Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Pineal Res ; 75(4): e12909, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37721126

RESUMEN

Huntington's disease (HD) is a progressive neurodegenerative brain disorder associated with uncontrolled body movements, cognitive decline, and reduced circulating melatonin levels. Melatonin is a potent antioxidant and exogenous melatonin treatment is neuroprotective in experimental HD models. In neurons, melatonin is exclusively synthesized in the mitochondrial matrix. Thus, we investigated the integrity of melatonin biosynthesis pathways in pineal and extrapineal brain areas in human HD brain samples, in the R6/2 mouse model of HD and in full-length mutant huntingtin knock-in cells. Aralkylamine N-acetyltransferase (AANAT) is the rate-limiting step enzyme in the melatonin biosynthetic pathway. We found that AANAT expression is significantly decreased in the pineal gland and the striatum of HD patients compared to normal controls. In the R6/2 mouse forebrain, AANAT protein expression was decreased in synaptosomal, but not nonsynaptosomal, mitochondria and was associated with decreased synaptosomal melatonin levels compared to wild type mice. We also demonstrate sequestration of AANAT in mutant-huntingtin protein aggregates likely resulting in decreased AANAT bioavailability. Paradoxically, AANAT mRNA expression is increased in tissues where AANAT protein expression is decreased, suggesting a potential feedback loop that is, ultimately unsuccessful. In conclusion, we demonstrate that pineal, extrapineal, and synaptosomal melatonin levels are compromised in the brains of HD patients and R6/2 mice due, at least in part, to protein aggregation.


Asunto(s)
Enfermedad de Huntington , Melatonina , Glándula Pineal , Humanos , Ratones , Animales , Melatonina/metabolismo , Glándula Pineal/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(33): 16593-16602, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31346086

RESUMEN

Mutant huntingtin (mHTT), the causative protein in Huntington's disease (HD), associates with the translocase of mitochondrial inner membrane 23 (TIM23) complex, resulting in inhibition of synaptic mitochondrial protein import first detected in presymptomatic HD mice. The early timing of this event suggests that it is a relevant and direct pathophysiologic consequence of mHTT expression. We show that, of the 4 TIM23 complex proteins, mHTT specifically binds to the TIM23 subunit and that full-length wild-type huntingtin (wtHTT) and mHTT reside in the mitochondrial intermembrane space. We investigated differences in mitochondrial proteome between wtHTT and mHTT cells and found numerous proteomic disparities between mHTT and wtHTT mitochondria. We validated these data by quantitative immunoblotting in striatal cell lines and human HD brain tissue. The level of soluble matrix mitochondrial proteins imported through the TIM23 complex is lower in mHTT-expressing cell lines and brain tissues of HD patients compared with controls. In mHTT-expressing cell lines, membrane-bound TIM23-imported proteins have lower intramitochondrial levels, whereas inner membrane multispan proteins that are imported via the TIM22 pathway and proteins integrated into the outer membrane generally remain unchanged. In summary, we show that, in mitochondria, huntingtin is located in the intermembrane space, that mHTT binds with high-affinity to TIM23, and that mitochondria from mHTT-expressing cells and brain tissues of HD patients have reduced levels of nuclearly encoded proteins imported through TIM23. These data demonstrate the mechanism and biological significance of mHTT-mediated inhibition of mitochondrial protein import, a mechanism likely broadly relevant to other neurodegenerative diseases.


Asunto(s)
Proteína Huntingtina/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mutantes/metabolismo , Proteostasis , Línea Celular , Núcleo Celular/metabolismo , Corteza Cerebral/patología , Cuerpo Estriado/patología , Humanos , Enfermedad de Huntington , Membranas Mitocondriales/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteínas Mitocondriales/metabolismo , Unión Proteica , Proteoma/metabolismo
3.
Proc Natl Acad Sci U S A ; 116(2): 650-659, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30584104

RESUMEN

Neuritic retraction in the absence of overt neuronal death is a shared feature of normal aging and neurodegenerative disorders, but the intracellular mechanisms modulating this process are not understood. We propose that cumulative distal mitochondrial protein damage results in impaired protein import, leading to mitochondrial dysfunction and focal activation of the canonical apoptosis pathway in neurites. This is a controlled process that may not lead to neuronal death and, thus, we term this phenomenon "neuritosis." Consistent with our hypothesis, we show that in primary cerebrocortical neurons, mitochondrial distance from the soma correlates with increased mitochondrial protein damage, PINK1 accumulation, reactive oxygen species production, and decreased mitochondrial membrane potential and depolarization threshold. Furthermore, we demonstrate that the distance-dependent mitochondrial membrane potential gradient exists in vivo in mice. We demonstrate that impaired distal mitochondria have a lower threshold for focal/nonlethal neuritic caspase-3 activation in normal neurons that is exacerbated in aging, stress, and neurodegenerative conditions, thus delineating a fundamental mechanistic underpinning for synaptic vulnerability.


Asunto(s)
Apoptosis , Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Neuritas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Ratones , Ratones Transgénicos , Mitocondrias/genética , Mitocondrias/patología , Neuritas/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
4.
Neurobiol Dis ; 158: 105451, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34298088

RESUMEN

In healthy neurons, a mitochondrial membrane potential gradient exists whereby membrane potential is highest in the soma and decreases with distance from the nucleus. Correspondingly, distal mitochondria have more oxidative damage and slower protein import than somal mitochondria. Due to these differences, distal mitochondria have an intrinsic first stressor that somal mitochondria do not have, resulting in synaptic mitochondrial vulnerability. A second stressor may result from mutant protein expression, situational stress, or aging, exacerbating vulnerable mitochondria activating stress responses. Under these conditions, distal mitochondria release cytochrome c and mitochondrial DNA, leading to compartmentalized sub-lethal caspase-3 activation and cytokine production. In this two-hit mitochondrial-driven synaptic loss model, synapse vulnerability during neurodegeneration is explained as a superposition of pre-existing lower synaptic mitochondrial membrane potential (hit one) with additional mitochondrial stress (hit two). This two-hit mechanism occurs in synaptic mitochondria, activating signaling pathways leading to synaptic degeneration, as a potential preamble to neuronal death.


Asunto(s)
Mitocondrias/patología , Enfermedades Neurodegenerativas/patología , Sinapsis/patología , Animales , Modelos Animales de Enfermedad , Humanos , Potencial de la Membrana Mitocondrial , Estrés Oxidativo
5.
Proc Natl Acad Sci U S A ; 114(38): E7997-E8006, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28874589

RESUMEN

G protein-coupled receptors (GPCRs) are classically characterized as cell-surface receptors transmitting extracellular signals into cells. Here we show that central components of a GPCR signaling system comprised of the melatonin type 1 receptor (MT1), its associated G protein, and ß-arrestins are on and within neuronal mitochondria. We discovered that the ligand melatonin is exclusively synthesized in the mitochondrial matrix and released by the organelle activating the mitochondrial MT1 signal-transduction pathway inhibiting stress-mediated cytochrome c release and caspase activation. These findings coupled with our observation that mitochondrial MT1 overexpression reduces ischemic brain injury in mice delineate a mitochondrial GPCR mechanism contributing to the neuroprotective action of melatonin. We propose a new term, "automitocrine," analogous to "autocrine" when a similar phenomenon occurs at the cellular level, to describe this unexpected intracellular organelle ligand-receptor pathway that opens a new research avenue investigating mitochondrial GPCR biology.


Asunto(s)
Lesiones Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Melatonina/biosíntesis , Mitocondrias/metabolismo , Receptor de Melatonina MT1/metabolismo , Transducción de Señal , Animales , Lesiones Encefálicas/genética , Isquemia Encefálica/genética , Citocromos c/genética , Citocromos c/metabolismo , Masculino , Melatonina/genética , Ratones , Mitocondrias/genética , Receptor de Melatonina MT1/genética
6.
J Mol Cell Cardiol ; 126: 50-59, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30448480

RESUMEN

Inflammation is critical in the pathobiology of atherosclerosis. An essential player in the inflammatory process in atherosclerosis are macrophages that scavenge oxidatively modified low-density lipoproteins (OxLDL) deposited in the subendothelium of systemic arteries that secrete a myriad of pro-inflammatory mediators. Here, we identified that a subunit of the Skp-Cullin-F-box ubiquitin E3 ligase apparatus, termed FBXO3, modulates the inflammatory response in atherosclerosis. Specifically, individuals with a hypofunctioning genetic variant of FBXO3 develop less atherosclerosis. FBXO3 protein is present in cells of monocytic lineage within carotid plaques and its levels increase in those with symptomatic compared with asymptomatic atherosclerosis. Further, cellular depletion or small molecule inhibition of FBXO3 significantly reduced the inflammatory response to OxLDL by macrophages without altering OxLDL uptake. Thus, FBXO3 potentiates vascular inflammation and atherosclerosis that can be effectively mitigated by a small molecule inhibitor.


Asunto(s)
Aterosclerosis/enzimología , Inflamación/enzimología , Ubiquitina-Proteína Ligasas/metabolismo , Adulto , Anciano , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/patología , Endocitosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas F-Box/genética , Femenino , Variación Genética , Humanos , Lipoproteínas LDL/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Bibliotecas de Moléculas Pequeñas/farmacología , Células THP-1
7.
Neurobiol Dis ; 105: 156-163, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28461247

RESUMEN

Modulation of miRNA expression has been shown to be beneficial in the context of multiple diseases. The purpose of this study was to determine if an inhibitor of miR-337-3p is neuroprotective for hypoxic injury after tail vein injection. We evaluated miR-337-3p expression levels and in brain tissue in vivo before and after permanent middle cerebral artery occlusion (pMCAO) in mice. Subsequently, a custom locked nucleic acid (LNA) antimir-337-3p oligonucleotide was developed and tested in vitro after induction of oxygen glucose-deprivation (OGD) and in vivo by injection into the mouse tail vein for 3 consecutive days before pMCAO. Ischemic lesion volume was measured by TTC staining. We show that systemically administered LNA antimir-337-3p crosses the blood brain-brain-barrier (BBB), penetrates into neurosn, downregulates endogenous miR-337-3p expression and reduces ischemic brain injury. The findings support the use of similar antimir-LNA constructs as novel therapies in neurological disease.


Asunto(s)
Anticuerpos/administración & dosificación , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/etiología , Infarto de la Arteria Cerebral Media/complicaciones , MicroARNs/metabolismo , Análisis de Varianza , Animales , Presión Sanguínea/efectos de los fármacos , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Células Cultivadas , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Glucosa/deficiencia , Hipoxia/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Examen Neurológico , Neuronas/efectos de los fármacos , Oligonucleótidos/uso terapéutico , Factores de Tiempo
9.
Neurobiol Dis ; 55: 26-35, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23537713

RESUMEN

Caspase-mediated cell death contributes to the pathogenesis of motor neuron degeneration in the mutant SOD1(G93A) transgenic mouse model of amyotrophic lateral sclerosis (ALS), along with other factors such as inflammation and oxidative damage. By screening a drug library, we found that melatonin, a pineal hormone, inhibited cytochrome c release in purified mitochondria and prevented cell death in cultured neurons. In this study, we evaluated whether melatonin would slow disease progression in SOD1(G93A) mice. We demonstrate that melatonin significantly delayed disease onset, neurological deterioration and mortality in ALS mice. ALS-associated ventral horn atrophy and motor neuron death were also inhibited by melatonin treatment. Melatonin inhibited Rip2/caspase-1 pathway activation, blocked the release of mitochondrial cytochrome c, and reduced the overexpression and activation of caspase-3. Moreover, for the first time, we determined that disease progression was associated with the loss of both melatonin and the melatonin receptor 1A (MT1) in the spinal cord of ALS mice. These results demonstrate that melatonin is neuroprotective in transgenic ALS mice, and this protective effect is mediated through its effects on the caspase-mediated cell death pathway. Furthermore, our data suggest that melatonin and MT1 receptor loss may play a role in the pathological phenotype observed in ALS. The above observations indicate that melatonin and modulation of Rip2/caspase-1/cytochrome c or MT1 pathways may be promising therapeutic approaches for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Antioxidantes/uso terapéutico , Muerte Celular/efectos de los fármacos , Muerte Celular/ética , Melatonina/uso terapéutico , Transducción de Señal/efectos de los fármacos , Esclerosis Amiotrófica Lateral/genética , Análisis de Varianza , Animales , Caspasa 3/metabolismo , Citocromos c/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Ratones , Ratones Transgénicos , Receptor de Melatonina MT1/metabolismo , Superóxido Dismutasa/genética
10.
Respir Res ; 14: 119, 2013 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-24499207

RESUMEN

BACKGROUND: The ability of chemicals to disrupt neonatal development can be studied using embryonic stem cells (ESC). One such chemical is nicotine. Prenatal nicotine exposure is known to affect postnatal lung function, although the mechanisms by which it has this effect are not clear. Since fibroblasts are a critical component of the developing lung, providing structure and secreting paracrine factors that are essential to epithelialization, this study focuses on the differentiation of ESC into fibroblasts using a directed differentiation protocol. METHODS: Fibroblasts obtained from non-human primate ESC (nhpESC) differentiation were analyzed by immunohistochemistry, immunostaining, Affymetrix gene expression array, qPCR, and immunoblotting. RESULTS: Results of these analyses demonstrated that although nhpESCs differentiate into fibroblasts in the presence of nicotine and appear normal by some measures, including H&E and SMA staining, they have an altered gene expression profile. Network analysis of expression changes demonstrated an over-representation of cell-cycle related genes with downregulation of N-myc as a central regulator in the pathway. Further investigation demonstrated that cells differentiated in the presence of nicotine had decreased N-myc mRNA and protein expression and longer doubling times, a biological effect consistent with downregulation of N-myc. CONCLUSIONS: This study is the first to use primate ESC to demonstrate that nicotine can affect cellular differentiation from pluripotency into fibroblasts, and in particular, mediate N-myc expression in differentiating ESCs. Given the crucial role of fibroblasts throughout the body, this has important implications for the effect of cigarette smoke exposure on human development not only in the lung, but in organogenesis in general.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/patología , Nicotina/farmacología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Línea Celular , Células Cultivadas , Células Madre Embrionarias/efectos de los fármacos , Fibroblastos/patología , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas In Vitro , Pulmón/embriología , Pulmón/metabolismo , Pulmón/patología , Modelos Animales , Primates , Proteínas Proto-Oncogénicas c-myc/genética , ARN Mensajero/metabolismo , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo
11.
Neurosci Lett ; 795: 137043, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36586530

RESUMEN

Microglial activation is readily detected following cerebral ischemia/reperfusion-induced injury. Activated microglia polarize into either classic pro-inflammatory M1 or protective M2 microglia following ischemia/reperfusion-induced injury. Melatonin is protective immediately after ischemia/reperfusion-induced brain injury. However, the ability of melatonin to affect longer-term recovery from ischemic/reperfusion-induced injury as well as its ability to modulate microglia/macrophage polarization are unknown. The goal of this study is to understand the impact of melatonin on mice 14 days after injury, as well as to understand how melatonin affects microglial polarization of neuronal MT1 activation following cerebral ischemia/reperfusion. We utilized NSEMT1-GFP transgenic mice which overexpress MT1 (melatonin type 1 receptor) in neurons. Melatonin-treated or vehicle treated wild type and NSEMT1-GFP mice underwent middle cerebral artery occlusion (MCAO)/reperfusion and followed for 14 days. Neuronal MT1 overexpression significantly reduced infarct volumes, improved motor function, and ameliorated weight loss. Additionally, melatonin treatment reduced infarct volume in NSEMT1-GFP mice as compared to untreated wild type, melatonin treated wild type, and untreated NSEMT1-GFP mice. Melatonin improved neurological function and prevented weight loss in NSEMT1-GFP mice compared with melatonin treated wild type mice. Finally, melatonin treatment in combination with MT1 overexpression reduced the numbers of Iba1+/CD16+ M1 microglia and increased the numbers of Iba1+/ CD206+ M2 microglia after ischemic injury. In conclusion, neuronal MT1 mediates melatonin-induced long-term recovery after cerebral ischemia, at least in part, by shifting microglial polarization toward the neuroprotective M2 phenotype.


Asunto(s)
Isquemia Encefálica , Melatonina , Daño por Reperfusión , Ratones , Animales , Microglía/fisiología , Melatonina/farmacología , Isquemia Encefálica/prevención & control , Infarto de la Arteria Cerebral Media , Receptores de Melatonina , Reperfusión , Neuronas
12.
Sci Rep ; 11(1): 18916, 2021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34556702

RESUMEN

Amyotrophic lateral sclerosis is an adult-onset neurodegenerative disorder characterized by loss of motor neurons. Mitochondria are essential for neuronal survival but the developmental timing and mechanistic importance of mitochondrial dysfunction in sporadic ALS (sALS) neurons is not fully understood. We used human induced pluripotent stem cells and generated a developmental timeline by differentiating sALS iPSCs to neural progenitors and to motor neurons and comparing mitochondrial parameters with familial ALS (fALS) and control cells at each developmental stage. We report that sALS and fALS motor neurons have elevated reactive oxygen species levels, depolarized mitochondria, impaired oxidative phosphorylation, ATP loss and defective mitochondrial protein import compared with control motor neurons. This phenotype develops with differentiation into motor neurons, the affected cell type in ALS, and does not occur in the parental undifferentiated sALS cells or sALS neural progenitors. Our work demonstrates a developmentally regulated unifying mitochondrial phenotype between patient derived sALS and fALS motor neurons. The occurrence of a unifying mitochondrial phenotype suggests that mitochondrial etiology known to SOD1-fALS may applicable to sALS. Furthermore, our findings suggest that disease-modifying treatments focused on rescue of mitochondrial function may benefit both sALS and fALS patients.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Diferenciación Celular , Mitocondrias/patología , Neuronas Motoras/patología , Células-Madre Neurales/patología , Biopsia , Células Cultivadas , Fibroblastos , Humanos , Células Madre Pluripotentes Inducidas , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Células-Madre Neurales/citología , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Piel/patología
13.
Neuroreport ; 31(3): 235-239, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31876686

RESUMEN

MiR-155 negatively regulates translation of mRNA targets to proteins involved in processes that modulate ischemic brain injury including neuroinflammation, blood-brain barrier (BBB) permeability, and apoptosis. However, reports of the effect of cerebral miR-155 expression changes after ischemic brain injury are equivocal and miR-155 modulates molecular pathways with opposing effects on these processes. The role of miR-155 in postischemic cerebral hemorrhagic transformation remains unknown. To understand the net effect of complete inactivation of miR-155, miR-155 knockout mice were studied in a cerebral ischemia/reperfusion (I/R) model of infarction and hemorrhagic transformation as compared with those of wild type mice. Wild type and miR-155 knockout mice underwent one hour of middle cerebral artery occlusion (MCAO) followed by up to 71 hours of reperfusion. The effects of miR-155 knockout on cerebral infarct size, incidence and extent of hemorrhagic transformation, and neurological outcome were determined. We found that miR-155 was significantly upregulated after cerebral I/R in wild type mice, and miR-155 knockout mice had comparably smaller cerebral infarct size and improved neurological deficits. Similarly, wild type mice had significant hemorrhagic burden after cerebral I/R, the incidence and volume of which was reduced in miR-155 knockout mice. Although miR-155 can have opposite effects on cerebral I/R-injury-related processes, the net effect of miR-155 knockout is neuroprotective. Thus, the increase in miR-155 expression observed after cerebral I/R may be considered deleterious and inhibition of this expression and its effects a potential therapeutic target.


Asunto(s)
Isquemia Encefálica/metabolismo , Hemorragia Cerebral/metabolismo , MicroARNs/metabolismo , Daño por Reperfusión/metabolismo , Animales , Isquemia Encefálica/complicaciones , Hemorragia Cerebral/etiología , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión/complicaciones
14.
J Clin Invest ; 130(6): 3124-3136, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32182222

RESUMEN

Chronic inflammation is a pathologic feature of neurodegeneration and aging; however, the mechanism regulating this process is not understood. Melatonin, an endogenous free radical scavenger synthesized by neuronal mitochondria, decreases with aging and neurodegeneration. We proposed that insufficient melatonin levels impair mitochondrial homeostasis, resulting in mitochondrial DNA (mtDNA) release and activation of cytosolic DNA-mediated inflammatory response in neurons. We found increased mitochondrial oxidative stress and decreased mitochondrial membrane potential, with higher mtDNA release in brain and primary cerebro-cortical neurons of melatonin-deficient aralkylamine N-acetyltransferase (AANAT) knockout mice. Cytosolic mtDNA activated the cGAS/STING/IRF3 pathway, stimulating inflammatory cytokine generation. We found that Huntington's disease mice had increased mtDNA release, cGAS activation, and inflammation, all inhibited by exogenous melatonin. Thus, we demonstrated that cytosolic mtDNA activated the inflammatory response in aging and neurodegeneration, a process modulated by melatonin. Furthermore, our data suggest that AANAT knockout mice are a model of accelerated aging.


Asunto(s)
Envejecimiento/metabolismo , Citosol/metabolismo , ADN Mitocondrial/metabolismo , Enfermedad de Huntington/metabolismo , Melatonina/farmacología , Neuronas/metabolismo , Transducción de Señal/efectos de los fármacos , Envejecimiento/genética , Envejecimiento/patología , Animales , Citosol/patología , ADN Mitocondrial/genética , Femenino , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Noqueados , Neuronas/patología
15.
J Huntingtons Dis ; 8(4): 449-457, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31594239

RESUMEN

BACKGROUND: Selective serotonin reuptake inhibitors (SSRIs) target the serotonin transporter (SERT) and are commonly prescribed for depression in Huntington's disease (HD) patients. However, SERT expression in HD has not been carefully evaluated in patients or mouse models. OBJECTIVE: In this study, we investigated SERT levels in HD patients and HD mouse models. METHODS: We obtained HD patient brain striatal samples and matched controls, as well as brain tissue from CAG140 and R6/2 mice. SERT mRNA and protein levels were analyzed using quantitative RT-PCR and immunoblotting. RESULTS AND CONCLUSIONS: We found that SERT protein, but not mRNA is markedly increased in grade 4 HD patient striatal tissue. These findings suggest posttranscriptional or translational SERT dysregulation as a possible etiologic factor modulating psychopathology in HD. Interestingly, SERT expression is variable in mouse models of the disease. Increased SERT levels are demonstrated in the brain of CAG140 mice, a full-length knock-in mouse model of the disease, but not in the striatum of the R6/2 fragment murine model of the disease. Based on this parameter, the CAG140 huntingtin knock-in mouse model is more suitable than the R6/2 model for the study of serotonergic pathway pathology in Huntington's disease.


Asunto(s)
Cuerpo Estriado/metabolismo , Enfermedad de Huntington/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , ARN Mensajero/metabolismo
16.
Neurosurgery ; 85(2): E284-E293, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30335165

RESUMEN

BACKGROUND: Many studies have explored molecular markers of carotid plaque development and vulnerability to rupture, usually having examined whole carotid plaques. However, there are regional differences in plaque morphology and known shear-related mechanisms in areas surrounding the lipid core. OBJECTIVE: To determine whether there are regional differences in protein expression along the long axis of the carotid plaque and how that might produce gaps in our understanding of the carotid plaque molecular signature. METHODS: Levels of 7 inflammatory cytokines (IL-1ß, IL-6, IL-8, IL-10, IL-12 p70, IFN-γ, and TNF-α) and caspase-3 were analyzed in prebifurcation, bifurcation, and postbifurcation segments of internal carotid plaques surgically removed from symptomatic and asymptomatic patients. Expression profiles of miRNAs and mRNAs were determined with microarrays for the rupture-prone postbifurcation segment for comparison with published whole plaque results. RESULTS: Expression levels of all proteins examined, except IL-10, were lowest in the prebifurcation segment and significantly higher in the postbifurcation segment. Patient group differences in protein expression were observed for the prebifurcation segment; however, no significant differences were observed in the postbifurcation segment between symptomatic and asymptomatic patients. Expression profiles from postbifurcation carotid plaques identified 4 novel high priority miRNAs differentially expressed between patient groups (miR-214, miR-484, miR-942, and miR-1287) and 3 high-confidence miRNA:mRNA targets, including miR-214:APOD, miR-484:DACH1, and miR-942:GPR56. CONCLUSION: The results demonstrate regional differences in protein expression for the first time and show that focus on the rupture-prone postbifurcation region leads to prioritization for further study of novel miRNA gene regulation mechanisms.


Asunto(s)
Citocinas/metabolismo , Proteínas del Ojo/metabolismo , Placa Aterosclerótica/metabolismo , Anciano , Anciano de 80 o más Años , Biomarcadores/metabolismo , Estenosis Carotídea/genética , Caspasa 3/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Interferón gamma/metabolismo , Interleucinas/metabolismo , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Factores de Transcripción , Factor de Necrosis Tumoral alfa/metabolismo
17.
Biomaterials ; 180: 225-239, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30053658

RESUMEN

The chronic performance of implantable neural electrodes is hindered by inflammatory brain tissue responses, including microglia activation, glial scarring, and neuronal loss. Melatonin (MT) has shown remarkable neuroprotective and neurorestorative effects in treating central nervous system (CNS) injuries and degeneration by inhibiting caspase-1, -3, and -9 activation and mitochondrial cytochrome c release, as well as reducing oxidative stress and neuroinflammation. This study examined the effect of MT administration on the quality and longevity of neural recording from an implanted microelectrode in the visual cortex of mice for 16 weeks. MT (30 mg/kg) was administered via daily intraperitoneal injection for acute (3 days before and 14 days post-implantation) and chronic (3 days before and 16 weeks post-implantation) exposures. During the first 4 weeks, both MT groups showed significantly higher single-unit (SU) yield, signal-to-noise ratio (SNR), and amplitude compared to the vehicle control group. However, after 4 weeks of implantation, the SU yield of the acute treatment group dropped to the same level as the control group, while the chronic treatment group maintained significantly higher SU yield compared to both acute (week 5-16) and control (week 0-16) mice. Histological studies revealed a significant increase in neuronal viability and decrease in neuronal apoptosis around the implanted electrode at week 16 in the chronic group in comparison to control and acute subjects, which is correlated with reduced oxidative stress and increased number of pro-regeneration arginase-1 positive microglia cells. These results demonstrate the potent effect of MT treatment in maintaining a high-quality electrode-tissue interface and suggest that MT promotes neuroprotection possibly through its anti-apoptotic, anti-inflammatory, and anti-oxidative properties.


Asunto(s)
Gliosis/metabolismo , Melatonina/farmacología , Animales , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Gliosis/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Masculino , Melatonina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Microelectrodos , Estrés Oxidativo/efectos de los fármacos
18.
J Neurosci Methods ; 263: 1-6, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26808294

RESUMEN

BACKGROUND: Functional and structural properties of mitochondria are highly tissue and cell dependent, but isolation of highly purified human neuronal mitochondria is not currently available. NEW METHOD: We developed and validated a procedure to isolate purified neuronal mitochondria from brain tissue. The method combines Percoll gradient centrifugation to obtain synaptosomal fraction with nitrogen cavitation mediated synaptosome disruption and extraction of mitochondria using anti mitochondrial outer membrane protein antibodies conjugated to magnetic beads. The final products of isolation are non-synaptosomal mitochondria, which are a mixture of mitochondria isolated from different brain cells (i.e. neurons, astrocytes, oligodendrocytes, microglia) and synaptic mitochondria, which are of neuronal origin. This method is well suited for preparing functional mitochondria from human cortex tissue that is surgically extracted. RESULTS: The procedure produces mitochondria with minimal cytoplasmic contaminations that are functionally active based on measurements of mitochondrial respiration as well as mitochondrial protein import. The procedure requires approximately four hours for the isolation of human neuronal mitochondria and can also be used to isolate mitochondria from mouse/rat/monkey brains. COMPARISON WITH EXISTING METHODS AND CONCLUSIONS: This method will allow researchers to study highly enriched neuronal mitochondria without the confounding effect of cellular and organelle contaminants.


Asunto(s)
Corteza Cerebral/citología , Mitocondrias/fisiología , Neuronas/ultraestructura , Anticuerpos/metabolismo , Fraccionamiento Celular , Antígenos HLA/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/inmunología , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteínas Mitocondriales/metabolismo , Sinaptosomas/metabolismo , Sinaptosomas/ultraestructura
20.
Clin Cancer Res ; 8(8): 2684-9, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12171901

RESUMEN

Human solid tumors frequently have a relatively small growth fraction,which interferes with the action of many chemotherapeutic agents that target actively cycling cells. Several polyamine analogues are currently being developed for clinical application against human solid tumors including N1,N11-bis(ethyl)norspermine. Therefore, an effort was made to examine the effects of growth rate on polyamine-analogue efficacy. Low growth fraction (LGF) cell cultures of the human non-small cell lung cancer cell line NCI-H157 were generated to partially mimic solid tumors with low mitotic indices. Log-phase cells were compared with LGF cells with respect to cell survival and biochemical effects after exposure to polyamine analogues. The results demonstrate generally that LGF NCI-H157 cells were sensitive to analogue treatment. However, the dose necessary to elicit a response in LGF cells was an order of magnitude higher than the dose needed in log-phase cells. Additionally, the biochemical effects of analogues were similar between log phase and LGF cells with regard to a down-regulation of polyamine biosynthesis as measured by ornithine decarboxylase activity and an increase in polyamine catabolism as indicated by an increase in spermidine/spermine N1-acetyltransferase activity. However, biochemical effects were less dramatic in the LGF cells than those observed in the log-phase cells. The overall results of these studies suggest that the growth status of solid tumors can significantly affect the response to antitumor polyamine analogues, and growth fraction must be considered in the continued development and use of the polyamine analogues.


Asunto(s)
Neoplasias Pulmonares/tratamiento farmacológico , Poliaminas/uso terapéutico , División Celular , Separación Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Humanos , Poliaminas/química , Poliaminas/metabolismo , Factores de Tiempo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA