardiac hypertrophy. The results reinforced the different tissue requirement in FRDA and the pleiotropic effects of leriglitazone that could be a promising therapy for FRDA.


Assuntos
Ataxia de Friedreich/metabolismo , Proteínas de Ligação ao Ferro/efeitos dos fármacos , Gotículas Lipídicas/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , PPAR gama/agonistas , Tiazolidinedionas/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Ataxia de Friedreich/patologia , Ataxia de Friedreich/fisiopatologia , Humanos , Proteínas de Ligação ao Ferro/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Neuritos/efeitos dos fármacos , Neuritos/patologia , Neurônios/metabolismo , Neurônios/patologia , Ratos , Frataxina
5.
IUBMB Life ; 73(3): 543-553, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33675183

RESUMO

Friedreich Ataxia is a neuro-cardiodegenerative disease caused by the deficiency of frataxin, a mitochondrial protein. Many evidences indicate that frataxin deficiency causes an unbalance of iron homeostasis. Nevertheless, in the last decade many results also highlighted the importance of calcium unbalance in the deleterious downstream effects caused by frataxin deficiency. In this review, the role of these two metals has been gathered to give a whole view of how iron and calcium dyshomeostasys impacts on cellular functions and, as a result, which strategies can be followed to find an effective therapy for the disease.


Assuntos
Cálcio/metabolismo , Ataxia de Friedreich/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Ferro/metabolismo , Mitocôndrias/metabolismo , Ataxia de Friedreich/tratamento farmacológico , Ataxia de Friedreich/fisiopatologia , Homeostase , Humanos , Quelantes de Ferro/farmacologia , Proteínas de Ligação ao Ferro/química , Frataxina
6.
Neurochem Res ; 46(1): 108-119, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32249386

RESUMO

Calpains are calcium-dependent proteases activated in apoptotic cell death and neurodegeneration. Friedreich Ataxia is a neurodegenerative rare disease caused by frataxin deficiency, a mitochondrial protein. Dorsal root ganglion (DRG) sensory neurons are among the cellular types most affected in this disease. We have previously demonstrated that frataxin-deficient DRGs show calpain activation, alteration in calcium levels and decreased content of the Na+/Ca2+ exchanger (NCLX). This transporter is involved in mitochondrial calcium efflux. In this study, we have performed a time-course analysis of several parameters altered in a frataxin-deficient DRGs. These include decline of NCLX levels, calcium accumulation, mitochondrial depolarization, α-fodrin fragmentation and apoptotic cell death. Furthermore, we have analysed the effect of the calpain inhibitors MDL28170 and Calpeptin on these parameters. We have observed that these inhibitors increase NCLX levels, protect sensory neurons from neurite degeneration and calcium accumulation, and restore mitochondrial membrane potential. In addition, calpain 1 reduction alleviated neurodegeneration in frataxin-deficient DRG neurons. These results strengthen the hypothesis of a central role for calcium homeostasis and calpains in frataxin-deficient dorsal root ganglia neurons.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Dipeptídeos/farmacologia , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Neurônios/efeitos dos fármacos , Trocador de Sódio e Cálcio/metabolismo , Animais , Cálcio/metabolismo , Calpaína/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Gânglios Espinais/citologia , Proteínas de Ligação ao Ferro/metabolismo , Proteínas dos Microfilamentos/metabolismo , Mitocôndrias/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Ratos , Frataxina
7.
J Cell Mol Med ; 22(2): 834-848, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28980774

RESUMO

Friedreich ataxia (FA) is a rare disease caused by deficiency of frataxin, a mitochondrial protein. As there is no cure available for this disease, many strategies have been developed to reduce the deleterious effects of such deficiency. One of these approaches is based on delivering frataxin to the tissues by coupling the protein to trans-activator of transcription (TAT) peptides, which enables cell membranes crossing. In this study, we tested the efficiency of TAT-MTScs-FXN fusion protein to decrease neurodegeneration markers on frataxin-depleted neurons obtained from dorsal root ganglia (DRG), one of the most affected tissues. In mice models of the disease, we tested the ability of TAT-MTScs-FXN to penetrate the mitochondria and its effect on lifespan. In DRG neurons, treatment with TAT-MTScs-FXN increased cell survival, decreased neurite degeneration and reduced apoptotic markers, such as α-fodrin cleavage and caspase 9 activation. Also, we show that heat-shock protein 60 (HSP60), a molecular chaperone targeted to mitochondria, suffered an impaired processing in frataxin-deficient neurons that was relieved by TAT-MTScs-FXN addition. In mice models of the disease, administration of TAT-MTScs-FXN was able to reach muscle mitochondria, restore the activity of the succinate dehydrogenase and produce a significant lifespan increase. These results support the use of TAT-MTScs-FXN as a treatment for Friedreich ataxia.


Assuntos
Ataxia de Friedreich/patologia , Ataxia de Friedreich/terapia , Proteínas de Ligação ao Ferro/metabolismo , Neurônios/patologia , Sinais Direcionadores de Proteínas , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Animais , Proteínas de Transporte/metabolismo , Sobrevivência Celular , Chaperonina 60/metabolismo , Modelos Animais de Doenças , Gânglios Espinais/patologia , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Mitocôndrias/metabolismo , Músculos/metabolismo , Degeneração Neural/patologia , Neuritos/metabolismo , Ratos , Análise de Sobrevida , Frataxina
8.
Cell Calcium ; 86: 102150, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31918031

RESUMO

Aggregation or phosphorylation of the microtubule-associated protein tau is the pathological hallmark in a number of diseases termed tauopathies, which include the most common neurodegenerative disorder, Alzheimer's disease; or frontotemporal dementia, linked to mutations in the gene MAPT encoding tau. Although misfolded tau has strong familial and histopathological (as in intracellular tangles) association with neurodegenerative disorders, the cellular mechanism of tau-induced pathology remains to be controversial. Here we studied the effect of tau on the cytosolic and mitochondrial calcium homeostasis using primary cortical cultures treated with the protein and iPSC-derived neurons bearing the 10 + 16 MAPT mutation linked to frontotemporal dementia. We found that incubation of the primary cortical co-cultures of neurons and astrocytes with tau induced spontaneous Ca2+ oscillations in the neurons, which were also observed in iPSC-neurons with the 10 + 16 MAPT mutation. Importantly, tau inhibited mitochondrial calcium efflux via the mitochondrial Na+/Ca2+ exchanger (NCLX) in both neurons and astrocytes. This inhibition led to mitochondrial depolarisation in response to physiological and pathological calcium stimuli and made these cells vulnerable to calcium-induced caspase 3 activation and cell death. Thus, inhibition of the mitochondrial NCLX in neurons with misfolded or mutated tau can be involved in the mechanism of neurodegeneration.


Assuntos
Cálcio/metabolismo , Mitocôndrias/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Proteínas tau/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Transporte Biológico/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Citosol/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mutação/genética , Neurônios/efeitos dos fármacos , Ratos Sprague-Dawley , Trocador de Sódio e Cálcio/metabolismo , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/genética
9.
Neuronal Signal ; 2(4): NS20180061, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32714593

RESUMO

Calcium is utilised by cells in signalling and in regulating ATP production; it also contributes to cell survival and, when concentrations are unbalanced, triggers pathways for cell death. Mitochondria contribute to calcium buffering, meaning that mitochondrial calcium uptake and release is intimately related to cytosolic calcium concentrations. This review focuses on the proteins contributing to mitochondrial calcium homoeostasis, the roles of the mitochondrial permeability transition pore (MPTP) and mitochondrial calcium-activated proteins, and their relevance in neurodegenerative pathologies. It also covers alterations to calcium homoeostasis in Friedreich ataxia (FA).

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA