Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Int J Mol Sci ; 24(2)2023 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-36674605

RESUMO

Lafora disease is a rare, fatal form of progressive myoclonus epilepsy characterized by continuous neurodegeneration with epileptic seizures, characterized by the intracellular accumulation of aberrant polyglucosan granules called Lafora bodies. Several works have provided numerous evidence of molecular and cellular alterations in neural tissue from experimental mouse models deficient in either laforin or malin, two proteins related to the disease. Oxidative stress, alterations in proteostasis, and deregulation of inflammatory signals are some of the molecular alterations underlying this condition in both KO animal models. Lafora bodies appear early in the animal's life, but many of the aforementioned molecular aberrant processes and the consequent neurological symptoms ensue only as animals age. Here, using small RNA-seq and quantitative PCR on brain extracts from laforin and malin KO male mice of different ages, we show that two different microRNA species, miR-155 and miR-146a, are overexpressed in an age-dependent manner. We also observed altered expression of putative target genes for each of the microRNAs studied in brain extracts. These results open the path for a detailed dissection of the molecular consequences of laforin and malin deficiency in brain tissue, as well as the potential role of miR-155 and miR-146a as specific biomarkers of disease progression in LD.


Assuntos
Doença de Lafora , MicroRNAs , Camundongos , Masculino , Animais , MicroRNAs/genética , MicroRNAs/metabolismo , Doença de Lafora/genética , Doença de Lafora/metabolismo , Doenças Neuroinflamatórias , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Estresse Oxidativo/genética , Ubiquitina-Proteína Ligases/metabolismo
2.
Proteomics ; 16(1): 150-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26507101

RESUMO

Niemann-Pick type C (NPC) disease is an inherited lysosomal storage disorder, characterized by severe neurodegeneration. It is mostly produced by mutations in the NPC1 gene, encoding for a protein of the late endosomes/lysosomes membrane, involved in cholesterol metabolism. However, the specific role of this protein in NPC disease still remains unknown. We aimed to identify Npc1-binding proteins in order to define new putative NPC1 lysosomal functions. By affinity chromatography using an Npc1 peptide (amino acids 1032-1066 of loop I), as bait, we fished 31 lysosomal proteins subsequently identified by LC-MS/MS. Most of them were involved in proteolysis and lipid catabolism and included the protease cathepsin D. Cathepsin D and NPC1 interaction was validated by immunoprecipitation and the functional relevance of this interaction was studied. We found that fibroblasts from NPC patients with low levels of NPC1 protein have high amounts of procathepsin D but reduced quantities of the mature protein, thus showing a diminished cathepsin D activity. The increase of NPC1 protein levels in NPC cells by treatment with the proteasome inhibitor bortezomib, induced an elevation of cathepsin D activity. All these results suggest a new lysosomal function of NPC1 as a regulator of cathepsin D processing and activity.


Assuntos
Proteínas de Transporte/metabolismo , Catepsina D/metabolismo , Precursores Enzimáticos/metabolismo , Glicoproteínas de Membrana/metabolismo , Doenças de Niemann-Pick/metabolismo , Proteínas/metabolismo , Sequência de Aminoácidos , Proteínas de Transporte/análise , Catepsina D/análise , Linhagem Celular , Cromatografia Líquida , Precursores Enzimáticos/análise , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Glicoproteínas de Membrana/análise , Dados de Sequência Molecular , Proteína C1 de Niemann-Pick , Mapas de Interação de Proteínas , Proteínas/análise , Espectrometria de Massas em Tandem
3.
Acta Neuropathol ; 129(3): 399-415, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25549970

RESUMO

X-linked adrenoleukodystrophy (X-ALD) is a rare neurometabolic disease characterized by the accumulation of very long chain fatty acids (VLCFAs) due to a loss of function of the peroxisomal transporter ABCD1. Here, using in vivo and in vitro models, we demonstrate that autophagic flux was impaired due to elevated mammalian target of rapamycin (mTOR) signaling, which contributed to X-ALD pathogenesis. We also show that excess VLCFAs downregulated autophagy in human fibroblasts. Furthermore, mTOR inhibition by a rapamycin derivative (temsirolimus) restored autophagic flux and inhibited the axonal degenerative process as well as the associated locomotor impairment in the Abcd1 (-) /Abcd2 (-/-) mouse model. This process was mediated through the restoration of proteasome function and redox as well as metabolic homeostasis. These findings provide the first evidence that links impaired autophagy to X-ALD, which may yield a therapy based on autophagy activators for adrenomyeloneuropathy patients.


Assuntos
Adrenoleucodistrofia/patologia , Adrenoleucodistrofia/fisiopatologia , Autofagia/fisiologia , Degeneração Neural/fisiopatologia , Adulto , Animais , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Degeneração Neural/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/metabolismo
4.
Brain ; 137(Pt 3): 806-18, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24430976

RESUMO

Lafora progressive myoclonus epilepsy (Lafora disease) is a fatal autosomal recessive neurodegenerative disorder characterized by the presence of glycogen-like intracellular inclusions called Lafora bodies. The vast majority of patients carry mutations in either the EPM2A or EPM2B genes, encoding laforin, a glucan phosphatase, and malin, an E3 ubiquitin ligase, respectively. Although the precise physiological role of these proteins is not fully understood, work in past years has established a link between glycogen synthesis, Lafora bodies formation and Lafora disease development. To determine the role of the phosphatase activity of laforin in disease development we generated two Epm2a(-/-) mouse lines expressing either wild-type laforin or a mutant (C265S) laforin lacking only the phosphatase activity. Our results demonstrate that expression of either transgene blocks formation of Lafora bodies and restores the impairment in macroautophagy, preventing the development of Lafora bodies in Epm2a(-/-) mice. These data indicate that the critical pathogenic process is the control of abnormal glycogen accumulation through intracellular proteolytic systems by the laforin-malin complex, and not glycogen dephosphorylation by laforin. Understanding which is the essential process leading to Lafora disease pathogenesis represents a critical conceptual advance that should facilitate development of appropriate therapeutics.


Assuntos
Fosfatases de Especificidade Dupla/deficiência , Fosfatases de Especificidade Dupla/metabolismo , Doença de Lafora/metabolismo , Animais , Autofagia/genética , Modelos Animais de Doenças , Fosfatases de Especificidade Dupla/genética , Feminino , Doença de Lafora/enzimologia , Doença de Lafora/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação , Fosforilação/genética , Proteínas Tirosina Fosfatases não Receptoras
5.
Hum Mol Genet ; 21(7): 1521-33, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22186026

RESUMO

Lafora disease (LD), a fatal neurodegenerative disorder characterized by the presence of intracellular inclusions called Lafora bodies (LBs), is caused by loss-of-function mutations in laforin or malin. Previous studies suggested a role of these proteins in the regulation of glycogen biosynthesis, in glycogen dephosphorylation and in the modulation of the intracellular proteolytic systems. However, the contribution of each of these processes to LD pathogenesis is unclear. We have generated a malin-deficient (Epm2b-/-) mouse with a phenotype similar to that of LD patients. By 3-6 months of age, Epm2b-/- mice present neurological and behavioral abnormalities that correlate with a massive presence of LBs in the cortex, hippocampus and cerebellum. Sixteen-day-old Epm2b-/- mice, without detectable LBs, show an impairment of macroautophagy (hereafter called autophagy), which remains compromised in adult animals. These data demonstrate similarities between the Epm2a-/- and Epm2b-/- mice that provide further insights into LD pathogenesis. They illustrate that the dysfunction of autophagy is a consequence of the lack of laforin-malin complexes and a common feature of both mouse models of LD. Because this dysfunction precedes other pathological manifestations, we propose that decreased autophagy plays a primary role in the formation of LBs and it is critical in LD pathogenesis.


Assuntos
Autofagia , Doença de Lafora/patologia , Ubiquitina-Proteína Ligases/genética , Animais , Comportamento Animal , Encéfalo/metabolismo , Encéfalo/patologia , Fosfatases de Especificidade Dupla/análise , Fosfatases de Especificidade Dupla/metabolismo , Glucanos/química , Doença de Lafora/genética , Doença de Lafora/fisiopatologia , Camundongos , Camundongos Knockout , Transtornos das Habilidades Motoras/genética , Miocárdio/ultraestrutura , Proteínas Tirosina Fosfatases não Receptoras , Ubiquitina/análise , Ubiquitina-Proteína Ligases/deficiência
6.
J Cell Sci ; 125(Pt 1): 92-107, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22266906

RESUMO

Macroautophagy is a major lysosomal catabolic process activated particularly under starvation in eukaryotic cells. A new organelle, the autophagosome, engulfs cytoplasmic substrates, which are degraded after fusion with endosomes and/or lysosomes. During a shotgun proteome analysis of purified lysosomal membranes from mouse fibroblasts, a Ca(2+)-dependent phospholipid-binding protein, annexin A5, was found to increase on lysosomal membranes under starvation. This suggests a role for this protein, an abundant annexin with a still unknown intracellular function, in starvation-induced lysosomal degradation. Transient overexpression and silencing experiments showed that annexin A5 increased lysosomal protein degradation, and colocalisation experiments, based on GFP sensitivity to lysosomal acidic pH, indicated that this was mainly the result of inducing autophagosome-lysosome fusion. Annexin A5 also inhibited the endocytosis of a fluid-phase marker and cholera toxin, but not receptor-mediated endocytosis. Therefore, we propose a double and opposite role of annexin A5 in regulating the endocytic and autophagic pathways and the fusion of autophagosomes with lysosomes and endosomes.


Assuntos
Anexina A5/metabolismo , Autofagia , Endocitose , Animais , Anexina A5/deficiência , Anexina A5/genética , Sinalização do Cálcio , Toxina da Cólera/metabolismo , Eletroforese em Gel Bidimensional , Endossomos/metabolismo , Fibroblastos , Privação de Alimentos , Complexo de Golgi/metabolismo , Células HEK293 , Humanos , Membranas Intracelulares/metabolismo , Lisossomos/metabolismo , Fusão de Membrana , Camundongos , Células NIH 3T3 , Fagossomos/metabolismo , Transporte Proteico , Proteólise , Proteômica
7.
Hum Mol Genet ; 19(14): 2867-76, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20453062

RESUMO

Lafora disease (LD) is an autosomal recessive, progressive myoclonus epilepsy, which is characterized by the accumulation of polyglucosan inclusion bodies, called Lafora bodies, in the cytoplasm of cells in the central nervous system and in many other organs. However, it is unclear at the moment whether Lafora bodies are the cause of the disease, or whether they are secondary consequences of a primary metabolic alteration. Here we describe that the major genetic lesion that causes LD, loss-of-function of the protein laforin, impairs autophagy. This phenomenon is confirmed in cell lines from human patients, mouse embryonic fibroblasts from laforin knockout mice and in tissues from such mice. Conversely, laforin expression stimulates autophagy. Laforin regulates autophagy via the mammalian target of rapamycin kinase-dependent pathway. The changes in autophagy mediated by laforin regulate the accumulation of diverse autophagy substrates and would be predicted to impact on the Lafora body accumulation and the cell stress seen in this disease that may eventually contribute to cell death.


Assuntos
Autofagia/genética , Doença de Lafora/genética , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Doença de Lafora/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mutantes/genética , Proteínas Mutantes/fisiologia , Fagossomos/genética , Fagossomos/metabolismo , Processamento de Proteína Pós-Traducional/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR
8.
Transplant Proc ; 53(9): 2721-2723, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34600758

RESUMO

BACKGROUND: To analyze the relationship of the antigen carbohydrate 125 (CA125) biomarker with the cellular rejection of the heart graft during the first year after transplantation. METHODS: Retrospective study of consecutive heart transplant (HTx) patients for 1.5 years. The total number of patients included in the study was 23 with a total of 103 follow-ups. In all patients, CA125 was determined before HTx and determined post-HTx in every follow-up. These were performed during months 1, 2, 4, 6, 9, and 12. Endomyocardial biopsy was performed in all revisions to assess the degree of graft rejection in the pathologic study. The biopsy results were grouped into 1. absence of rejection and 2. presence of some degree of rejection. RESULTS: The mean pretransplant CA125 value presented a median of 120 U/mL with an interquartile range of 28.8 U/mL. One month after transplantation, the value was reduced by 20% and at 2 months by 81%. In subsequent reviews, plasma values were always between 10 and 20 U/mL. When comparing the values by periods and according to the presence or absence of rejection, no significant differences were found other than a slight elevation at the 6-month checkup (P = .03) but without clinical relevance, because the CA125 value was slightly higher in biopsy results without rejection. CONCLUSION: The rapid reduction of CA125 corroborates its usefulness as a marker of congestion in heart failure. This biomarker is not useful for predicting rejection. However, in cases of very severe rejections that occurred with systemic congestion, it could be raised. It would be necessary to corroborate this hypothesis in a larger study with a higher number of severe rejections.


Assuntos
Transplante de Coração , Transplante de Células-Tronco Hematopoéticas , Biomarcadores , Biópsia , Carboidratos , Rejeição de Enxerto/diagnóstico , Transplante de Coração/efeitos adversos , Humanos , Estudos Retrospectivos
9.
Hum Mol Genet ; 17(5): 667-78, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18029386

RESUMO

Lafora progressive myoclonus epilepsy (LD) is a fatal autosomal recessive neurodegenerative disorder characterized by the presence of glycogen-like intracellular inclusions called Lafora bodies. LD is caused by mutations in two genes, EPM2A and EPM2B, encoding respectively laforin, a dual-specificity protein phosphatase, and malin, an E3 ubiquitin ligase. Previously, we and others have suggested that the interactions between laforin and PTG (a regulatory subunit of type 1 protein phosphatase) and between laforin and malin are critical in the pathogenesis of LD. Here, we show that the laforin-malin complex downregulates PTG-induced glycogen synthesis in FTO2B hepatoma cells through a mechanism involving ubiquitination and degradation of PTG. Furthermore, we demonstrate that the interaction between laforin and malin is a regulated process that is modulated by the AMP-activated protein kinase (AMPK). These findings provide further insights into the critical role of the laforin-malin complex in the control of glycogen metabolism and unravel a novel link between the energy sensor AMPK and glycogen metabolism. These data advance our understanding of the functional role of laforin and malin, which hopefully will facilitate the development of appropriate LD therapies.


Assuntos
Proteínas de Transporte/genética , Glicogênio/biossíntese , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Quinases Ativadas por AMP , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Transporte/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Fenômenos Fisiológicos Celulares , Escherichia coli/genética , Glicogênio/análise , Proteínas de Fluorescência Verde/metabolismo , Humanos , Rim/citologia , Modelos Biológicos , Dados de Sequência Molecular , Mutação , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Tirosina Fosfatases não Receptoras/química , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Estatística como Assunto , Transfecção , Técnicas do Sistema de Duplo-Híbrido , Ubiquitina-Proteína Ligases , Ubiquitinação
10.
Cell Mol Life Sci ; 66(15): 2427-43, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19399586

RESUMO

In higher organisms, dietary proteins are broken down into amino acids within the digestive tract but outside the cells, which incorporate the resulting amino acids into their metabolism. However, under certain conditions, an organism loses more nitrogen than is assimilated in the diet. This additional loss was found in the past century to come from intracellular proteins and started an intensive research that produced an enormous expansion of the field and a dispersed literature. Therefore, our purpose is to provide an updated summary of the current knowledge on the proteolytic machinery involved in intracellular protein degradation and its physiological and pathological relevance, especially addressed to newcomers in the field who may find further details in more specialized reviews. However, even providing a general overview, this is an extremely wide field and, therefore, we mainly focus on mammalian cells, while other cells will be mentioned only for comparison purposes.


Assuntos
Proteínas/metabolismo , Aminoácidos/metabolismo , Animais , Autofagia/fisiologia , Humanos , Lisossomos/metabolismo , Fagossomos/metabolismo , Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo
11.
Mol Neurobiol ; 57(3): 1607-1621, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31808062

RESUMO

Lafora disease (LD) is a rare, fatal form of progressive myoclonus epilepsy. The molecular basis of this devastating disease is still poorly understood, and no treatment is available yet, which leads to the death of the patients around 10 years from the onset of the first symptoms. The hallmark of LD is the accumulation of insoluble glycogen-like inclusions in the brain and peripheral tissues, as a consequence of altered glycogen homeostasis. In addition, other determinants in the pathophysiology of LD have been suggested, such as proteostasis impairment, with reduction in autophagy, and oxidative stress, among others. In order to gain a general view of the genes involved in the pathophysiology of LD, in this work, we have performed RNA-Seq transcriptome analyses of whole-brain tissue from two independent mouse models of the disease, namely Epm2a-/- and Epm2b-/- mice, at different times of age. Our results provide strong evidence for three major facts: first, in both models of LD, we found a common set of upregulated genes, most of them encoding mediators of inflammatory response; second, there was a progression with the age in the appearance of these inflammatory markers, starting at 3 months of age; and third, reactive glia was responsible for the expression of these inflammatory genes. These results clearly indicate that neuroinflammation is one of the most important traits to be considered in order to fully understand the pathophysiology of LD, and define reactive glia as novel therapeutic targets in the disease.


Assuntos
Fatores Etários , Doença de Lafora/metabolismo , Epilepsias Mioclônicas Progressivas/metabolismo , Neuroglia/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Animais , Modelos Animais de Doenças , Progressão da Doença , Glicogênio/metabolismo , Corpos de Inclusão/metabolismo , Doença de Lafora/genética , Camundongos Knockout , Epilepsias Mioclônicas Progressivas/genética , Estresse Oxidativo/fisiologia
12.
Biochem Biophys Res Commun ; 369(3): 964-8, 2008 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-18328803

RESUMO

In this work, we have examined the possible role of AMP-activated protein kinase (a key energy sensor) in regulating intracellular protein degradation. We have found that AICAR, a known activator of AMPK, has a dual effect. On one hand, it inhibits autophagy by a mechanism independent of AMPK activity; AICAR decreases class III PI3-kinase binding to beclin-1 and this effect counteracts and reverses the known positive effect of AMPK activity on autophagy. On the other hand, AICAR inhibits the proteasomal degradation of proteins by an AMPK-dependent mechanism. This is a novel function of AMPK that allows the regulation of proteasomal activity under conditions of energy demand.


Assuntos
Autofagia , Complexos Multienzimáticos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Ativadas por AMP , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/efeitos dos fármacos , Proteína Beclina-1 , Células Cultivadas , Regulação para Baixo , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Humanos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteassoma , Ribonucleosídeos/farmacologia , Vacúolos/efeitos dos fármacos , Vacúolos/enzimologia
13.
FEBS J ; 285(11): 2071-2090, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29645350

RESUMO

Lafora disease (LD) is a fatal neurodegenerative disorder caused mostly by mutations in either of two genes encoding laforin and malin. LD is characterized by accumulation of a poorly branched form of glycogen in the cytoplasm of neurons and other cells. We previously reported dysfunctional mitochondria in different LD models. Now, using mitochondrial uncouplers and respiratory chain inhibitors, we have investigated with human fibroblasts a possible alteration in the selective degradation of damaged mitochondria (mitophagy) in LD. By flow cytometry of MitoTracker-labelled cells and measuring the levels of various mitochondrial proteins by western blot, we found in LD fibroblasts a partial impairment in the increased mitochondrial degradation produced by these treatments. In addition, colocalization of mitochondrial and lysosomal markers decreased in LD fibroblasts. All these results are consistent with a partial impairment in the induced autophagic degradation of dysfunctional mitochondria in LD fibroblasts. However, canonical recruitment of Parkin to mitochondria under these conditions remained unaffected in LD fibroblasts, and also in SH-SY5Y cells after malin and laforin overexpression. Neither mitochondrial localization nor protein levels of Bcl-2-like protein 13, another component of the mitophagic machinery that operates under these conditions, were affected in LD fibroblasts. In contrast, although these treatments raised autophagy in both control and LD fibroblasts, this enhanced autophagy was clearly lower in the latter cells. Therefore, the autophagic degradation of altered mitochondria is impaired in LD, which is due to a partial defect in the autophagic response and not in the canonical mitophagy signalling pathways.


Assuntos
Autofagia/genética , Doença de Lafora/genética , Mitocôndrias/genética , Mitofagia/genética , Proteínas de Transporte/genética , Fibroblastos/metabolismo , Fibroblastos/patologia , Citometria de Fluxo , Regulação da Expressão Gênica/genética , Humanos , Doença de Lafora/metabolismo , Doença de Lafora/patologia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Tirosina Fosfatases não Receptoras/genética , Ubiquitina-Proteína Ligases
14.
Biochim Biophys Acta Mol Basis Dis ; 1864(10): 3234-3246, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30006152

RESUMO

Circulating histones have been proposed as targets for therapy in sepsis and hyperinflammatory symptoms. However, the proposed strategies have failed in clinical trials. Although different mechanisms for histone-related cytotoxicity are being explored, those mediated by circulating histones are not fully understood. Extracellular histones induce endothelial cell death, thereby contributing to the pathogenesis of complex diseases such as sepsis and septic shock. Therefore, the comprehension of cellular responses triggered by histones is capital to design effective therapeutic strategies. Here we report how extracellular histones induce autophagy and apoptosis in a dose-dependent manner in cultured human endothelial cells. In addition, we describe how histones regulate these pathways via Sestrin2/AMPK/ULK1-mTOR and AKT/mTOR. Furthermore, we evaluate the effect of Toll-like receptors in mediating autophagy and apoptosis demonstrating how TLR inhibitors do not prevent apoptosis and/or autophagy induced by histones. Our results confirm that histones and autophagic pathways can be considered as novel targets to design therapeutic strategies in endothelial damage.


Assuntos
Histonas/farmacologia , Células Endoteliais da Veia Umbilical Humana/citologia , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
15.
FEBS Lett ; 581(18): 3415-21, 2007 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-17610878

RESUMO

Intracellular protein degradation is a regulated process with several proteolytic pathways. Although regulation of macroautophagy has been investigated in some detail in hepatocytes and in few other cells, less is known on this regulation in other cells and proteolytic pathways. We show that in human fibroblasts insulin and amino acids reduce protein degradation by different signalling pathways and that this inhibition proceeds in part via the mammalian target of rapamycin, especially with amino acids, which probably increase lysosomal pH. Moreover, the regulatory amino acids (Phe, Arg, Met, Tyr, Trp and Cys) are partially different from other cells. Finally, and in addition to macroautophagy, insulin and amino acids modify, to different extents and sometimes in opposite directions, the activities of other proteolytic pathways.


Assuntos
Aminoácidos/farmacologia , Insulina/farmacologia , Peptídeo Hidrolases/metabolismo , Células Cultivadas , Fibroblastos , Humanos , Microscopia Eletrônica , Proteínas Quinases/metabolismo , Serina-Treonina Quinases TOR
16.
Ann Transplant ; 22: 285-295, 2017 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-28484204

RESUMO

BACKGROUND Cardioplegic arrest is a common procedure for many types of cardiac surgery, and different formulations have been proposed to enhance its cardio-protective effect. Hydrogen sulfide is an important signaling molecule that has cardio-protective properties. We therefore studied the cardio-protective effect of hydrogen sulfide in cardiac cell culture and its potential therapeutic use in combination with cardioplegia formulations. MATERIAL AND METHODS We added hydrogen sulfide donor GYY4137 to HL-1 cells to study its protective effect in nutrient starved conditions. In addition, we tested the potential use of GYY4137 when it is added into two different cardioplegia formulations: Cardi-Braun® solution and del Nido solution in an ex vivo Langendorff perfused rat hearts model. RESULTS We observed that eight-hour pre-treatment with GYY4137 significantly suppressed apoptosis in nutrient-starved HL-1 cells (28% less compared to untreated cells; p<0.05), maintained ATP content, and reduced protein synthesis. In ex vivo experiments, Cardi-Braun® and del Nido cardioplegia solutions supplemented with GYY4137 significantly reduced the pro-apoptotic protein caspase-3 content and preserved ATP content. Furthermore, GYY4137 supplemented cardioplegia solutions decreased the S-(5-adenosyl)-L-methionine/S-(adenosyl)-L-homocysteine ratio, reducing the oxidative stress in cardiac tissue. Finally, heart beating analysis revealed the preservation of the inter-beat interval and the heart rate in del Nido cardioplegia solution supplemented with GYY4137. CONCLUSIONS GYY4137 preconditioning preserved energetic state during starved conditions, attenuating the cardiomyocytes apoptosis in vitro. The addition of GYY4137 to cardioplegia solutions prevented apoptosis, ATP consumption, and oxidative stress in perfused rat hearts, restoring its electrophysiological status after cardiac arrest. These findings suggested that GYY4137 sulfide donor may improve the cardioplegia solution performance during cardiac surgery.


Assuntos
Apoptose/efeitos dos fármacos , Parada Cardíaca/metabolismo , Coração/efeitos dos fármacos , Sulfeto de Hidrogênio/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Animais , Soluções Cardioplégicas , Caspase 3/metabolismo , Linhagem Celular , Células Cultivadas , Masculino , Miócitos Cardíacos/metabolismo , Ratos , Ratos Wistar
17.
Front Mol Biosci ; 3: 81, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28018906

RESUMO

Distal hereditary motor neuropathies (dHMN) are a group of rare hereditary neuromuscular disorders characterized by an atrophy that affects peroneal muscles in the absence of sensory symptoms. To date, 23 genes are thought to be responsible for dHMN, four of which encode chaperones: DNAJB2, which encodes a member of the HSP40/DNAJ co-chaperone family; and HSPB1, HSPB3, and HSPB8, encoding three members of the small heat shock protein family. While around 30 different mutations in HSPB1 have been identified, the remaining three genes are altered in many fewer cases. Indeed, a mutation of HSPB3 has only been described in one case, whereas a few cases have been reported carrying mutations in DNAJB2 and HSPB8, most of them caused by a founder c.352+1G>A mutation in DNAJB2 and by mutations affecting the K141 residue in the HSPB8 chaperone. Hence, their rare occurrence makes it difficult to understand the pathological mechanisms driven by such mutations in this neuropathy. Chaperones can assemble into multi-chaperone complexes that form an integrated chaperone network within the cell. Such complexes fulfill relevant roles in a variety of processes, such as the correct folding of newly synthesized proteins, in which chaperones escort them to precise cellular locations, and as a response to protein misfolding, which includes the degradation of proteins that fail to refold properly. Despite this range of functions, mutations in some of these chaperones lead to diseases with a similar clinical profile, suggesting common pathways. This review provides an overview of the genetics of those dHMNs that share a common disease mechanism and that are caused by mutations in four genes encoding chaperones: DNAJB2, HSPB1, HSPB3, and HSPB8.

18.
Methods Mol Biol ; 1449: 299-311, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27613045

RESUMO

Lysosomes participate within the cells in the degradation of organelles, macromolecules, and a wide variety of substrates. In any study on specific roles of lysosomes, both under physiological and pathological conditions, it is advisable to include methods that allow their reproducible and reliable isolation. However, purification of lysosomes is a difficult task, particularly in the case of cultured cells. This is mainly because of the heterogeneity of these organelles, along with their low number and high fragility. Also, isolation methods, while disrupting plasma membranes, have to preserve the integrity of lysosomes, as the breakdown of their membranes releases enzymes that could damage all cell organelles, including themselves. The protocols described below have been routinely used in our laboratory for the specific isolation of lysosomes from rat liver, NIH/3T3, and other cultured cells, but can be adapted to other mammalian tissues or cell lines.


Assuntos
Fracionamento Celular/métodos , Organelas/metabolismo , Animais , Linhagem Celular , Membrana Celular , Células Cultivadas , Centrifugação com Gradiente de Concentração/métodos , Fígado/citologia , Lisossomos , Camundongos , Ratos , Frações Subcelulares/metabolismo
19.
Free Radic Biol Med ; 88(Pt A): 30-41, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25680286

RESUMO

Lafora disease (LD; OMIM 254780, ORPHA501) is a devastating neurodegenerative disorder characterized by the presence of glycogen-like intracellular inclusions called Lafora bodies and caused, in most cases, by mutations in either the EPM2A or the EPM2B gene, encoding respectively laforin, a phosphatase with dual specificity that is involved in the dephosphorylation of glycogen, and malin, an E3-ubiquitin ligase involved in the polyubiquitination of proteins related to glycogen metabolism. Thus, it has been reported that laforin and malin form a functional complex that acts as a key regulator of glycogen metabolism and that also plays a crucial role in protein homeostasis (proteostasis). Regarding this last function, it has been shown that cells are more sensitive to ER stress and show defects in proteasome and autophagy activities in the absence of a functional laforin-malin complex. More recently, we have demonstrated that oxidative stress accompanies these proteostasis defects and that various LD models show an increase in reactive oxygen species and oxidative stress products together with a dysregulated antioxidant enzyme expression and activity. In this review we discuss possible connections between the multiple defects in protein homeostasis present in LD and oxidative stress.


Assuntos
Doença de Lafora/etiologia , Estresse Oxidativo , Deficiências na Proteostase/metabolismo , Antioxidantes/metabolismo , Proteínas de Transporte/genética , Glicogênio/metabolismo , Humanos , Doença de Lafora/metabolismo , Doença de Lafora/fisiopatologia , Mutação , Proteínas Tirosina Fosfatases não Receptoras/genética , Ubiquitina-Proteína Ligases
20.
PLoS One ; 10(12): e0144273, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26642043

RESUMO

GTPBP3 is an evolutionary conserved protein presumably involved in mitochondrial tRNA (mt-tRNA) modification. In humans, GTPBP3 mutations cause hypertrophic cardiomyopathy with lactic acidosis, and have been associated with a defect in mitochondrial translation, yet the pathomechanism remains unclear. Here we use a GTPBP3 stable-silencing model (shGTPBP3 cells) for a further characterization of the phenotype conferred by the GTPBP3 defect. We experimentally show for the first time that GTPBP3 depletion is associated with an mt-tRNA hypomodification status, as mt-tRNAs from shGTPBP3 cells were more sensitive to digestion by angiogenin than tRNAs from control cells. Despite the effect of stable silencing of GTPBP3 on global mitochondrial translation being rather mild, the steady-state levels and activity of Complex I, and cellular ATP levels were 50% of those found in the controls. Notably, the ATPase activity of Complex V increased by about 40% in GTPBP3 depleted cells suggesting that mitochondria consume ATP to maintain the membrane potential. Moreover, shGTPBP3 cells exhibited enhanced antioxidant capacity and a nearly 2-fold increase in the uncoupling protein UCP2 levels. Our data indicate that stable silencing of GTPBP3 triggers an AMPK-dependent retrograde signaling pathway that down-regulates the expression of the NDUFAF3 and NDUFAF4 Complex I assembly factors and the mitochondrial pyruvate carrier (MPC), while up-regulating the expression of UCP2. We also found that genes involved in glycolysis and oxidation of fatty acids are up-regulated. These data are compatible with a model in which high UCP2 levels, together with a reduction in pyruvate transport due to the down-regulation of MPC, promote a shift from pyruvate to fatty acid oxidation, and to an uncoupling of glycolysis and oxidative phosphorylation. These metabolic alterations, and the low ATP levels, may negatively affect heart function.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas de Transporte de Ânions/metabolismo , Proteínas de Ligação ao GTP/genética , Canais Iônicos/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Trifosfato de Adenosina/metabolismo , Proteínas de Transporte de Ânions/genética , Proteínas de Ligação a Calmodulina/genética , Proteínas de Ligação a Calmodulina/metabolismo , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Escherichia coli/genética , Ácidos Graxos/genética , Ácidos Graxos/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Glicólise/genética , Células HEK293 , Humanos , Canais Iônicos/genética , Mitocôndrias/genética , Proteínas de Transporte da Membrana Mitocondrial , Proteínas Mitocondriais/genética , Transportadores de Ácidos Monocarboxílicos , Fosforilação Oxidativa , RNA de Transferência de Lisina/metabolismo , Ribonuclease Pancreático/química , Ribonuclease Pancreático/metabolismo , Proteína Desacopladora 2
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa