Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 15(3): 203, 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38467609

RESUMEN

Organelles form membrane contact sites between each other, allowing for the transfer of molecules and signals. Mitochondria-endoplasmic reticulum (ER) contact sites (MERCS) are cellular subdomains characterized by close apposition of mitochondria and ER membranes. They have been implicated in many diseases, including neurodegenerative, metabolic, and cardiac diseases. Although MERCS have been extensively studied, much remains to be explored. To uncover novel regulators of MERCS, we conducted a genome-wide, flow cytometry-based screen using an engineered MERCS reporter cell line. We found 410 genes whose downregulation promotes MERCS and 230 genes whose downregulation decreases MERCS. From these, 29 genes were selected from each population for arrayed screening and 25 were validated from the high population and 13 from the low population. GET4 and BAG6 were highlighted as the top 2 genes that upon suppression increased MERCS from both the pooled and arrayed screens, and these were subjected to further investigation. Multiple microscopy analyses confirmed that loss of GET4 or BAG6 increased MERCS. GET4 and BAG6 were also observed to interact with the known MERCS proteins, inositol 1,4,5-trisphosphate receptors (IP3R) and glucose-regulated protein 75 (GRP75). In addition, we found that loss of GET4 increased mitochondrial calcium uptake upon ER-Ca2+ release and mitochondrial respiration. Finally, we show that loss of GET4 rescues motor ability, improves lifespan and prevents neurodegeneration in a Drosophila model of Alzheimer's disease (Aß42Arc). Together, these results suggest that GET4 is involved in decreasing MERCS and that its loss is neuroprotective.


Asunto(s)
Sistemas CRISPR-Cas , Membranas Asociadas a Mitocondrias , Sistemas CRISPR-Cas/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Calcio/metabolismo
2.
Sci Rep ; 13(1): 20616, 2023 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-37996473

RESUMEN

Biological pathways between alcohol consumption and alcohol liver disease (ALD) are not fully understood. We selected genes with known effect on (1) alcohol consumption, (2) liver function, and (3) gene expression. Expression of the orthologs of these genes in Caenorhabditis elegans and Drosophila melanogaster was suppressed using mutations and/or RNA interference (RNAi). In humans, association analysis, pathway analysis, and Mendelian randomization analysis were performed to identify metabolic changes due to alcohol consumption. In C. elegans, we found a reduction in locomotion rate after exposure to ethanol for RNAi knockdown of ACTR1B and MAPT. In Drosophila, we observed (1) a change in sedative effect of ethanol for RNAi knockdown of WDPCP, TENM2, GPN1, ARPC1B, and SCN8A, (2) a reduction in ethanol consumption for RNAi knockdown of TENM2, (3) a reduction in triradylglycerols (TAG) levels for RNAi knockdown of WDPCP, TENM2, and GPN1. In human, we observed (1) a link between alcohol consumption and several metabolites including TAG, (2) an enrichment of the candidate (alcohol-associated) metabolites within the linoleic acid (LNA) and alpha-linolenic acid (ALA) metabolism pathways, (3) a causal link between gene expression of WDPCP to liver fibrosis and liver cirrhosis. Our results imply that WDPCP might be involved in ALD.


Asunto(s)
Caenorhabditis elegans , Drosophila melanogaster , Metabolismo de los Lípidos , Hepatopatías Alcohólicas , Animales , Humanos , Consumo de Bebidas Alcohólicas/genética , Caenorhabditis elegans/genética , Drosophila melanogaster/genética , Etanol/metabolismo , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Cirrosis Hepática/patología , Hepatopatías Alcohólicas/metabolismo
3.
Dis Model Mech ; 16(4)2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-37083954

RESUMEN

Eukaryotic Tribbles proteins are pseudoenzymes that regulate multiple aspects of intracellular signalling. Both Drosophila melanogaster and mammalian members of this family of pseudokinases act as negative regulators of insulin signalling. Mammalian tribbles pseudokinase (TRIB) genes have also been linked to insulin resistance and type 2 diabetes mellitus. Type 2 diabetes mellitus is associated with increased body weight, sleep problems and increased long-term mortality. Here, we investigated how manipulating the expression of Tribbles impacts body weight, sleep and mortality. We showed that the overexpression of Drosophila tribbles (trbl) in the fly fat body reduces both body weight and lifespan in adult flies without affecting food intake. Furthermore, it decreases the levels of Drosophila insulin-like peptide 2 (DILP2; ILP2) and increases night-time sleep. The three genes encoding TRIBs of mammals, TRIB1, TRIB2 and TRIB3, show both common and unique features. As the three human TRIB genes share features with Drosophila trbl, we further explored the links between TRIB genetic variants and both body weight and sleep in the human population. We identified associations between the polymorphisms and expression levels of the pseudokinases and markers of body weight and sleep duration. We conclude that Tribbles pseudokinases are involved in the control of body weight, lifespan and sleep.


Asunto(s)
Diabetes Mellitus Tipo 2 , Neuropéptidos , Animales , Humanos , Peso Corporal , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas de Ciclo Celular/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mamíferos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Duración del Sueño , Regulación hacia Arriba/genética
4.
Dis Model Mech ; 16(1)2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36695500

RESUMEN

Parkinson's disease (PD), an age-dependent neurodegenerative disease, is characterised by the selective loss of dopaminergic neurons in the substantia nigra (SN). Mitochondrial dysfunction is a hallmark of PD, and mutations in PINK1, a gene necessary for mitochondrial fitness, cause PD. Drosophila melanogaster flies with pink1 mutations exhibit mitochondrial defects and dopaminergic cell loss and are used as a PD model. To gain an integrated view of the cellular changes caused by defects in the PINK1 pathway of mitochondrial quality control, we combined metabolomics and transcriptomics analysis in pink1-mutant flies with human induced pluripotent stem cell (iPSC)-derived neural precursor cells (NPCs) with a PINK1 mutation. We observed alterations in cysteine metabolism in both the fly and human PD models. Mitochondrial dysfunction in the NPCs resulted in changes in several metabolites that are linked to cysteine synthesis and increased glutathione levels. We conclude that alterations in cysteine metabolism may compensate for increased oxidative stress in PD, revealing a unifying mechanism of early-stage PD pathology that may be targeted for drug development. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Proteínas de Drosophila , Células Madre Pluripotentes Inducidas , Células-Madre Neurales , Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Animales , Humanos , Drosophila melanogaster/metabolismo , Cisteína , Enfermedad de Parkinson/metabolismo , Células-Madre Neurales/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas Quinasas/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Serina-Treonina Quinasas/genética
5.
Geroscience ; 44(4): 1961-1974, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35355221

RESUMEN

Mitochondrial reactive oxygen species (mtROS) are cellular messengers essential for cellular homeostasis. In response to stress, reverse electron transport (RET) through respiratory complex I generates high levels of mtROS. Suppression of ROS production via RET (ROS-RET) reduces survival under stress, while activation of ROS-RET extends lifespan in basal conditions. Here, we demonstrate that ROS-RET signalling requires increased electron entry and uninterrupted electron flow through the electron transport chain (ETC). We find that in old fruit flies, ROS-RET is abolished when electron flux is decreased and that their mitochondria produce consistently high levels of mtROS. Finally, we demonstrate that in young flies, limiting electron exit, but not entry, from the ETC phenocopies mtROS generation observed in old individuals. Our results elucidate the mechanism by which ROS signalling is lost during ageing.


Asunto(s)
Dípteros , Electrones , Animales , Especies Reactivas de Oxígeno , Transporte de Electrón , Envejecimiento
6.
Nat Aging ; 2(4): 317-331, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-37117744

RESUMEN

The innate immune response mounts a defense against foreign invaders and declines with age. An inappropriate induction of this response can cause diseases. Previous studies showed that mitochondria can be repurposed to promote inflammatory signaling. Damaged mitochondria can also trigger inflammation and promote diseases. Mutations in pink1, a gene required for mitochondrial health, cause Parkinson's disease, and Drosophila melanogaster pink1 mutants accumulate damaged mitochondria. Here, we show that defective mitochondria in pink1 mutants activate Relish targets and demonstrate that inflammatory signaling causes age-dependent intestinal dysfunction in pink1-mutant flies. These effects result in the death of intestinal cells, metabolic reprogramming and neurotoxicity. We found that Relish signaling is activated downstream of a pathway stimulated by cytosolic DNA. Suppression of Relish in the intestinal midgut of pink1-mutant flies restores mitochondrial function and is neuroprotective. We thus conclude that gut-brain communication modulates neurotoxicity in a fly model of Parkinson's disease through a mechanism involving mitochondrial dysfunction.


Asunto(s)
Proteínas de Drosophila , Enfermedades Gastrointestinales , Enfermedades Intestinales , Enfermedad de Parkinson , Animales , Drosophila/metabolismo , Drosophila melanogaster/genética , Proteínas de Drosophila/genética , Proteínas Serina-Treonina Quinasas/genética , Enfermedad de Parkinson/genética
8.
Cell Death Dis ; 12(7): 651, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34172715

RESUMEN

Alzheimer's disease is the most common age-related neurodegenerative disorder. Familial forms of Alzheimer's disease associated with the accumulation of a toxic form of amyloid-ß (Aß) peptides are linked to mitochondrial impairment. The coenzyme nicotinamide adenine dinucleotide (NAD+) is essential for both mitochondrial bioenergetics and nuclear DNA repair through NAD+-consuming poly (ADP-ribose) polymerases (PARPs). Here we analysed the metabolomic changes in flies overexpressing Aß and showed a decrease of metabolites associated with nicotinate and nicotinamide metabolism, which is critical for mitochondrial function in neurons. We show that increasing the bioavailability of NAD+ protects against Aß toxicity. Pharmacological supplementation using NAM, a form of vitamin B that acts as a precursor for NAD+ or a genetic mutation of PARP rescues mitochondrial defects, protects neurons against degeneration and reduces behavioural impairments in a fly model of Alzheimer's disease. Next, we looked at links between PARP polymorphisms and vitamin B intake in patients with Alzheimer's disease. We show that polymorphisms in the human PARP1 gene or the intake of vitamin B are associated with a decrease in the risk and severity of Alzheimer's disease. We suggest that enhancing the availability of NAD+ by either vitamin B supplements or the inhibition of NAD+-dependent enzymes such as PARPs are potential therapies for Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/genética , Proteínas de Drosophila/genética , Mitocondrias/genética , Mutación , NAD/metabolismo , Neuronas/enzimología , Poli(ADP-Ribosa) Polimerasa-1/genética , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Animales Modificados Genéticamente , Conducta Animal , Modelos Animales de Enfermedad , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Humanos , Metaboloma , Metabolómica , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/ultraestructura , Actividad Motora , Degeneración Nerviosa , Neuronas/efectos de los fármacos , Neuronas/patología , Niacinamida/farmacología , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Polimorfismo de Nucleótido Simple
9.
Int J Mol Sci ; 22(9)2021 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-33925631

RESUMEN

In Drosophila, endoplasmic reticulum (ER) stress activates the protein kinase R-like endoplasmic reticulum kinase (dPerk). dPerk can also be activated by defective mitochondria in fly models of Parkinson's disease caused by mutations in pink1 or parkin. The Perk branch of the unfolded protein response (UPR) has emerged as a major toxic process in neurodegenerative disorders causing a chronic reduction in vital proteins and neuronal death. In this study, we combined microarray analysis and quantitative proteomics analysis in adult flies overexpressing dPerk to investigate the relationship between the transcriptional and translational response to dPerk activation. We identified tribbles and Heat shock protein 22 as two novel Drosophila activating transcription factor 4 (dAtf4) regulated transcripts. Using a combined bioinformatics tool kit, we demonstrated that the activation of dPerk leads to translational repression of mitochondrial proteins associated with glutathione and nucleotide metabolism, calcium signalling and iron-sulphur cluster biosynthesis. Further efforts to enhance these translationally repressed dPerk targets might offer protection against Perk toxicity.


Asunto(s)
eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo , Factor de Transcripción Activador 4/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Biología Computacional/métodos , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Retículo Endoplásmico/patología , Estrés del Retículo Endoplásmico , Factor 2 Eucariótico de Iniciación/metabolismo , Proteínas de Choque Térmico/metabolismo , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Proteómica/métodos , Transducción de Señal , Factores de Transcripción/metabolismo , Transcriptoma , Ubiquitina-Proteína Ligasas/metabolismo , Respuesta de Proteína Desplegada/genética , Respuesta de Proteína Desplegada/fisiología
10.
Redox Biol ; 32: 101450, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32146156

RESUMEN

Reactive Oxygen Species (ROS) are essential cellular messengers required for cellular homeostasis and regulate the lifespan of several animal species. The main site of ROS production is the mitochondrion, and within it, respiratory complex I (CI) is the main ROS generator. ROS produced by CI trigger several physiological responses that are essential for the survival of neurons, cardiomyocytes and macrophages. Here, we show that CI produces ROS when electrons flow in either the forward (Forward Electron Transport, FET) or reverse direction (Reverse Electron Transport, RET). We demonstrate that ROS production via RET (ROS-RET) is activated under thermal stress conditions and that interruption of ROS-RET production, through ectopic expression of the alternative oxidase AOX, attenuates the activation of pro-survival pathways in response to stress. Accordingly, we find that both suppressing ROS-RET signalling or decreasing levels of mitochondrial H2O2 by overexpressing mitochondrial catalase (mtCAT), reduces survival dramatically in flies under stress. Our results uncover a specific ROS signalling pathway where hydrogen peroxide (H2O2) generated by CI via RET is required to activate adaptive mechanisms, maximising survival under stress conditions.


Asunto(s)
Drosophila melanogaster , Complejo I de Transporte de Electrón , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Transporte de Electrón , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Peróxido de Hidrógeno , Especies Reactivas de Oxígeno/metabolismo
11.
Biol Open ; 9(1)2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31822473

RESUMEN

Eukaryotic cells are complex systems containing internal compartments with specialised functions. Among these compartments, the endoplasmic reticulum (ER) plays a major role in processing proteins for modification and delivery to other organelles, whereas mitochondria generate energy in the form of ATP. Mitochondria and the ER form physical interactions, defined as mitochondria-ER contact sites (MERCs) to exchange metabolites such as calcium ions (Ca2+) and lipids. Sites of contact between mitochondria and the ER can regulate biological processes such as ATP generation and mitochondrial division. The interactions between mitochondria and the ER are dynamic and respond to the metabolic state of cells. Changes in MERCs have been linked to metabolic pathologies such as diabetes, neurodegenerative diseases and sleep disruption. Here we explored the consequences of increasing contacts between mitochondria and the ER in flies using a synthetic linker. We showed that enhancing MERCs increases locomotion and extends lifespan. We also showed that, in a Drosophila model of Alzheimer's disease linked to toxic amyloid beta (Aß), linker expression can suppress motor impairment and extend lifespan. We conclude that strategies for increasing contacts between mitochondria and the ER may improve symptoms of diseases associated with mitochondria dysfunction. A video abstract for this article is available at https://youtu.be/_YWA4oKZkes.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/patología , Animales , Transporte Biológico , Calcio/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Drosophila , Retículo Endoplásmico/ultraestructura , Técnica del Anticuerpo Fluorescente , Expresión Génica , Genes Reporteros , Humanos , Locomoción , Mitocondrias/ultraestructura , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
12.
Cell Death Dis ; 10(4): 288, 2019 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-30911005

RESUMEN

Mutations in the mitochondrial GTPase mitofusin 2 (MFN2) cause Charcot-Marie-Tooth disease type 2 (CMT2A), a form of peripheral neuropathy that compromises axonal function. Mitofusins promote mitochondrial fusion and regulate mitochondrial dynamics. They are also reported to be involved in forming contacts between mitochondria and the endoplasmic reticulum. The fruit fly, Drosophila melanogaster, is a powerful tool to model human neurodegenerative diseases, including CMT2A. Here, we have downregulated the expression of the Drosophila mitofusin (dMfn RNAi) in adult flies and showed that this activates mitochondrial retrograde signalling and is associated with an upregulation of genes involved in folic acid (FA) metabolism. Additionally, we demonstrated that pharmacological and genetic interventions designed to increase the FA metabolism pathway suppresses the phenotype of the dMfn RNAi flies. We conclude that strategies to increase FA metabolism may ameliorate diseases, such as peripheral neuropathies, that are associated with loss of mitochondrial function. A video abstract for this article is available at  https://youtu.be/fs1G-QRo6xI .


Asunto(s)
Regulación hacia Abajo/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Ácido Fólico/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Factor de Transcripción Activador 4/metabolismo , Animales , Transporte Axonal/genética , Enfermedad de Charcot-Marie-Tooth/metabolismo , Modelos Animales de Enfermedad , Ácido Fólico/genética , Locomoción/genética , Masculino , Mitocondrias/metabolismo , Fenotipo , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo
13.
Dis Model Mech ; 12(2)2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30760495

RESUMEN

Lung cancer is the leading cause of cancer-related death. Two-thirds of cases are diagnosed at an advanced stage that is refractory to curative treatment. Therefore, strategies for the early detection of lung cancer are urgently sought. Total circulating free DNA (cfDNA) and tumour-derived circulating tumour DNA (ctDNA) are emerging as important biomarkers within a 'liquid biopsy' for monitoring human disease progression and response to therapy. Owing to the late clinical diagnosis of lung adenocarcinoma, the potential for cfDNA and ctDNA as early detection biomarkers remains unexplored. Here, using a Cre-regulated genetically engineered mouse model of lung adenocarcinoma development, driven by KrasG12D (the KrasLSL-G12D mouse), we serially tracked the release of cfDNA/ctDNA and compared this with tumour burden as determined by micro-computed tomography (CT). To monitor ctDNA, a droplet digital PCR assay was developed to permit discrimination of the KrasLox-G12D allele from the KrasLSL-G12D and KrasWT alleles. We show that micro-CT correlates with endpoint histology and is able to detect pre-malignant tumours with a combined volume larger than 7 mm3 Changes in cfDNA/ctDNA levels correlate with micro-CT measurements in longitudinal sampling and are able to monitor the emergence of lesions before the adenoma-adenocarcinoma transition. Potentially, this work has implications for the early detection of human lung adenocarcinoma using ctDNA/cfDNA profiling.A video abstract for this article is available at https://youtu.be/Ku8xJJyGs3UThis article has an associated First Person interview with the joint first authors of the paper.


Asunto(s)
Ácidos Nucleicos Libres de Células/sangre , Detección Precoz del Cáncer , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/diagnóstico , Mutación/genética , Lesiones Precancerosas/sangre , Lesiones Precancerosas/diagnóstico , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Alelos , Animales , Modelos Animales de Enfermedad , Femenino , Integrasas/metabolismo , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/patología , Masculino , Ratones Endogámicos C57BL , Lesiones Precancerosas/diagnóstico por imagen , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas p21(ras)/sangre , Recombinación Genética/genética , Reproducibilidad de los Resultados , Carga Tumoral , Microtomografía por Rayos X
14.
Cell Death Differ ; 26(9): 1861, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30546072

RESUMEN

Following publication of the article, Dr. Roberta Tufi of the Mitochondrial Biology Unit at the University of Cambridge was concerned to note that her own contribution to the study during her postdoc in Leicester at the MRC Toxicology Unit had not been acknowledged. Specifically, the data in Fig. 1 (panels a, b, and d) were produced though her work.

15.
Cell Death Dis ; 8(10): e3119, 2017 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-29022916

RESUMEN

HtrA2 (high-temperature requirement 2) is a human mitochondrial protease that has a role in apoptosis and Parkinson's disease. The structure of HtrA2 with an intact catalytic triad was determined, revealing a conformational change in the active site loops, involving mainly the regulatory LD loop, which resulted in burial of the catalytic serine relative to the previously reported structure of the proteolytically inactive mutant. Mutations in the loops surrounding the active site that significantly restricted their mobility, reduced proteolytic activity both in vitro and in cells, suggesting that regulation of HtrA2 activity cannot be explained by a simple transition to an activated conformational state with enhanced active site accessibility. Manipulation of solvent viscosity highlighted an unusual bi-phasic behavior of the enzymatic activity, which together with MD calculations supports the importance of motion in the regulation of the activity of HtrA2. HtrA2 is an unusually thermostable enzyme (TM=97.3 °C), a trait often associated with structural rigidity, not dynamic motion. We suggest that this thermostability functions to provide a stable scaffold for the observed loop motions, allowing them a relatively free conformational search within a rather restricted volume.


Asunto(s)
Dominio Catalítico/fisiología , Serina Peptidasa A2 que Requiere Temperaturas Altas/genética , Serina Peptidasa A2 que Requiere Temperaturas Altas/metabolismo , Termodinámica , Línea Celular , Clonación Molecular , Eliminación de Gen , Humanos , Mitocondrias/metabolismo , Modelos Moleculares , Simulación de Dinámica Molecular , Estructura Secundaria de Proteína , Viscosidad
16.
Brain ; 140(9): 2444-2459, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-29050400

RESUMEN

The mitochondrial proteins TRAP1 and HTRA2 have previously been shown to be phosphorylated in the presence of the Parkinson's disease kinase PINK1 but the downstream signalling is unknown. HTRA2 and PINK1 loss of function causes parkinsonism in humans and animals. Here, we identified TRAP1 as an interactor of HTRA2 using an unbiased mass spectrometry approach. In our human cell models, TRAP1 overexpression is protective, rescuing HTRA2 and PINK1-associated mitochondrial dysfunction and suggesting that TRAP1 acts downstream of HTRA2 and PINK1. HTRA2 regulates TRAP1 protein levels, but TRAP1 is not a direct target of HTRA2 protease activity. Following genetic screening of Parkinson's disease patients and healthy controls, we also report the first TRAP1 mutation leading to complete loss of functional protein in a patient with late onset Parkinson's disease. Analysis of fibroblasts derived from the patient reveal that oxygen consumption, ATP output and reactive oxygen species are increased compared to healthy individuals. This is coupled with an increased pool of free NADH, increased mitochondrial biogenesis, triggering of the mitochondrial unfolded protein response, loss of mitochondrial membrane potential and sensitivity to mitochondrial removal and apoptosis. These data highlight the role of TRAP1 in the regulation of energy metabolism and mitochondrial quality control. Interestingly, the diabetes drug metformin reverses mutation-associated alterations on energy metabolism, mitochondrial biogenesis and restores mitochondrial membrane potential. In summary, our data show that TRAP1 acts downstream of PINK1 and HTRA2 for mitochondrial fine tuning, whereas TRAP1 loss of function leads to reduced control of energy metabolism, ultimately impacting mitochondrial membrane potential. These findings offer new insight into mitochondrial pathologies in Parkinson's disease and provide new prospects for targeted therapies.


Asunto(s)
Proteínas HSP90 de Choque Térmico/genética , Metformina/uso terapéutico , Mitocondrias/efectos de los fármacos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Adenosina Trifosfato/metabolismo , Apoptosis/efectos de los fármacos , Estudios de Casos y Controles , Células Cultivadas , Fibroblastos/metabolismo , Proteínas HSP90 de Choque Térmico/biosíntesis , Serina Peptidasa A2 que Requiere Temperaturas Altas , Humanos , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Mutación , NAD/metabolismo , Biogénesis de Organelos , Consumo de Oxígeno , Enfermedad de Parkinson/genética , Proteínas Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Serina Endopeptidasas/metabolismo
18.
Cell Rep ; 19(9): 1739-1749, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28564594

RESUMEN

Motor neurons (MNs) and astrocytes (ACs) are implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS), but their interaction and the sequence of molecular events leading to MN death remain unresolved. Here, we optimized directed differentiation of induced pluripotent stem cells (iPSCs) into highly enriched (> 85%) functional populations of spinal cord MNs and ACs. We identify significantly increased cytoplasmic TDP-43 and ER stress as primary pathogenic events in patient-specific valosin-containing protein (VCP)-mutant MNs, with secondary mitochondrial dysfunction and oxidative stress. Cumulatively, these cellular stresses result in synaptic pathology and cell death in VCP-mutant MNs. We additionally identify a cell-autonomous VCP-mutant AC survival phenotype, which is not attributable to the same molecular pathology occurring in VCP-mutant MNs. Finally, through iterative co-culture experiments, we uncover non-cell-autonomous effects of VCP-mutant ACs on both control and mutant MNs. This work elucidates molecular events and cellular interplay that could guide future therapeutic strategies in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Astrocitos/patología , Modelos Biológicos , Neuronas Motoras/patología , Proteína que Contiene Valosina/metabolismo , Supervivencia Celular , Proteínas de Unión al ADN/metabolismo , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Estrés del Retículo Endoplásmico , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Mutación/genética , Degeneración Nerviosa/patología , Neurogénesis , Estrés Oxidativo , Fenotipo , Sinapsis/patología
19.
Biol Open ; 6(2): 141-147, 2017 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-28011627

RESUMEN

Familial forms of Parkinson's disease (PD) caused by mutations in PINK1 are linked to mitochondrial impairment. Defective mitochondria are also found in Drosophila models of PD with pink1 mutations. The co-enzyme nicotinamide adenine dinucleotide (NAD+) is essential for both generating energy in mitochondria and nuclear DNA repair through NAD+-consuming poly(ADP-ribose) polymerases (PARPs). We found alterations in NAD+ salvage metabolism in Drosophila pink1 mutants and showed that a diet supplemented with the NAD+ precursor nicotinamide rescued mitochondrial defects and protected neurons from degeneration. Additionally, a mutation of Parp improved mitochondrial function and was neuroprotective in the pink1 mutants. We conclude that enhancing the availability of NAD+ by either the use of a diet supplemented with NAD+ precursors or the inhibition of NAD+-dependent enzymes, such as PARPs, which compete with mitochondria for NAD+, is a viable approach to preventing neurotoxicity associated with mitochondrial defects.

20.
Exp Cell Res ; 333(1): 73-9, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25728007

RESUMEN

Mutations in Parkin, an E3 ubiquitin ligase, are associated to autosomal recessive Parkinson's disease (PD). Parkin has been mainly implicated, along with Pink1, in mitochondrial autophagy in response to stress. In this study, a yeast model was developed to analyse the biological function of human Parkin. We observed that Parkin increases yeast chronological lifespan and oxidative stress resistance, through a mitochondrial-dependent pathway. Moreover, in response to H2O2, Parkin translocate to mitochondria, leading to a higher mitochondrial degradation. Parkin-induced H2O2 resistance is dependent on the autophagic pathway and on the mitochondrial protein Por1p. Although expression of Pink1 induces an H2O2 resistance phenotype similar to Parkin, co-expression of both proteins does not result in a synergistic effect. Concerning H2O2 resistance, this may indicate that these two proteins independently affect the same pathway. Altogether, this work establishes a yeast model for Parkin, which may provide new insights on Parkin function and potential mechanisms of pathogenicity.


Asunto(s)
Ubiquitina-Proteína Ligasas/biosíntesis , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Peróxido de Hidrógeno/farmacología , Oxidantes/farmacología , Estrés Oxidativo , Enfermedad de Parkinson/metabolismo , Transporte de Proteínas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Saccharomyces cerevisiae/metabolismo , Ubiquitina-Proteína Ligasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA