Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
EMBO J ; 38(2)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30504269

RESUMEN

The Parkinson's disease-associated protein kinase PINK1 and ubiquitin ligase Parkin coordinate the ubiquitination of mitochondrial proteins, which marks mitochondria for degradation. Miro1, an atypical GTPase involved in mitochondrial trafficking, is one of the substrates tagged by Parkin after mitochondrial damage. Here, we demonstrate that a small pool of Parkin interacts with Miro1 before mitochondrial damage occurs. This interaction does not require PINK1, does not involve ubiquitination of Miro1 and also does not disturb Miro1 function. However, following mitochondrial damage and PINK1 accumulation, this initial pool of Parkin becomes activated, leading to the ubiquitination and degradation of Miro1. Knockdown of Miro proteins reduces Parkin translocation to mitochondria and suppresses mitophagic removal of mitochondria. Moreover, we demonstrate that Miro1 EF-hand domains control Miro1's ubiquitination and Parkin recruitment to damaged mitochondria, and they protect neurons from glutamate-induced mitophagy. Together, our results suggest that Miro1 functions as a calcium-sensitive docking site for Parkin on mitochondria.


Asunto(s)
Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Mitofagia , Dominios Proteicos , Transporte de Proteínas , Proteolisis , Ratas , Ubiquitinación , Proteínas de Unión al GTP rho/química , Proteínas de Unión al GTP rho/genética
2.
Development ; 143(11): 1981-92, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27122166

RESUMEN

During early development, neurons undergo complex morphological rearrangements to assemble into neuronal circuits and propagate signals. Rapid growth requires a large quantity of building materials, efficient intracellular transport and also a considerable amount of energy. To produce this energy, the neuron should first generate new mitochondria because the pre-existing mitochondria are unlikely to provide a sufficient acceleration in ATP production. Here, we demonstrate that mitochondrial biogenesis and ATP production are required for axonal growth and neuronal development in cultured rat cortical neurons. We also demonstrate that growth signals activating the CaMKKß, LKB1-STRAD or TAK1 pathways also co-activate the AMPK-PGC-1α-NRF1 axis leading to the generation of new mitochondria to ensure energy for upcoming growth. In conclusion, our results suggest that neurons are capable of signalling for upcoming energy requirements. Earlier activation of mitochondrial biogenesis through these pathways will accelerate the generation of new mitochondria, thereby ensuring energy-producing capability for when other factors for axonal growth are synthesized.


Asunto(s)
Axones/metabolismo , Biogénesis de Organelos , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Adenilato Quinasa/metabolismo , Animales , Animales Recién Nacidos , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Proliferación Celular , Células Cultivadas , Corteza Cerebral/citología , Metabolismo Energético , Quinasas Quinasa Quinasa PAM/metabolismo , Mitocondrias/metabolismo , Modelos Biológicos , Neurogénesis , Factor Nuclear 1 de Respiración/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Ratas Wistar , Factor de Crecimiento Transformador beta/metabolismo
3.
PLoS Biol ; 14(7): e1002511, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27434582

RESUMEN

Deficiency of the protein Wolfram syndrome 1 (WFS1) is associated with multiple neurological and psychiatric abnormalities similar to those observed in pathologies showing alterations in mitochondrial dynamics. The aim of this study was to examine the hypothesis that WFS1 deficiency affects neuronal function via mitochondrial abnormalities. We show that down-regulation of WFS1 in neurons leads to dramatic changes in mitochondrial dynamics (inhibited mitochondrial fusion, altered mitochondrial trafficking, and augmented mitophagy), delaying neuronal development. WFS1 deficiency induces endoplasmic reticulum (ER) stress, leading to inositol 1,4,5-trisphosphate receptor (IP3R) dysfunction and disturbed cytosolic Ca2+ homeostasis, which, in turn, alters mitochondrial dynamics. Importantly, ER stress, impaired Ca2+ homeostasis, altered mitochondrial dynamics, and delayed neuronal development are causatively related events because interventions at all these levels improved the downstream processes. Our data shed light on the mechanisms of neuronal abnormalities in Wolfram syndrome and point out potential therapeutic targets. This work may have broader implications for understanding the role of mitochondrial dynamics in neuropsychiatric diseases.


Asunto(s)
Mitocondrias/metabolismo , Dinámicas Mitocondriales , Neurogénesis , Neuronas/metabolismo , Animales , Animales Recién Nacidos , Encéfalo/citología , Encéfalo/metabolismo , Calcio/metabolismo , Células Cultivadas , Estrés del Retículo Endoplásmico/genética , Transferencia Resonante de Energía de Fluorescencia , Homeostasis , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Potencial de la Membrana Mitocondrial/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Noqueados , Microscopía Confocal , Mitocondrias/genética , Mitofagia/genética , Neuronas/citología , Células PC12 , Interferencia de ARN , Ratas , Ratas Wistar , Imagen de Lapso de Tiempo/métodos , Síndrome de Wolfram/genética , Síndrome de Wolfram/metabolismo
4.
Cell Mol Life Sci ; 75(24): 4643-4666, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30255402

RESUMEN

The Ca2+-binding protein parvalbumin (PV) and mitochondria play important roles in Ca2+ signaling, buffering and sequestration. Antagonistic regulation of PV and mitochondrial volume is observed in in vitro and in vivo model systems. Changes in mitochondrial morphology, mitochondrial volume and dynamics (fusion, fission, mitophagy) resulting from modulation of PV were investigated in MDCK epithelial cells with stable overexpression/downregulation of PV. Increased PV levels resulted in smaller, roundish cells and shorter mitochondria, the latter phenomenon related to reduced fusion rates and decreased expression of genes involved in mitochondrial fusion. PV-overexpressing cells displayed increased mitophagy, a likely cause for the decreased mitochondrial volumes and the smaller overall cell size. Cells showed lower mobility in vitro, paralleled by reduced protrusions. Constitutive PV down-regulation in PV-overexpressing cells reverted mitochondrial morphology and fractional volume to the state present in control MDCK cells, resulting from increased mitochondrial movement and augmented fusion rates. PV-modulated, bi-directional and reversible mitochondrial dynamics are key to regulation of mitochondrial volume.


Asunto(s)
Células Epiteliales/citología , Mitocondrias/ultraestructura , Dinámicas Mitocondriales , Parvalbúminas/metabolismo , Animales , Señalización del Calcio , Tamaño de la Célula , Perros , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Células de Riñón Canino Madin Darby , Mitocondrias/metabolismo , Mitofagia
5.
PLoS Biol ; 11(12): e1001755, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24391475

RESUMEN

Mitochondria are the power houses of the cell, but unlike the static structures portrayed in textbooks, they are dynamic organelles that move about the cell to deliver energy to locations in need. These organelles fuse with each other then split apart; some appear anchored and others more free to move around, and when damaged they are engulfed by autophagosomes. Together, these processes-mitochondrial trafficking, fusion and fission, and mitophagy-are best described by the term "mitochondrial dynamics". The molecular machineries behind these events are relatively well known yet the precise dynamics in neurons remains under debate. Neurons pose a peculiar logistical challenge to mitochondria; how do these energy suppliers manage to traffic down long axons to deliver the requisite energy supply to distant parts of the cell? To date, the majority of neuronal mitochondrial dynamics studies have used cultured neurons, Drosophila larvae, zebrafish embryos, with occasional experiments in resting mouse nerves. However, a new study in this issue of PLOS Biology from Marija Sajic and colleagues provides an in vivo look at mitochondrial dynamics along resting and electrically active neurons of live anaesthetized mice.


Asunto(s)
Mitocondrias/fisiología , Conducción Nerviosa/fisiología , Nervios Periféricos/fisiología , Animales , Masculino
6.
J Cell Sci ; 126(Pt 10): 2187-97, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23525002

RESUMEN

Mitochondrial fusion-fission dynamics play a crucial role in many important cell processes. These dynamics control mitochondrial morphology, which in turn influences several important mitochondrial properties including mitochondrial bioenergetics and quality control, and they appear to be affected in several neurodegenerative diseases. However, an integrated and quantitative understanding of how fusion-fission dynamics control mitochondrial morphology has not yet been described. Here, we took advantage of modern visualisation techniques to provide a clear explanation of how fusion and fission correlate with mitochondrial length and motility in neurons. Our main findings demonstrate that: (1) the probability of a single mitochondrion splitting is determined by its length; (2) the probability of a single mitochondrion fusing is determined primarily by its motility; (3) the fusion and fission cycle is driven by changes in mitochondrial length and deviations from this cycle serves as a corrective mechanism to avoid extreme mitochondrial length; (4) impaired mitochondrial motility in neurons overexpressing 120Q Htt or Tau suppresses mitochondrial fusion and leads to mitochondrial shortening whereas stimulation of mitochondrial motility by overexpressing Miro-1 restores mitochondrial fusion rates and sizes. Taken together, our results provide a novel insight into the complex crosstalk between different processes involved in mitochondrial dynamics. This knowledge will increase understanding of the dynamic mitochondrial functions in cells and in particular, the pathogenesis of mitochondrial-related neurodegenerative diseases.


Asunto(s)
Mitocondrias/metabolismo , Dinámicas Mitocondriales , Proteínas Mitocondriales/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas/ultraestructura , Proteínas de Unión al GTP rho/metabolismo , Animales , Humanos , Proteína Huntingtina , Mitocondrias/ultraestructura , Proteínas Mitocondriales/genética , Tamaño Mitocondrial/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células PC12 , Ratas , Ratas Wistar , Transgenes/genética , Proteínas de Unión al GTP rho/genética , Proteínas tau/genética , Proteínas tau/metabolismo
7.
J Cell Sci ; 125(Pt 3): 625-33, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22331352

RESUMEN

Calcium pumping into the endoplasmic reticulum (ER) lumen is thought to be coupled to a countertransport of protons through sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA) and the members of the ClC family of chloride channels. However, pH in the ER lumen remains neutral, which suggests a mechanism responsible for proton re-entry. We studied whether cation-proton exchangers could act as routes for such a re-entry. ER Ca(2+) uptake was measured in permeabilized immortalized hypothalamic neurons, primary rat cortical neurons and mouse cardiac fibers. Replacement of K(+) in the uptake solution with Na(+) or tetraethylammonium led to a strong inhibition of Ca(2+) uptake in neurons and cardiomyocytes. Furthermore, inhibitors of the potassium-proton exchanger (quinine or propranolol) but not of the sodium-proton exchanger reduced ER Ca(2+) uptake by 56-82%. Externally added nigericin, a potassium-proton exchanger, attenuated the inhibitory effect of propranolol. Inhibitors of small conductance calcium-sensitive K(+) (SK(Ca)) channels (UCL 1684, dequalinium) blocked the uptake of Ca(2+) by the ER in all preparations by 48-94%, whereas inhibitors of other K(+) channels (IK(Ca), BK(Ca) and K(ATP)) had no effect. Fluorescence microscopy and western blot analysis revealed the presence of both SK(Ca) channels and the potassium-proton exchanger leucine zipper-EF-hand-containing transmembrane protein 1 (LETM1) in ER in situ and in the purified ER fraction. The data obtained demonstrate that SK(Ca) channels and LETM1 reside in the ER membrane and that their activity is essential for ER Ca(2+) uptake.


Asunto(s)
Retículo Endoplásmico/metabolismo , Miocitos Cardíacos/metabolismo , Neuronas/metabolismo , Antiportadores de Potasio-Hidrógeno/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Transporte Iónico/efectos de los fármacos , Ratones , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Neuronas/efectos de los fármacos , Nigericina/farmacología , Antiportadores de Potasio-Hidrógeno/antagonistas & inhibidores , Propranolol/farmacología , Ratas , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores
9.
Front Endocrinol (Lausanne) ; 14: 1234925, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37900147

RESUMEN

Aim: Wolfram Syndrome (WS) is a rare condition caused by mutations in Wfs1, with a poor prognosis and no cure. Mono-agonists targeting the incretin glucagon-like-peptide 1 (GLP-1) have demonstrated disease-modifying potential in pre-clinical and clinical settings. Dual agonists that target GLP-1 and glucose-dependent insulinotropic polypeptide (GIP-1) are reportedly more efficacious; hence, we evaluated the therapeutic potential of dual incretin agonism in a loss-of-function rat model of WS. Methods: Eight-month-old Wfs1 knock-out (KO) and wild-type control rats were continuously treated with either the dual agonist DA-CH5 or saline for four months. Glycemic profile, visual acuity and hearing sensitivity were longitudinally monitored pre-treatment, and then at 10.5 and 12 months. Pancreata and retina were harvested for immunohistological analysis. Results: DA-CH5 therapy reversed glucose intolerance in KO rats and provided lasting anti-diabetogenic protection. Treatment also reversed intra-islet alterations, including reduced endocrine islet area and ß-cell density, indicating its regenerative potential. Although no rescue effect was noted for hearing loss, visual acuity and retinal ganglion cell density were better preserved in DA-CH5-treated rats. Conclusion: We present preclinical evidence for the pleiotropic therapeutic effects of long-term dual incretin agonist treatment; effects were seen despite treatment beginning after symptom-onset, indicating reversal of disease progression. Dual incretins represent a promising therapeutic avenue for WS patients.


Asunto(s)
Células Secretoras de Insulina , Síndrome de Wolfram , Humanos , Ratas , Animales , Lactante , Incretinas/farmacología , Síndrome de Wolfram/tratamiento farmacológico , Péptido 1 Similar al Glucagón/farmacología , Polipéptido Inhibidor Gástrico
10.
J Biol Chem ; 286(12): 10814-24, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21252228

RESUMEN

Parkinson disease is characterized by the accumulation of aggregated α-synuclein as the major component of the Lewy bodies. α-Synuclein accumulation in turn leads to compensatory effects that may include the up-regulation of autophagy. Another common feature of Parkinson disease (PD) is mitochondrial dysfunction. Here, we provide evidence that the overactivation of autophagy may be a link that connects the intracellular accumulation of α-synuclein with mitochondrial dysfunction. We found that the activation of macroautophagy in primary cortical neurons that overexpress mutant A53T α-synuclein leads to massive mitochondrial destruction and loss, which is associated with a bioenergetic deficit and neuronal degeneration. No mitochondrial removal or net loss was observed when we suppressed the targeting of mitochondria to autophagosomes by silencing Parkin, overexpressing wild-type Mitofusin 2 and dominant negative Dynamin-related protein 1 or blocking autophagy by silencing autophagy-related genes. The inhibition of targeting mitochondria to autophagosomes or autophagy was also partially protective against mutant A53T α-synuclein-induced neuronal cell death. These data suggest that overactivated mitochondrial removal could be one of the contributing factors that leads to the mitochondrial loss observed in PD models.


Asunto(s)
Autofagia , Mitocondrias/metabolismo , Mutación Missense , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Sustitución de Aminoácidos , Animales , Modelos Animales de Enfermedad , GTP Fosfohidrolasas , Silenciador del Gen , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mitocondrias/genética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Células PC12 , Enfermedad de Parkinson/genética , Ratas , Ratas Wistar , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , alfa-Sinucleína/genética
11.
Autophagy ; 18(9): 2249-2251, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35090371

RESUMEN

If cellular reactive oxygen species (ROS) production surpasses the intracellular antioxidant capacity, thus altering the ROS homeostasis, the cell needs to eradicate faulty mitochondria responsible for these excessive ROS. We have shown that even moderate ROS production breaks the KEAP1-PGAM5 complex, inhibiting the proteasomal removal of PGAM5. This leads to an accumulation of PGAM5 interfering with PINK1 processing that sensitizes mitochondria to autophagic removal. We propose that such a negative feedback system maintains cell ROS homeostasis.


Asunto(s)
Proteínas Mitocondriales , Mitofagia , Autofagia , Retroalimentación , Homeostasis , Proteína 1 Asociada A ECH Tipo Kelch , Proteínas Mitocondriales/metabolismo , Factor 2 Relacionado con NF-E2 , Fosfoproteínas Fosfatasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
12.
Cells ; 11(1)2021 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-35011599

RESUMEN

Mitochondria in the cell are the center for energy production, essential biomolecule synthesis, and cell fate determination. Moreover, the mitochondrial functional versatility enables cells to adapt to the changes in cellular environment and various stresses. In the process of discharging its cellular duties, mitochondria face multiple types of challenges, such as oxidative stress, protein-related challenges (import, folding, and degradation) and mitochondrial DNA damage. They mitigate all these challenges with robust quality control mechanisms which include antioxidant defenses, proteostasis systems (chaperones and proteases) and mitochondrial biogenesis. Failure of these quality control mechanisms leaves mitochondria as terminally damaged, which then have to be promptly cleared from the cells before they become a threat to cell survival. Such damaged mitochondria are degraded by a selective form of autophagy called mitophagy. Rigorous research in the field has identified multiple types of mitophagy processes based on targeting signals on damaged or superfluous mitochondria. In this review, we provide an in-depth overview of mammalian mitophagy and its importance in human health and diseases. We also attempted to highlight the future area of investigation in the field of mitophagy.


Asunto(s)
Mamíferos/metabolismo , Animales , Humanos , Mitofagia/genética , Modelos Biológicos , Biogénesis de Organelos , Receptores de Superficie Celular/metabolismo , Ubiquitina/metabolismo
13.
Sci Signal ; 14(702): eabc6165, 2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34582248

RESUMEN

Mutations in WFS1 (which encodes Wolframin, WFS1) and CISD2 (which encodes CDGSH iron sulfur domain 2) result in Wolfram syndrome (WS), a rare genetic disorder that starts with juvenile diabetes and progresses to neurological dysfunction. WFS1 and CISD2 belong to different protein families with distinct properties. Despite differences between WFS1 and CISD2, loss-of-function mutations in these proteins result in similar disease phenotypes, suggesting that they have convergent roles. WFS1 and CISD2 both localize at the endoplasmic reticulum (ER), the main intracellular calcium (Ca2+) store, which is implicated in several diseases, including WS. Here, we not only review the roles of WFS1 and CISD2 in Ca2+ signaling modulation but also point out knowledge gaps. Because WFS1 and CISD2 form complexes with Ca2+ transporters and Ca2+ channels, it is thought that they influence the activity of these transport systems in cells. Together, the studies reviewed here provide a better understanding of the pathogenesis and the severe disease burden of WS and may contribute to the development of therapeutics.


Asunto(s)
Síndrome de Wolfram , Humanos , Proteínas de la Membrana/genética , Transducción de Señal , Síndrome de Wolfram/genética
14.
Trends Cell Biol ; 31(7): 598-612, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33678551

RESUMEN

Organelles cooperate with each other to control cellular homeostasis and cell functions by forming close connections through membrane contact sites. Important contacts are present between the endoplasmic reticulum (ER), the main intracellular Ca2+-storage organelle, and the mitochondria, the organelle responsible not only for the majority of cellular ATP production but also for switching on cell death processes. Several Ca2+-transport systems focalize at these contact sites, thereby enabling the efficient transmission of Ca2+ signals from the ER toward mitochondria. This provides tight control of mitochondrial functions at the microdomain level. Here, we discuss how ER-mitochondrial Ca2+ transfers support cell function and how their dysregulation underlies, drives, or contributes to pathogenesis and pathophysiology, with a major focus on cancer and neurodegeneration but also with attention to other diseases such as diabetes and rare genetic diseases.


Asunto(s)
Señalización del Calcio , Calcio , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo
15.
Cells ; 10(11)2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34831417

RESUMEN

Wolfram syndrome (WS), also known as a DIDMOAD (diabetes insipidus, early-onset diabetes mellitus, optic nerve atrophy and deafness) is a rare autosomal disorder caused by mutations in the Wolframin1 (WFS1) gene. Previous studies have revealed that glucagon-like peptide-1 receptor agonist (GLP1 RA) are effective in delaying and restoring blood glucose control in WS animal models and patients. The GLP1 RA liraglutide has also been shown to have neuroprotective properties in aged WS rats. WS is an early-onset, chronic condition. Therefore, early diagnosis and lifelong pharmacological treatment is the best solution to control disease progression. Hence, the aim of this study was to evaluate the efficacy of the long-term liraglutide treatment on the progression of WS symptoms. For this purpose, 2-month-old WS rats were treated with liraglutide up to the age of 18 months and changes in diabetes markers, visual acuity, and hearing sensitivity were monitored over the course of the treatment period. We found that treatment with liraglutide delayed the onset of diabetes and protected against vision loss in a rat model of WS. Therefore, early diagnosis and prophylactic treatment with the liraglutide may also prove to be a promising treatment option for WS patients by increasing the quality of life.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Receptor del Péptido 1 Similar al Glucagón/agonistas , Pérdida Auditiva Sensorineural/tratamiento farmacológico , Liraglutida/uso terapéutico , Degeneración Nerviosa/tratamiento farmacológico , Vías Visuales/patología , Síndrome de Wolfram/tratamiento farmacológico , Animales , Péptido C/metabolismo , Diabetes Mellitus Experimental/complicaciones , Modelos Animales de Enfermedad , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Pérdida Auditiva Sensorineural/complicaciones , Liraglutida/farmacología , Masculino , Degeneración Nerviosa/complicaciones , Nervio Óptico/efectos de los fármacos , Nervio Óptico/patología , Nervio Óptico/ultraestructura , Fenotipo , Ratas , Vías Visuales/efectos de los fármacos , Síndrome de Wolfram/complicaciones
16.
Redox Biol ; 48: 102186, 2021 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-34801863

RESUMEN

When ROS production exceeds the cellular antioxidant capacity, the cell needs to eliminate the defective mitochondria responsible for excessive ROS production. It has been proposed that the removal of these defective mitochondria involves mitophagy, but the mechanism of this regulation remains unclear. Here, we demonstrate that moderate mitochondrial superoxide and hydrogen peroxide production oxidates KEAP1, thus breaking the interaction between this protein and PGAM5, leading to the inhibition of its proteasomal degradation. Accumulated PGAM5 interferes with the processing of the PINK1 in the mitochondria leading to the accumulation of PINK1 on the outer mitochondrial membrane. In turn, PINK1 promotes Parkin recruitment to mitochondria and sensitizes mitochondria for autophagic removal. We also demonstrate that inhibitors of the KEAP1-PGAM5 protein-protein interaction (including CPUY192018) mimic the effect of mitochondrial ROS and sensitize mitophagy machinery, suggesting that these inhibitors could be used as pharmacological regulators of mitophagy. Together, our results show that KEAP1/PGAM5 complex senses mitochondrially generated superoxide/hydrogen peroxide to induce mitophagy.

17.
Genes (Basel) ; 12(11)2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34828323

RESUMEN

Wolfram syndrome (WS) 1 is a rare monogenic neurodegenerative disorder caused by mutations in the gene encoding WFS1. Knowledge of the pathophysiology of WS is incomplete and to date, there is no treatment available. Here, we describe early deviations in the renin-angiotensin-aldosterone system (RAAS) and bradykinin pathway (kallikrein kinin system, KKS) observed in a rat model of WS (Wfs1 KO) and the modulative effect of glucagon-like peptide-1 receptor agonist liraglutide (LIR) and anti-epileptic drug valproate (VPA), which have been proven effective in delaying WS progression in WS animal models. We found that the expression of key receptors of the RAAS and KKS, Agtr2 and Bdkrb1, were drastically downregulated both in vitro and in vivo at an early stage in a rat model of WS. Moreover, in Wfs1, KO serum aldosterone levels were substantially decreased and bradykinin levels increased compared to WT animals. Neither treatment nor their combination affected the gene expression levels seen in the Wfs1 KO animals. However, all the treatments elevated serum aldosterone and decreased bradykinin in the Wfs1 KO rats, as well as increasing angiotensin II levels independent of genotype. Altogether, our results indicate that Wfs1 deficiency might disturb the normal functioning of RAAS and KKS and that LIR and VPA have the ability to modulate these systems.


Asunto(s)
Proteínas de Unión a Calmodulina/genética , Regulación hacia Abajo , Proteínas de la Membrana/genética , Receptor de Angiotensina Tipo 2/genética , Receptor de Bradiquinina B1/genética , Síndrome de Wolfram/genética , Aldosterona/sangre , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Técnicas de Inactivación de Genes , Liraglutida/farmacología , Liraglutida/uso terapéutico , Masculino , Ratas , Sistema Renina-Angiotensina/efectos de los fármacos , Ácido Valproico/farmacología , Ácido Valproico/uso terapéutico , Síndrome de Wolfram/sangre , Síndrome de Wolfram/tratamiento farmacológico
18.
Autophagy ; 15(5): 930-931, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30806158

RESUMEN

The Parkinson disease-associated proteins PINK1 and PRKN coordinate the ubiquitination of mitochondrial outer membrane proteins to tag them either for degradation or for autophagic clearance of the mitochondrion. The proteins include the mitochondrial trafficking proteins RHOT1 and RHOT2, the removal of which may be required for immobilization of mitochondria prior to mitophagy. Here, we demonstrate that RHOT1 and RHOT2 are not only substrates for PINK1-PRKN-dependent degradation but that they also play an active role in the process of mitophagy. RHOT1, and likely also RHOT2, may act as a docking site for inactive PRKN prior to mitochondrial damage, thus keeping PRKN in close proximity to its potential substrates and thereby facilitating mitophagy. We also show that RHOT1 functions as a calcium-sensing docking site for PRKN, and we suggest that calcium binding to RHOT is a key step in the calcium-dependent activation of mitophagy machinery.


Asunto(s)
Autofagia , Mitofagia , Proteínas Portadoras , Mitocondrias , Proteínas Mitocondriales , Proteínas Quinasas , Ubiquitina-Proteína Ligasas
19.
Sci Rep ; 9(1): 5457, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30932003

RESUMEN

Neuronal growth regulator 1 (NEGR1) belongs to the immunoglobulin (IgLON) superfamily of cell adhesion molecules involved in cortical layering. Recent functional and genomic studies implicate the role of NEGR1 in a wide spectrum of psychiatric disorders, such as major depression, schizophrenia and autism. Here, we investigated the impact of Negr1 deficiency on brain morphology, neuronal properties and social behavior of mice. In situ hybridization shows Negr1 expression in the brain nuclei which are central modulators of cortical-subcortical connectivity such as the island of Calleja and the reticular nucleus of thalamus. Brain morphological analysis revealed neuroanatomical abnormalities in Negr1-/- mice, including enlargement of ventricles and decrease in the volume of the whole brain, corpus callosum, globus pallidus and hippocampus. Furthermore, decreased number of parvalbumin-positive inhibitory interneurons was evident in Negr1-/- hippocampi. Behaviorally, Negr1-/- mice displayed hyperactivity in social interactions and impairments in social hierarchy. Finally, Negr1 deficiency resulted in disrupted neurite sprouting during neuritogenesis. Our results provide evidence that NEGR1 is required for balancing the ratio of excitatory/inhibitory neurons and proper formation of brain structures, which is prerequisite for adaptive behavioral profiles. Therefore, Negr1-/- mice have a high potential to provide new insights into the neural mechanisms of neuropsychiatric disorders.


Asunto(s)
Conducta Animal , Encéfalo/patología , Moléculas de Adhesión Celular Neuronal/genética , Endofenotipos , Trastornos Mentales/patología , Animales , Encéfalo/metabolismo , Trastornos Mentales/metabolismo , Ratones , Ratones Noqueados
20.
Neurotoxicology ; 29(1): 101-8, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17981335

RESUMEN

N-Methyl-d-aspartate (NMDA) receptor stimulation promotes neuronal survival and differentiation under both in vitro and in vivo conditions. We studied the effects of various NMDA receptor antagonists acting at different NMDA receptor binding sites and non-NMDA receptor antagonists on the development and survival of cerebellar granule cell (CGC) culture. Only three of the drugs tested induced neurotoxicity-MK-801 (non-competitive NMDA channel blocking antagonist), ifenprodil (an antagonist of the NR2B site and polyamine site of the NMDA receptor) and L-701.324 (full antagonist at glycine site), while CGP-37849 (a competitive NMDA antagonist), (+)-HA-966 (a partial agonist of the glycine site of the NMDA receptor), and NBQX (a competitively acting AMPA receptor antagonist) were not toxic at any concentration (1-100 microM) used. Among these drugs, only MK-801 was toxic for the immature CGC on second day in vitro (2DIV), and toxicity was diminished parallel to the neuronal maturation. In more mature neurons (7DIV), MK-801 demonstrated some neuroprotection, which diminished spontaneously occurring neuronal death in culture. Neither NMDA nor glutamate were able to prevent the neurotoxic effect of MK-801 at 2DIV. MK-801, ifenprodil and L-701.324 induced DNA fragmentation on 2DIV in CGC culture measured by the TUNEL method. The BOC-D-FMK, the universal caspase inhibitor, completely reversed MK-801-induced DNA fragmentation, suggesting an apoptotic pathway of MK-801-induced cell death. Neurite outgrowth as a characteristic feature of the development of CGC was diminished after treatment with MK-801, ifenprodil and L-701.324. In conclusion, the results of the present study demonstrate that only nonselective channel blocker MK-801 decreases cell viability, induces apoptosis and inhibits neurite outgrowth of CGC in a development-dependent manner.


Asunto(s)
Cerebelo/citología , Antagonistas de Aminoácidos Excitadores/farmacología , Neuronas/efectos de los fármacos , Análisis de Varianza , Animales , Animales Recién Nacidos , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/farmacología , Etiquetado Corte-Fin in Situ , N-Metilaspartato/farmacología , Ratas , Ratas Wistar , Sales de Tetrazolio , Tiazoles , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA