Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cell ; 155(6): 1351-64, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24290359

RESUMEN

Parkinson's disease (PD) is characterized by loss of A9 dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). An association has been reported between PD and exposure to mitochondrial toxins, including environmental pesticides paraquat, maneb, and rotenone. Here, using a robust, patient-derived stem cell model of PD allowing comparison of A53T α-synuclein (α-syn) mutant cells and isogenic mutation-corrected controls, we identify mitochondrial toxin-induced perturbations in A53T α-syn A9 DA neurons (hNs). We report a pathway whereby basal and toxin-induced nitrosative/oxidative stress results in S-nitrosylation of transcription factor MEF2C in A53T hNs compared to corrected controls. This redox reaction inhibits the MEF2C-PGC1α transcriptional network, contributing to mitochondrial dysfunction and apoptotic cell death. Our data provide mechanistic insight into gene-environmental interaction (GxE) in the pathogenesis of PD. Furthermore, using small-molecule high-throughput screening, we identify the MEF2C-PGC1α pathway as a therapeutic target to combat PD.


Asunto(s)
Interacción Gen-Ambiente , Mitocondrias/efectos de los fármacos , Paraquat/toxicidad , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factores de Transcripción MEF2 , Mutación/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo , Enfermedad de Parkinson/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Especies de Nitrógeno Reactivo/metabolismo , Sustancia Negra/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33833060

RESUMEN

Parkinson's disease is characterized by accumulation of α-synuclein (αSyn). Release of oligomeric/fibrillar αSyn from damaged neurons may potentiate neuronal death in part via microglial activation. Heretofore, it remained unknown if oligomeric/fibrillar αSyn could activate the nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome in human microglia and whether anti-αSyn antibodies could prevent this effect. Here, we show that αSyn activates the NLRP3 inflammasome in human induced pluripotent stem cell (hiPSC)-derived microglia (hiMG) via dual stimulation involving Toll-like receptor 2 (TLR2) engagement and mitochondrial damage. In vitro, hiMG can be activated by mutant (A53T) αSyn secreted from hiPSC-derived A9-dopaminergic neurons. Surprisingly, αSyn-antibody complexes enhanced rather than suppressed inflammasome-mediated interleukin-1ß (IL-1ß) secretion, indicating these complexes are neuroinflammatory in a human context. A further increase in inflammation was observed with addition of oligomerized amyloid-ß peptide (Aß) and its cognate antibody. In vivo, engraftment of hiMG with αSyn in humanized mouse brain resulted in caspase-1 activation and neurotoxicity, which was exacerbated by αSyn antibody. These findings may have important implications for antibody therapies aimed at depleting misfolded/aggregated proteins from the human brain, as they may paradoxically trigger inflammation in human microglia.


Asunto(s)
Inflamasomas/metabolismo , Microglía/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedad de Parkinson/inmunología , alfa-Sinucleína/inmunología , Péptidos beta-Amiloides/inmunología , Anticuerpos/inmunología , Diferenciación Celular , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/citología , Microglía/citología , Receptor Toll-Like 2/metabolismo , alfa-Sinucleína/genética
3.
J Neurosci ; 42(14): 3011-3024, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35169022

RESUMEN

Dysregulation of autophagic pathways leads to accumulation of abnormal proteins and damaged organelles in many neurodegenerative disorders, including Parkinson's disease (PD) and Lewy body dementia (LBD). Autophagy-related dysfunction may also trigger secretion and spread of misfolded proteins, such as α-synuclein (α-syn), the major misfolded protein found in PD/LBD. However, the mechanism underlying these phenomena remains largely unknown. Here, we used cell-based models, including human induced pluripotent stem cell-derived neurons, CRISPR/Cas9 technology, and male transgenic PD/LBD mice, plus vetting in human postmortem brains (both male and female). We provide mechanistic insight into this pathologic pathway. We find that aberrant S-nitrosylation of the autophagic adaptor protein p62 causes inhibition of autophagic flux and intracellular buildup of misfolded proteins, with consequent secretion resulting in cell-to-cell spread. Thus, our data show that pathologic protein S-nitrosylation of p62 represents a critical factor not only for autophagic inhibition and demise of individual neurons, but also for α-syn release and spread of disease throughout the nervous system.SIGNIFICANCE STATEMENT In Parkinson's disease and Lewy body dementia, dysfunctional autophagy contributes to accumulation and spread of aggregated α-synuclein. Here, we provide evidence that protein S-nitrosylation of p62 inhibits autophagic flux, contributing to α-synuclein aggregation and spread.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad por Cuerpos de Lewy , Enfermedad de Parkinson , Proteínas de Unión al ARN , alfa-Sinucleína , Animales , Autofagia , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad por Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/patología , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Proteína S/metabolismo , Proteínas de Unión al ARN/metabolismo , alfa-Sinucleína/metabolismo
4.
Mol Psychiatry ; 26(10): 5751-5765, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-32467645

RESUMEN

Beginning at early stages, human Alzheimer's disease (AD) brains manifest hyperexcitability, contributing to subsequent extensive synapse loss, which has been linked to cognitive dysfunction. No current therapy for AD is disease-modifying. Part of the problem with AD drug discovery is that transgenic mouse models have been poor predictors of potential human treatment. While it is undoubtedly important to test drugs in these animal models, additional evidence for drug efficacy in a human context might improve our chances of success. Accordingly, in order to test drugs in a human context, we have developed a platform of physiological assays using patch-clamp electrophysiology, calcium imaging, and multielectrode array (MEA) experiments on human (h)iPSC-derived 2D cortical neuronal cultures and 3D cerebral organoids. We compare hiPSCs bearing familial AD mutations vs. their wild-type (WT) isogenic controls in order to characterize the aberrant electrical activity in such a human context. Here, we show that these AD neuronal cultures and organoids manifest increased spontaneous action potentials, slow oscillatory events (~1 Hz), and hypersynchronous network activity. Importantly, the dual-allosteric NMDAR antagonist NitroSynapsin, but not the FDA-approved drug memantine, abrogated this hyperactivity. We propose a novel model of synaptic plasticity in which aberrant neural networks are rebalanced by NitroSynapsin. We propose that hiPSC models may be useful for screening drugs to treat hyperexcitability and related synaptic damage in AD.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Potenciales de Acción , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Animales , Modelos Animales de Enfermedad , Ratones , Redes Neurales de la Computación , Neuronas
5.
Proc Natl Acad Sci U S A ; 113(47): E7564-E7571, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27821734

RESUMEN

Recent studies have pointed to protein S-nitrosylation as a critical regulator of cellular redox homeostasis. For example, S-nitrosylation of peroxiredoxin-2 (Prx2), a peroxidase widely expressed in mammalian neurons, inhibits both enzymatic activity and protective function against oxidative stress. Here, using in vitro and in vivo approaches, we identify a role and reaction mechanism of the reductase sulfiredoxin (Srxn1) as an enzyme that denitrosylates (thus removing -SNO) from Prx2 in an ATP-dependent manner. Accordingly, by decreasing S-nitrosylated Prx2 (SNO-Prx2), overexpression of Srxn1 protects dopaminergic neural cells and human-induced pluripotent stem cell (hiPSC)-derived neurons from NO-induced hypersensitivity to oxidative stress. The pathophysiological relevance of this observation is suggested by our finding that SNO-Prx2 is dramatically increased in murine and human Parkinson's disease (PD) brains. Our findings therefore suggest that Srxn1 may represent a therapeutic target for neurodegenerative disorders such as PD that involve nitrosative/oxidative stress.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Enfermedad de Parkinson/metabolismo , Peroxirredoxinas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/citología , Humanos , Hidrólisis , Células Madre Pluripotentes Inducidas/citología , Ratones , Óxido Nítrico/química , Estrés Oxidativo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Peroxirredoxinas/química , Fosforilación
6.
J Neurochem ; 133(6): 898-908, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25692407

RESUMEN

Cyanide is a life-threatening, bioterrorist agent, preventing cellular respiration by inhibiting cytochrome c oxidase, resulting in cardiopulmonary failure, hypoxic brain injury, and death within minutes. However, even after treatment with various antidotes to protect cytochrome oxidase, cyanide intoxication in humans can induce a delayed-onset neurological syndrome that includes symptoms of Parkinsonism. Additional mechanisms are thought to underlie cyanide-induced neuronal damage, including generation of reactive oxygen species. This may account for the fact that antioxidants prevent some aspects of cyanide-induced neuronal damage. Here, as a potential preemptive countermeasure against a bioterrorist attack with cyanide, we tested the CNS protective effect of carnosic acid (CA), a pro-electrophilic compound found in the herb rosemary. CA crosses the blood-brain barrier to up-regulate endogenous antioxidant enzymes via activation of the Nrf2 transcriptional pathway. We demonstrate that CA exerts neuroprotective effects on cyanide-induced brain damage in cultured rodent and human-induced pluripotent stem cell-derived neurons in vitro, and in vivo in various brain areas of a non-Swiss albino mouse model of cyanide poisoning that simulates damage observed in the human brain. Cyanide, a potential bioterrorist agent, can produce a chronic delayed-onset neurological syndrome that includes symptoms of Parkinsonism. Here, cyanide poisoning treated with the proelectrophillic compound carnosic acid, results in reduced neuronal cell death in both in vitro and in vivo models through activation of the Nrf2/ARE transcriptional pathway. Carnosic acid is therefore a potential treatment for the toxic central nervous system (CNS) effects of cyanide poisoning. ARE, antioxidant responsive element; Nrf2 (NFE2L2, Nuclear factor (erythroid-derived 2)-like 2).


Asunto(s)
Abietanos/farmacología , Lesiones Encefálicas/prevención & control , Cianuros/toxicidad , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Extractos Vegetales/farmacología , Animales , Antioxidantes/farmacología , Bioterrorismo , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , Ratas , Ratas Sprague-Dawley
7.
J Cell Sci ; 125(Pt 2): 287-94, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22302996

RESUMEN

Mutations in the store-operated Ca²âº entry pore protein ORAI1 have been reported to cause myopathies in human patients but the mechanism involved is not known. Cardiomyocytes express ORAI1 but its role in heart function is also unknown. Using reverse genetics in zebrafish, we demonstrated that inactivation of the highly conserved zebrafish orthologue of ORAI1 resulted in severe heart failure, reduced ventricular systolic function, bradycardia and skeletal muscle weakness. Electron microscopy of Orai1-deficient myocytes revealed progressive skeletal muscle instability with loss of myofiber integrity and ultrastructural abnormalities of the z-disc in both skeletal and cardiac muscle. Isolated Orai1-deficient cardiomyocytes showed loss of the calcineurin-associated protein calsarcin from the z-discs. Furthermore, we found mechanosignal transduction was affected in Orai1-depleted hearts, indicating an essential role for ORAI1 in establishing the cardiac signaling transduction machinery at the z-disc. Our findings identify ORAI1 as an important regulator of cardiac and skeletal muscle function and provide evidence linking ORAI1-mediated calcium signaling to sarcomere integrity and cardiomyocyte function.


Asunto(s)
Canales de Calcio/fisiología , Insuficiencia Cardíaca/etiología , Debilidad Muscular/etiología , Proteínas de Pez Cebra/fisiología , Animales , Canales de Calcio/deficiencia , Canales de Calcio/genética , Canales de Calcio/metabolismo , Señalización del Calcio , Cardiomegalia/metabolismo , Células Cultivadas , Corazón/fisiología , Mecanotransducción Celular , Ratones , Músculo Esquelético/ultraestructura , Miocardio/metabolismo , Miocardio/ultraestructura , Miocitos Cardíacos/metabolismo , Proteína ORAI1 , Ratas , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
8.
Adv Sci (Weinh) ; 11(12): e2306469, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38235614

RESUMEN

In Alzheimer's disease (AD), dysfunctional mitochondrial metabolism is associated with synaptic loss, the major pathological correlate of cognitive decline. Mechanistic insight for this relationship, however, is still lacking. Here, comparing isogenic wild-type and AD mutant human induced pluripotent stem cell (hiPSC)-derived cerebrocortical neurons (hiN), evidence is found for compromised mitochondrial energy in AD using the Seahorse platform to analyze glycolysis and oxidative phosphorylation (OXPHOS). Isotope-labeled metabolic flux experiments revealed a major block in activity in the tricarboxylic acid (TCA) cycle at the α-ketoglutarate dehydrogenase (αKGDH)/succinyl coenzyme-A synthetase step, metabolizing α-ketoglutarate to succinate. Associated with this block, aberrant protein S-nitrosylation of αKGDH subunits inhibited their enzyme function. This aberrant S-nitrosylation is documented not only in AD-hiN but also in postmortem human AD brains versus controls, as assessed by two separate unbiased mass spectrometry platforms using both SNOTRAP identification of S-nitrosothiols and chemoselective-enrichment of S-nitrosoproteins. Treatment with dimethyl succinate, a cell-permeable derivative of a TCA substrate downstream to the block, resulted in partial rescue of mitochondrial bioenergetic function as well as reversal of synapse loss in AD-hiN. These findings have therapeutic implications that rescue of mitochondrial energy metabolism can ameliorate synaptic loss in hiPSC-based models of AD.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Humanos , Enfermedad de Alzheimer/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Metabolismo Energético/fisiología , Glucólisis , Neuronas/metabolismo
9.
J Neurosci ; 32(45): 15837-42, 2012 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-23136422

RESUMEN

After transplantation, individual stem cell-derived neurons can functionally integrate into the host CNS; however, evidence that neurons derived from transplanted human embryonic stem cells (hESCs) can drive endogenous neuronal network activity in CNS tissue is still lacking. Here, using multielectrode array recordings, we report activation of high-frequency oscillations in the ß and γ ranges (10-100 Hz) in the host hippocampal network via targeted optogenetic stimulation of transplanted hESC-derived neurons.


Asunto(s)
Células Madre Embrionarias/trasplante , Hipocampo/fisiología , Células-Madre Neurales/trasplante , Neuronas/trasplante , Potenciales de Acción/fisiología , Animales , Células Madre Embrionarias/citología , Femenino , Hipocampo/citología , Humanos , Masculino , Células-Madre Neurales/citología , Neurogénesis/fisiología , Neuronas/citología , Optogenética , Ratas , Ratas Sprague-Dawley
10.
Cell Chem Biol ; 30(8): 965-975.e6, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37478858

RESUMEN

A causal relationship between mitochondrial metabolic dysfunction and neurodegeneration has been implicated in synucleinopathies, including Parkinson disease (PD) and Lewy body dementia (LBD), but underlying mechanisms are not fully understood. Here, using human induced pluripotent stem cell (hiPSC)-derived neurons with mutation in the gene encoding α-synuclein (αSyn), we report the presence of aberrantly S-nitrosylated proteins, including tricarboxylic acid (TCA) cycle enzymes, resulting in activity inhibition assessed by carbon-labeled metabolic flux experiments. This inhibition principally affects α-ketoglutarate dehydrogenase/succinyl coenzyme-A synthetase, metabolizing α-ketoglutarate to succinate. Notably, human LBD brain manifests a similar pattern of aberrantly S-nitrosylated TCA enzymes, indicating the pathophysiological relevance of these results. Inhibition of mitochondrial energy metabolism in neurons is known to compromise dendritic length and synaptic integrity, eventually leading to neuronal cell death. Our evidence indicates that aberrant S-nitrosylation of TCA cycle enzymes contributes to this bioenergetic failure.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Sinucleinopatías , Humanos , Sinucleinopatías/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Parkinson/metabolismo , Neuronas/metabolismo , Encéfalo/metabolismo
11.
J Mol Cell Cardiol ; 48(6): 1329-34, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20138887

RESUMEN

Cardiac hypertrophy is an independent risk for heart failure (HF) and sudden death. Deciphering signalling pathways dependent on extracellular calcium (Ca(2+)) influx that control normal and pathological cardiac growth may enable identification of novel therapeutic targets. The objective of the present study is to determine the role of the Ca(2+) release-activated Ca(2+) (CRAC) channel Orai1 and stromal interaction molecule 1 (Stim1) in postnatal cardiomyocyte store operated Ca(2+) entry (SOCE) and impact on normal and hypertrophic postnatal cardiomyocyte growth. Employing a combination of siRNA-mediated gene silencing, cultured neonatal rat ventricular cardiomyocytes together with indirect immunofluorescence, epifluorescent Ca(2+) imaging and site-specific protein phosphorylation and real-time mRNA expression analysis, we show for the first time that both Orai1 and Stim1 are present in cardiomyocytes and required for SOCE due to intracellular Ca(2+) store depletion by thapsigargin. Stim1-KD but not Orai1-KD significantly decreased diastolic Ca(2+) levels and caffeine-releasable Ca(2+) from the sarcoplasmic reticulum (SR). Conversely, Orai1-KD but not Stim1-KD significantly diminished basal NRCM cell size, anp and bnp mRNA levels and activity of the calcineurin (CnA) signalling pathway although diminishing both Orai1 and Stim1 proteins similarly attenuated calmodulin kinase II (CamKII) and ERK1/2 activity under basal conditions. Both Orai1- and Stim1-KD completely abrogated phenylephrine (PE) mediated hypertrophic NRCM growth and enhanced natriuretic factor expression by inhibiting G(q)-protein conveyed activation of the CamKII and ERK1/2 signalling pathway. Interestingly, only Orai1-KD but not Stim1-KD prevented Gq-mediated CaN-dependent prohypertrophic signalling. This study shows for the first time that both Orai1 and Stim1 have a key role in cardiomyocyte SOCE regulating both normal and hypertrophic postnatal cardiac growth in vitro.


Asunto(s)
Canales de Calcio/metabolismo , Regulación de la Expresión Génica , Glicoproteínas de Membrana/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Calcio/metabolismo , Proliferación Celular , Microscopía Fluorescente/métodos , Miocitos Cardíacos/citología , Proteína ORAI1 , Fenilefrina/química , Fosforilación , Ratas , Transducción de Señal , Molécula de Interacción Estromal 1 , Tapsigargina/química , Tapsigargina/farmacología
12.
Elife ; 82019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31782729

RESUMEN

Human Alzheimer's disease (AD) brains and transgenic AD mouse models manifest hyperexcitability. This aberrant electrical activity is caused by synaptic dysfunction that represents the major pathophysiological correlate of cognitive decline. However, the underlying mechanism for this excessive excitability remains incompletely understood. To investigate the basis for the hyperactivity, we performed electrophysiological and immunofluorescence studies on hiPSC-derived cerebrocortical neuronal cultures and cerebral organoids bearing AD-related mutations in presenilin-1 or amyloid precursor protein vs. isogenic gene corrected controls. In the AD hiPSC-derived neurons/organoids, we found increased excitatory bursting activity, which could be explained in part by a decrease in neurite length. AD hiPSC-derived neurons also displayed increased sodium current density and increased excitatory and decreased inhibitory synaptic activity. Our findings establish hiPSC-derived AD neuronal cultures and organoids as a relevant model of early AD pathophysiology and provide mechanistic insight into the observed hyperexcitability.


Asunto(s)
Potenciales de Acción , Enfermedad de Alzheimer/fisiopatología , Cerebro/citología , Excitabilidad Cortical , Fenómenos Electrofisiológicos , Células Madre Pluripotentes Inducidas/fisiología , Neuronas/fisiología , Precursor de Proteína beta-Amiloide/genética , Animales , Tamaño de la Célula , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Modelos Teóricos , Proteínas Mutantes/genética , Organoides , Presenilina-1/genética
13.
J Genet ; 97(3): 729-751, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30027906

RESUMEN

Parkinson's disease (PD) is a debilitating neurodegenerative disorder, for which people above the age of 60 show an increased risk. Although there has been great advancement in understanding the disease-related abnormalities in brain circuitry and development of symptomatic treatments, a cure for PD remains elusive. The discovery of PD associated gene mutations and environmental toxins have yielded animal models of the disease. These models could recapitulate several key aspects of PD, and provide more insights into the disease pathogenesis. They have also revealed novel aspects of the disease mechanism including noncell autonomous events and spreading of pathogenic protein species across the brain. Nevertheless, none of these models so far can comprehensively represent all aspects of the human disease. While the field is still searching for the perfect model system, recent developments in stem cell biology have provided a new dimension to modelling PD, especially doing it in a patient-specific manner. In the current review, we attempt to summarize the key findings in the areas discussed above, and highlight how the core PD pathology distinguishes itself from other neurodegenerative disorders while also resembling them in many aspects.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad de Parkinson/patología , Animales , Interacción Gen-Ambiente , Humanos , Inflamación/patología , Modelos Biológicos , Mutación/genética , Enfermedad de Parkinson/genética
14.
Cell Rep ; 21(8): 2171-2182, 2017 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-29166608

RESUMEN

Mutations in PARK6 (PINK1) and PARK2 (Parkin) are linked to rare familial cases of Parkinson's disease (PD). Mutations in these genes result in pathological dysregulation of mitophagy, contributing to neurodegeneration. Here, we report that environmental factors causing a specific posttranslational modification on PINK1 can mimic these genetic mutations. We describe a molecular mechanism for impairment of mitophagy via formation of S-nitrosylated PINK1 (SNO-PINK1). Mitochondrial insults simulating age- or environmental-related stress lead to increased SNO-PINK1, inhibiting its kinase activity. SNO-PINK1 decreases Parkin translocation to mitochondrial membranes, disrupting mitophagy in cell lines and human-iPSC-derived neurons. We find levels of SNO-PINK1 in brains of α-synuclein transgenic PD mice similar to those in cell-based models, indicating the pathophysiological relevance of our findings. Importantly, SNO-PINK1-mediated deficits in mitophagy contribute to neuronal cell death. These results reveal a direct molecular link between nitrosative stress, SNO-PINK1 formation, and mitophagic dysfunction that contributes to the pathogenesis of PD.


Asunto(s)
Mitocondrias/genética , Mitofagia/genética , Enfermedad de Parkinson/genética , Proteínas Quinasas/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Mitocondrias/metabolismo , Mutación/genética , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Quinasas/metabolismo
15.
Nat Commun ; 7: 10242, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26743041

RESUMEN

Metabolic syndrome (MetS) and Type 2 diabetes mellitus (T2DM) increase risk for Alzheimer's disease (AD). The molecular mechanism for this association remains poorly defined. Here we report in human and rodent tissues that elevated glucose, as found in MetS/T2DM, and oligomeric ß-amyloid (Aß) peptide, thought to be a key mediator of AD, coordinately increase neuronal Ca(2+) and nitric oxide (NO) in an NMDA receptor-dependent manner. The increase in NO results in S-nitrosylation of insulin-degrading enzyme (IDE) and dynamin-related protein 1 (Drp1), thus inhibiting insulin and Aß catabolism as well as hyperactivating mitochondrial fission machinery. Consequent elevation in Aß levels and compromise in mitochondrial bioenergetics result in dysfunctional synaptic plasticity and synapse loss in cortical and hippocampal neurons. The NMDA receptor antagonist memantine attenuates these effects. Our studies show that redox-mediated posttranslational modification of brain proteins link Aß and hyperglycaemia to cognitive dysfunction in MetS/T2DM and AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Dinaminas/metabolismo , Glucosa/metabolismo , Hiperglucemia/metabolismo , Insulisina/metabolismo , Neuronas/metabolismo , Óxido Nítrico/metabolismo , Compuestos Nitrosos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Animales , Encéfalo/citología , Encéfalo/patología , Estudios de Casos y Controles , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Espinas Dendríticas , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , GTP Fosfohidrolasas/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Immunoblotting , Células Madre Pluripotentes Inducidas , Insulina/metabolismo , Potenciación a Largo Plazo , Masculino , Memantina/farmacología , Síndrome Metabólico/metabolismo , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Mitocondriales/metabolismo , Consumo de Oxígeno , Ratas , Especies de Nitrógeno Reactivo , Sinapsis/metabolismo
16.
J Comp Neurol ; 522(12): 2845-56, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24756727

RESUMEN

Neural transplantation is a promising strategy for restoring dopaminergic dysfunction and modifying disease progression in Parkinson's disease (PD). Human embryonic stem cells (hESCs) are a potential resource in this regard because of their ability to provide a virtually limitless supply of homogenous dopaminergic progenitors and neurons of appropriate lineage. The recent advances in developing robust cell culture protocols for directed differentiation of hESCs to near pure populations of ventral mesencephalic (A9-type) dopaminergic neurons has heightened the prospects for PD cell therapy. Here, we focus our review on current state-of-the-art techniques for harnessing hESC-based strategies toward development of a stem cell therapeutic for PD. Importantly, we also briefly describe a novel genetic-programming approach that may address many of the key challenges that remain in the field and that may hasten clinical translation.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Embrionarias/fisiología , Células-Madre Neurales/trasplante , Enfermedad de Parkinson/cirugía , Animales , Diferenciación Celular , Humanos , Células-Madre Neurales/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA