Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
FASEB J ; 33(10): 11270-11283, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31307210

RESUMEN

Ileal interposition (IT) is a surgical procedure that increases the delivery of incompletely digested nutrients and biliary and pancreatic secretions to the distal intestinal mucosa. Here, we investigated the metabolic impact of this intervention in 2-mo-old prediabetic University of California, Davis type 2 diabetes mellitus rats by assessing liver gene expression at 1.5 mo post-IT surgery. Pathway analysis indicated decreased signaling via TGF-ß/Smad (a family of proteins named mothers against decapentaplegic homologs), peroxisome proliferator-activated receptor (PPAR), and PI3K-Akt-AMPK-mechanistic target of rapamycin, likely targeting hepatic stellate cells because differentiation and activation of these cells is associated with decreased signaling via PPAR and TGF-ß/Smad. IT surgery up-regulated the expression of genes involved in regulation of cholesterol and terpenoid syntheses and down-regulated those involved in glycerophospholipid metabolism [including cardiolipin (CL)], lipogenesis, and gluconeogenesis. Consistent with the down-regulation of the hepatic CL pathway, IT surgery produced a metabolic switch in liver, kidney cortex, and fat depots toward decreased mitochondrial fatty acid ß-oxidation, the process required to fuel high energy-demanding pathways (e.g., gluconeogenesis and glyceroneogenesis), whereas opposite effects were observed in skeletal and cardiac muscles. This study demonstrates for the first time the presence of metabolic pathways that complement the effects of IT surgery to maximize its benefits and potentially identify similarly effective, durable, and less invasive therapeutic options for metabolic disease, including inhibitors of TGF-ß signaling.-Hung, C., Napoli, E., Ross-Inta, C., Graham, J., Flores-Torres, A. L., Stanhope, K. L., Froment, P., Havel, P. J., Giulivi, C. Ileal interposition surgery targets the hepatic TGF-ß pathway, influencing gluconeogenesis and mitochondrial bioenergetics in the UCD-T2DM rat model of diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/fisiología , Gluconeogénesis/fisiología , Íleon/metabolismo , Hígado/metabolismo , Mitocondrias/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Íleon/fisiopatología , Insulina/metabolismo , Metabolismo de los Lípidos/fisiología , Hígado/fisiopatología , Masculino , Mitocondrias/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Proteínas Smad/metabolismo
2.
Hum Mol Genet ; 22(5): 989-1004, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23197653

RESUMEN

A number of studies have been conducted that link mitochondrial dysfunction (MD) to Huntington's disease (HD); however, contradicting results had resulted in a lack of a clear mechanism that links expression of mutant Huntingtin protein and MD. Mouse homozygous (HM) and heterozygous (HT) mutant striatal cells with two or one allele encoding for a mutant huntingtin protein with 111 polyGln repeats showed a significant impairment of the mitochondrial disulfide relay system (MDRS). This system (consisting of two proteins, Gfer and Mia40) is involved in the mitochondrial import of Cys-rich proteins. The Gfer-to-Mia40 ratio was significantly altered in HM cells compared with controls, along with the expression of mitochondrial proteins considered substrates of the MDRS. In progenitors and differentiated neuron-like HM cells, impairment of MDRS were accompanied by deficient oxidative phosphorylation, Complex I, IV and V activities, decreased mtDNA copy number and transcripts, accumulation of mtDNA deletions and changes in mitochondrial morphology, consistent with other MDRS-deficient biological models, thus providing a framework for the energy deficits observed in this HD model. The majority (>90%) of the mitochondrial outcomes exhibited a gene-dose dependency with the expression of mutant Htt. Finally, decreases in the mtDNA copy number, along with the accumulation of mtDNA deletions, provide a mechanism for the progressive neurodegeneration observed in HD patients.


Asunto(s)
Enfermedad de Huntington/genética , Mitocondrias/genética , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Proteínas Nucleares/genética , Animales , Diferenciación Celular , Cuerpo Estriado , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/fisiopatología , Masculino , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Mutación , Neuronas/citología , Fosforilación Oxidativa , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Expansión de Repetición de Trinucleótido/genética
3.
J Neurochem ; 127(4): 520-30, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24024616

RESUMEN

The anterior piriform cortex (APC) is activated by, and is the brain area most sensitive to, essential (indispensable) amino acid (IAA) deficiency. The APC is required for the rapid (20 min) behavioral rejection of IAA deficient diets and increased foraging, both crucial adaptive functions supporting IAA homeostasis in omnivores. The biochemical mechanisms signaling IAA deficiency in the APC block initiation of translation in protein synthesis via uncharged tRNA and the general amino acid control kinase, general control nonderepressing kinase 2. Yet, how inhibition of protein synthesis activates the APC is unknown. The neuronal K(+) Cl(-) cotransporter, neural potassium chloride co-transporter (KCC2), and GABAA receptors are essential inhibitory elements in the APC with short plasmalemmal half-lives that maintain control in this highly excitable circuitry. After a single IAA deficient meal both proteins were reduced (vs. basal diet controls) in western blots of APC (but not neocortex or cerebellum) and in immunohistochemistry of APC. Furthermore, electrophysiological analyses support loss of inhibitory elements such as the GABAA receptor in this model. As the crucial inhibitory function of the GABAA receptor depends on KCC2 and the Cl(-) transmembrane gradient it establishes, these results suggest that loss of such inhibitory elements contributes to disinhibition of the APC in IAA deficiency. The circuitry of the anterior piriform cortex (APC) is finely balanced between excitatory (glutamate, +) and inhibitory (GABA, -) transmission. GABAA receptors use Cl(-), requiring the neural potassium chloride co-transporter (KCC2). Both are rapidly turning-over proteins, dependent on protein synthesis for repletion. In IAA (indispensable amino acid) deficiency, within 20 min, blockade of protein synthesis prevents restoration of these inhibitors; they are diminished; disinhibition ensues. GCN2 = general control non-derepressing kinase 2, eIF2α = α-subunit of the eukaryotic initiation factor 2.


Asunto(s)
Aminoácidos Esenciales/deficiencia , Vías Olfatorias/metabolismo , Receptores de GABA-A/biosíntesis , Simportadores/biosíntesis , Animales , Regulación hacia Abajo , Potenciales Postsinápticos Excitadores , Masculino , Ratas , Cotransportadores de K Cl
4.
Hum Mol Genet ; 20(15): 3079-92, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21558427

RESUMEN

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder that affects individuals who are carriers of small CGG premutation expansions in the fragile X mental retardation 1 (FMR1) gene. Mitochondrial dysfunction was observed as an incipient pathological process occurring in individuals who do not display overt features of FXTAS (1). Fibroblasts from premutation carriers had lower oxidative phosphorylation capacity (35% of controls) and Complex IV activity (45%), and higher precursor-to-mature ratios (P:M) of nDNA-encoded mitochondrial proteins (3.1-fold). However, fibroblasts from carriers with FXTAS symptoms presented higher FMR1 mRNA expression (3-fold) and lower Complex V (38%) and aconitase activities (43%). Higher P:M of ATPase ß-subunit (ATPB) and frataxin were also observed in cortex from patients that died with FXTAS symptoms. Biochemical findings observed in FXTAS cells (lower mature frataxin, lower Complex IV and aconitase activities) along with common phenotypic traits shared by Friedreich's ataxia and FXTAS carriers (e.g. gait ataxia, loss of coordination) are consistent with a defective iron homeostasis in both diseases. Higher P:M, and lower ZnT6 and mature frataxin protein expression suggested defective zinc and iron metabolism arising from altered ZnT protein expression, which in turn impairs the activity of mitochondrial Zn-dependent proteases, critical for the import and processing of cytosolic precursors, such as frataxin. In support of this hypothesis, Zn-treated fibroblasts showed a significant recovery of ATPB P:M, ATPase activity and doubling time, whereas Zn and desferrioxamine extended these recoveries and rescued Complex IV activity.


Asunto(s)
Síndrome del Cromosoma X Frágil/metabolismo , Transporte Iónico/fisiología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Zinc/metabolismo , Anciano , Western Blotting , Células Cultivadas , ADN Mitocondrial/genética , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/genética , Dosificación de Gen/genética , Humanos , Peróxido de Hidrógeno/metabolismo , Técnicas In Vitro , Transporte Iónico/genética , Masculino
5.
J Neurosci ; 31(5): 1583-90, 2011 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-21289166

RESUMEN

Protein synthesis requires a continuous supply of all of the indispensable (essential) amino acids (IAAs). If any IAA is deficient, animals must obtain the limiting amino acid by diet selection. Sensing of IAA deficiency requires an intact anterior piriform cortex (APC), but does it act alone? Shortly after rats begin eating an IAA-deficient diet, the meal ends and EPSPs are activated in the APC; from there, neurons project to feeding circuits; the meal ends within 20 min. Within the APC in vivo, uncharged tRNA activates the general amino acid control non-derepressing 2 (GCN2) enzyme system increasing phosphorylation of eukaryotic initiation factor (P-eIF2α), which blocks general protein synthesis. If this paleocortex is sufficient for sensing IAA depletion, both neuronal activation and P-eIF2α should occur in an isolated APC slice. We used standard techniques for electrophysiology and immunohistochemistry. After rats ate IAA-devoid or -imbalanced diets, their depleted slices responded to different stimuli with increased EPSP amplitudes. Slices from rats fed a control diet were bathed in artificial CSF replete with all amino acids with or without the IAA, threonine, or a tRNA synthetase blocker, l-threoninol, or its inactive isomer, d-threoninol. Thr depletion in vitro increased both EPSP amplitudes and P-eIF2α. l (but not d)-threoninol also increased EPSP amplitudes relative to control. Thus, we show independent excitation of the APC with responses parallel to those known in vivo. These data suggest a novel idea: in addition to classical processing of peripheral sensory input, direct primary sensing may occur in mammalian cortex.


Asunto(s)
Aminoácidos Esenciales/deficiencia , Corteza Cerebral/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Neuronas/metabolismo , Biosíntesis de Proteínas , Proteínas Quinasas/metabolismo , Aminoácidos Esenciales/biosíntesis , Aminoácidos Esenciales/metabolismo , Animales , Corteza Cerebral/fisiología , Dieta , Electrofisiología , Inmunohistoquímica , Masculino , Neuronas/fisiología , Técnicas de Cultivo de Órganos , Fosforilación , Ratas , Ratas Sprague-Dawley , Treonina/deficiencia
6.
J Biol Chem ; 286(1): 99-113, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-20978128

RESUMEN

Malignant hyperthermia (MH) and central core disease in humans have been associated with mutations in the skeletal ryanodine receptor (RyR1). Heterozygous mice expressing the human MH/central core disease RyR1 R163C mutation exhibit MH when exposed to halothane or heat stress. Considering that many MH symptoms resemble those that could ensue from a mitochondrial dysfunction (e.g. metabolic acidosis and hyperthermia) and that MH-susceptible mice or humans have a higher than normal cytoplasmic Ca(2+) concentration at rest, we evaluated the role of mitochondria in skeletal muscle from R163C compared with wild type mice under basal (untriggered) conditions. R163C skeletal muscle exhibited a significant increase in matrix Ca(2+), increased reactive oxygen species production, lower expression of mitochondrial proteins, and higher mtDNA copy number. These changes, in conjunction with lower myoglobin and glycogen contents, Myh4 and GAPDH transcript levels, GAPDH activity, and lower glucose utilization suggested a switch to a compromised bioenergetic state characterized by both low oxidative phosphorylation and glycolysis. The shift in bioenergetic state was accompanied by a dysregulation of Ca(2+)-responsive signaling pathways regulated by calcineurin and ERK1/2. Chronically elevated resting Ca(2+) in R163C skeletal muscle elicited the maintenance of a fast-twitch fiber program and the development of insulin resistance-like phenotype as part of a metabolic adaptation to the R163C RyR1 mutation.


Asunto(s)
Metabolismo Basal/genética , Técnicas de Sustitución del Gen , Predisposición Genética a la Enfermedad/genética , Hipertermia Maligna/metabolismo , Músculo Esquelético/metabolismo , Mutación , Canal Liberador de Calcio Receptor de Rianodina/genética , Animales , Calcineurina/metabolismo , Calcio/metabolismo , Diafragma/metabolismo , Diafragma/fisiopatología , Femenino , Regulación de la Expresión Génica , Humanos , Cinética , Hipertermia Maligna/genética , Hipertermia Maligna/patología , Hipertermia Maligna/fisiopatología , Ratones , Mitocondrias/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Estrés Oxidativo/genética , Oxígeno/metabolismo , Permeabilidad , Ratas , Transducción de Señal/genética
7.
Toxicol Appl Pharmacol ; 262(2): 194-204, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22583949

RESUMEN

Current medical countermeasures against organophosphate (OP) nerve agents are effective in reducing mortality, but do not sufficiently protect the CNS from delayed brain damage and persistent neurological symptoms. In this study, we examined the efficacy of neuregulin-1 (NRG-1) in protecting against delayed neuronal cell death following acute intoxication with the OP diisopropylflurophosphate (DFP). Adult male Sprague-Dawley rats were pretreated with pyridostigmine (0.1 mg/kg BW, i.m.) and atropine methylnitrate (20 mg/kg BW, i.m.) prior to DFP (9 mg/kg BW, i.p.) intoxication to increase survival and reduce peripheral signs of cholinergic toxicity but not prevent DFP-induced seizures or delayed neuronal injury. Pretreatment with NRG-1 did not protect against seizures in rats exposed to DFP. However, neuronal injury was significantly reduced in most brain regions by pretreatment with NRG-1 isoforms NRG-EGF (3.2 µg/kg BW, i.a) or NRG-GGF2 (48 µg/kg BW, i.a.) as determined by FluroJade-B labeling in multiple brain regions at 24 h post-DFP injection. NRG-1 also blocked apoptosis and oxidative stress-mediated protein damage in the brains of DFP-intoxicated rats. Administration of NRG-1 at 1h after DFP injection similarly provided significant neuroprotection against delayed neuronal injury. These findings identify NRG-1 as a promising adjuvant therapy to current medical countermeasures for enhancing neuroprotection against acute OP intoxication.


Asunto(s)
Encéfalo/efectos de los fármacos , Inhibidores de la Colinesterasa/toxicidad , Isoflurofato/toxicidad , Neurregulina-1/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Convulsiones/prevención & control , Animales , Atropina/farmacología , Encéfalo/citología , Encéfalo/metabolismo , Inmunohistoquímica , Masculino , Neuronas/metabolismo , Isoformas de Proteínas , Bromuro de Piridostigmina/farmacología , Ratas , Ratas Sprague-Dawley , Convulsiones/metabolismo
8.
Am J Pathol ; 177(4): 1958-68, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20724589

RESUMEN

Citrullinemia type I (CTLN1, OMIM# 215700) is an inherited urea cycle disorder that is caused by an argininosuccinate synthetase (ASS) enzyme deficiency. In this report, we describe two spontaneous hypomorphic alleles of the mouse Ass1 gene that serve as an animal model of CTLN1. These two independent mouse mutant alleles, also described in patients affected with CTLN1, interact to produce a range of phenotypes. While some mutant mice died within the first week after birth, others survived but showed severe retardation during postnatal development as well as alopecia, lethargy, and ataxia. Notable pathological findings were similar to findings in human CTLN1 patients and included citrullinemia and hyperammonemia along with delayed cerebellar development, epidermal hyperkeratosis, and follicular dystrophy. Standard treatments for CTLN1 were effective in rescuing the phenotype of these mutant mice. Based on our studies, we propose that defective cerebellar granule cell migration secondary to disorganization of Bergmann glial cell fibers cause cerebellar developmental delay in the hyperammonemic and citrullinemic brain, pointing to a possible role for nitric oxide in these processes. These mouse mutations constitute a suitable model for both mechanistic and preclinical studies of CTLN1 and other hyperammonemic encephalopathies and, at the same time, underscore the importance of complementing knockout mutations with hypomorphic mutations for the generation of animal models of human genetic diseases.


Asunto(s)
Argininosuccinato Sintasa/fisiología , Citrulinemia/etiología , Modelos Animales de Enfermedad , Hiperamonemia/etiología , Mutación Missense/genética , Alelos , Animales , Arginina/farmacología , Western Blotting , Movimiento Celular , Cerebelo/anomalías , Citrulinemia/tratamiento farmacológico , Discapacidades del Desarrollo/tratamiento farmacológico , Discapacidades del Desarrollo/etiología , Femenino , Trastornos del Crecimiento/tratamiento farmacológico , Trastornos del Crecimiento/etiología , Humanos , Hiperamonemia/tratamiento farmacológico , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Óxido Nítrico/metabolismo , Fenotipo , Benzoato de Sodio/farmacología , Síndrome
9.
Biochem J ; 429(3): 545-52, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20513237

RESUMEN

FXTAS (fragile X-associated tremor/ataxia syndrome) is a late-onset neurodegenerative disorder that affects individuals who are carriers of premutation expansions (55-200 CGG repeats) in the 5' untranslated region of the FMR1 (fragile X mental retardation 1) gene. The role of MD (mitochondrial dysfunction) in FXTAS was evaluated in fibroblasts and brain samples from premutation carriers with and without FXTAS symptoms, with a range of CGG repeats. This study resulted in several important conclusions: (i) decreased NAD- and FAD-linked oxygen uptake rates and uncoupling between electron transport and synthesis of ATP were observed in fibroblasts from premutation carriers; (ii) a lower expression of mitochondrial proteins preceded both in age and in CGG repeats the appearance of overt clinical involvement; (iii) the CGG repeat size required for altered mitochondrial protein expression was also smaller than that required to produce brain intranuclear inclusions from individuals with the premutation who died, suggesting that MD is an incipient pathological process occurring in individuals who do not display overt features of FXTAS; and (iv) on the basis of the CGG repeats, MD preceded the increase in oxidative/nitrative stress damage, indicating that the latter is a late event. MD in carriers of small CGG repeats, even when the allele size is not sufficient to produce FXTAS, may predispose them to other disorders (e.g. Parkinson's disease) that are likely to involve MD, and to environmental stressors, which may trigger the development of FXTAS symptoms. Detection of MD is of critical importance to the management of FXTAS, since it opens up additional treatment options for this disorder.


Asunto(s)
Ataxia/fisiopatología , Síndrome del Cromosoma X Frágil/fisiopatología , Mitocondrias/fisiología , Temblor/fisiopatología , Adenosina Trifosfato/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Ataxia/complicaciones , Transporte de Electrón , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/complicaciones , Humanos , Persona de Mediana Edad , Mutación , Biosíntesis de Proteínas , Temblor/complicaciones
10.
JAMA ; 304(21): 2389-96, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21119085

RESUMEN

CONTEXT: Impaired mitochondrial function may influence processes highly dependent on energy, such as neurodevelopment, and contribute to autism. No studies have evaluated mitochondrial dysfunction and mitochondrial DNA (mtDNA) abnormalities in a well-defined population of children with autism. OBJECTIVE: To evaluate mitochondrial defects in children with autism. DESIGN, SETTING, AND PATIENTS: Observational study using data collected from patients aged 2 to 5 years who were a subset of children participating in the Childhood Autism Risk From Genes and Environment study in California, which is a population-based, case-control investigation with confirmed autism cases and age-matched, genetically unrelated, typically developing controls, that was launched in 2003 and is still ongoing. Mitochondrial dysfunction and mtDNA abnormalities were evaluated in lymphocytes from 10 children with autism and 10 controls. MAIN OUTCOME MEASURES: Oxidative phosphorylation capacity, mtDNA copy number and deletions, mitochondrial rate of hydrogen peroxide production, and plasma lactate and pyruvate. RESULTS: The reduced nicotinamide adenine dinucleotide (NADH) oxidase activity (normalized to citrate synthase activity) in lymphocytic mitochondria from children with autism was significantly lower compared with controls (mean, 4.4 [95% confidence interval {CI}, 2.8-6.0] vs 12 [95% CI, 8-16], respectively; P = .001). The majority of children with autism (6 of 10) had complex I activity below control range values. Higher plasma pyruvate levels were found in children with autism compared with controls (0.23 mM [95% CI, 0.15-0.31 mM] vs 0.08 mM [95% CI, 0.04-0.12 mM], respectively; P = .02). Eight of 10 cases had higher pyruvate levels but only 2 cases had higher lactate levels compared with controls. These results were consistent with the lower pyruvate dehydrogenase activity observed in children with autism compared with controls (1.0 [95% CI, 0.6-1.4] nmol × [min × mg protein](-1) vs 2.3 [95% CI, 1.7-2.9] nmol × [min × mg protein](-1), respectively; P = .01). Children with autism had higher mitochondrial rates of hydrogen peroxide production compared with controls (0.34 [95% CI, 0.26-0.42] nmol × [min × mg of protein](-1) vs 0.16 [95% CI, 0.12-0.20] nmol × [min × mg protein](-1) by complex III; P = .02). Mitochondrial DNA overreplication was found in 5 cases (mean ratio of mtDNA to nuclear DNA: 239 [95% CI, 217-239] vs 179 [95% CI, 165-193] in controls; P = 10(-4)). Deletions at the segment of cytochrome b were observed in 2 cases (ratio of cytochrome b to ND1: 0.80 [95% CI, 0.68-0.92] vs 0.99 [95% CI, 0.93-1.05] for controls; P = .01). CONCLUSION: In this exploratory study, children with autism were more likely to have mitochondrial dysfunction, mtDNA overreplication, and mtDNA deletions than typically developing children.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/fisiopatología , ADN Mitocondrial/genética , Enfermedades Mitocondriales/fisiopatología , Estudios de Casos y Controles , Preescolar , Variaciones en el Número de Copia de ADN , Femenino , Eliminación de Gen , Humanos , Peróxido de Hidrógeno/metabolismo , Ácido Láctico/sangre , Linfocitos , Masculino , Complejos Multienzimáticos/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Fosforilación , Ácido Pirúvico/sangre
11.
Biochem J ; 415(2): 309-16, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18588503

RESUMEN

No studies have been performed on the mitochondria of malaria vector mosquitoes. This information would be valuable in understanding mosquito aging and detoxification of insecticides, two parameters that have a significant impact on malaria parasite transmission in endemic regions. In the present study, we report the analyses of respiration and oxidative phosphorylation in mitochondria of cultured cells [ASE (Anopheles stephensi Mos. 43) cell line] from A. stephensi, a major vector of malaria in India, South-East Asia and parts of the Middle East. ASE cell mitochondria share many features in common with mammalian muscle mitochondria, despite the fact that these cells are of larval origin. However, two major differences with mammalian mitochondria were apparent. One, the glycerol-phosphate shuttle plays as major a role in NADH oxidation in ASE cell mitochondria as it does in insect muscle mitochondria. In contrast, mammalian white muscle mitochondria depend primarily on lactate dehydrogenase, whereas red muscle mitochondria depend on the malate-oxaloacetate shuttle. Two, ASE mitochondria were able to oxidize proline at a rate comparable with that of alpha-glycerophosphate. However, the proline pathway appeared to differ from the currently accepted pathway, in that oxoglutarate could be catabolized completely by the tricarboxylic acid cycle or via transamination, depending on the ATP need.


Asunto(s)
Anopheles/metabolismo , Insectos Vectores/metabolismo , Redes y Vías Metabólicas , Mitocondrias/metabolismo , Aminoácidos/metabolismo , Animales , Anopheles/citología , Antimicina A/farmacología , Metabolismo de los Hidratos de Carbono , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/farmacología , Línea Celular , Respiración de la Célula/efectos de los fármacos , Cromatografía Liquida , Ciclo del Ácido Cítrico , Ácido Glutámico/metabolismo , Malaria/transmisión , Malatos/metabolismo , Oligomicinas/farmacología , Oxidación-Reducción , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno , Ácido Pirúvico/metabolismo , Espectrometría de Masas en Tándem
12.
Biosci Rep ; 28(5): 239-49, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18636966

RESUMEN

Mitochondria can synthesize a limited number of proteins encoded by mtDNA (mitochondrial DNA) by using their own biosynthetic machinery, whereas most of the proteins in mitochondria are imported from the cytosol. It could be hypothesized that the mitochondrial pool of amino acids follows the frequency of amino acids in mtDNA-encoded proteins or, alternatively, that the profile is the result of the participation of amino acids in pathways other than protein synthesis (e.g. haem biosynthesis and aminotransferase reactions). These hypotheses were tested by evaluating the pool of free amino acids and derivatives in highly-coupled purified liver mitochondria obtained from rats fed on a nutritionally adequate diet for growth. Our results indicated that the pool mainly reflects the amino acid composition of mtDNA-encoded proteins, suggesting that there is a post-translational control of protein synthesis. This conclusion was supported by the following findings: (i) correlation between the concentration of free amino acids in the matrix and the frequency of abundance of amino acids in mtDNA-encoded proteins; (ii) the similar ratios of essential-to-non-essential amino acids in mtDNA-encoded proteins and the mitochondrial pool of amino acids; and (iii), lack of a correlation between codon usage or tRNA levels and amino-acid concentrations. Quantitative information on the mammalian mitochondrial content of amino acids, such as that presented in the present study, along with functional studies, will help us to better understand the pathogenesis of mitochondrial diseases or the biochemical implications in mitochondrial metabolism.


Asunto(s)
Aminoácidos/metabolismo , ADN Mitocondrial/metabolismo , Mitocondrias Hepáticas/metabolismo , Proteínas Mitocondriales/biosíntesis , Biosíntesis de Proteínas/fisiología , Animales , Codón/metabolismo , Masculino , ARN de Transferencia/metabolismo , Ratas , Ratas Sprague-Dawley
13.
J Comp Neurol ; 494(3): 485-94, 2006 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-16320252

RESUMEN

The anterior piriform cortex (APC) has been shown to be an essential brain structure for the detection of dietary indispensable amino acid (IAA) deficiency, but little has been known about possible molecular detection mechanisms. Increased phosphorylation of the alpha-subunit of the eukaryotic initiation factor 2alpha (eIF2alpha) has been directly linked to amino acid deficiency in yeast. Recently, we have shown increased phosphorylation of eIF2alpha (p-eIF2alpha) in the rat APC 20 minutes after ingestion of an IAA-deficient meal. We suggest that if phosphorylation of eIF2alpha is an important mechanism in detection of IAA deficiency, then APC neurons that show p-eIF2alpha should also show molecular evidence of potentiation. The present research demonstrates increased expression and co-localization of p-eIF2alpha and phosphorylated extracellular signal-regulated protein kinase 1/2 (p-ERK1/2) in APC neurons, but not in the primary motor or agranular insular cortices in response to an IAA-deficient diet. ERK1/2 is an element of the mitogen-activated protein kinase cascade, an intraneuronal signaling mechanism associated with neuronal activation. The region of the APC that responds to IAA deficiency with increased p-eIF2alpha and p-ERK1/2 labeling ranges from 3.1 to 2.5 mm rostral of bregma. Within this region, only a few neurons respond to IAA deficiency with co-localization of abundant p-eIF2alpha and p-ERK1/2. These chemosensory neurons probably detect IAA deficiency and generate neuronal signaling to other portions of the brain, changing feeding behavior.


Asunto(s)
Factor 2 Eucariótico de Iniciación/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Giro Parahipocampal/enzimología , Treonina/deficiencia , Alimentación Animal , Animales , Células Quimiorreceptoras/enzimología , Inmunohistoquímica , Masculino , Neuronas/enzimología , Giro Parahipocampal/citología , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Estadísticas no Paramétricas , Treonina/metabolismo
14.
Front Neurosci ; 10: 159, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27147951

RESUMEN

Fragile X premutation alleles have 55-200 CGG repeats in the 5' UTR of the FMR1 gene. Altered zinc (Zn) homeostasis has been reported in fibroblasts from >60 years old premutation carriers, in which Zn supplementation significantly restored Zn-dependent mitochondrial protein import/processing and function. Given that mitochondria play a critical role in synaptic transmission, brain function, and cognition, we tested FMRP protein expression, brain bioenergetics, and expression of the Zn-dependent synaptic scaffolding protein SH3 and multiple ankyrin repeat domains 3 (Shank3) in a knock-in (KI) premutation mouse model with 180 CGG repeats. Mitochondrial outcomes correlated with FMRP protein expression (but not FMR1 gene expression) in KI mice and human fibroblasts from carriers of the pre- and full-mutation. Significant deficits in brain bioenergetics, Zn levels, and Shank3 protein expression were observed in the Zn-rich regions KI hippocampus and cerebellum at PND21, with some of these effects lasting into adulthood (PND210). A strong genotype × age interaction was observed for most of the outcomes tested in hippocampus and cerebellum, whereas in cortex, age played a major role. Given that the most significant effects were observed at the end of the lactation period, we hypothesized that KI milk might have a role at compounding the deleterious effects on the FMR1 genetic background. A higher gene expression of ZnT4 and ZnT6, Zn transporters abundant in brain and lactating mammary glands, was observed in the latter tissue of KI dams. A cross-fostering experiment allowed improving cortex bioenergetics in KI pups nursing on WT milk. Conversely, WT pups nursing on KI milk showed deficits in hippocampus and cerebellum bioenergetics. A highly significant milk type × genotype interaction was observed for all three-brain regions, being cortex the most influenced. Finally, lower milk-Zn levels were recorded in milk from lactating women carrying the premutation as well as other Zn-related outcomes (Zn-dependent alkaline phosphatase activity and lactose biosynthesis-whose limiting step is the Zn-dependent ß-1,4-galactosyltransferase). In premutation carriers, altered Zn homeostasis, brain bioenergetics and Shank3 levels could be compounded by Zn-deficient milk, increasing the risk of developing emotional and neurological/cognitive problems and/or FXTAS later in life.

15.
Brain Pathol ; 25(4): 441-53, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25117056

RESUMEN

Alaskan Husky encephalopathy (AHE(1) ) is a fatal brain disease associated with a mutation in SLC19A3.1 (c.624insTTGC, c.625C>A). This gene encodes for a thiamine transporter 2 with a predominately (CNS) central nervous system distribution. Considering that brain is particularly vulnerable to thiamine deficiency because of its reliance on thiamine pyrophosphate (TPP)-dependent metabolic pathways involved in energy metabolism and neurotransmitter synthesis, we characterized the impact of this mutation on thiamine status, brain bioenergetics and the contribution of oxidative stress to this phenotype. In silico modeling of the mutated transporter indicated a significant loss of alpha-helices resulting in a more open protein structure suggesting an impaired thiamine transport ability. The cerebral cortex and thalamus of affected dogs were severely deficient in TPP-dependent enzymes accompanied by decreases in mitochondrial mass and oxidative phosphorylation (OXPHOS) capacity, and increases in oxidative stress. These results along with the behavioral and pathological findings indicate that the phenotype associated with AHE is consistent with a brain-specific thiamine deficiency, leading to brain mitochondrial dysfunction and increased oxidative stress. While some of the biochemical deficits, neurobehavior and affected brain areas in AHE were shared by Wernicke's and Korsakoff's syndromes, several differences were noted likely arising from a tissue-specific vs. that from a whole-body thiamine deficiency.


Asunto(s)
Encéfalo/patología , Proteínas de Transporte de Membrana/genética , Mitocondrias/patología , Mutación/genética , Deficiencia de Tiamina , Animales , Encéfalo/ultraestructura , ADN Mitocondrial/genética , Perros , Mitocondrias/genética , Modelos Moleculares , Enfermedades del Sistema Nervioso/etiología , Tiamina/metabolismo , Deficiencia de Tiamina/genética , Deficiencia de Tiamina/patología , Deficiencia de Tiamina/veterinaria
16.
PLoS One ; 7(8): e42504, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22900024

RESUMEN

Etiology of aberrant social behavior consistently points to a strong polygenetic component involved in fundamental developmental pathways, with the potential of being enhanced by defects in bioenergetics. To this end, the occurrence of social deficits and mitochondrial outcomes were evaluated in conditional Pten (Phosphatase and tensin homolog) haplo-insufficient mice, in which only one allele was selectively knocked-out in neural tissues. Pten mutations have been linked to Alzheimer's disease and syndromic autism spectrum disorders, among others. By 4-6 weeks of age, Pten insufficiency resulted in the increase of several mitochondrial Complex activities (II-III, IV and V) not accompanied by increases in mitochondrial mass, consistent with an activation of the PI3K/Akt pathway, of which Pten is a negative modulator. At 8-13 weeks of age, Pten haplo-insufficient mice did not show significant behavioral abnormalities or changes in mitochondrial outcomes, but by 20-29 weeks, they displayed aberrant social behavior (social avoidance, failure to recognize familiar mouse, and repetitive self-grooming), macrocephaly, increased oxidative stress, decreased cytochrome c oxidase (CCO) activity (50%) and increased mtDNA deletions in cerebellum and hippocampus. Mitochondrial dysfunction was the result of a downregulation of p53-signaling pathway evaluated by lower protein expression of p21 (65% of controls) and the CCO chaperone SCO2 (47% of controls), two p53-downstream targets. This mechanism was confirmed in Pten-deficient striatal neurons and, HCT 116 cells with different p53 gene dosage. These results suggest a unique pathogenic mechanism of the Pten-p53 axis in mice with aberrant social behavior: loss of Pten (via p53) impairs mitochondrial function elicited by an early defective assembly of CCO and later enhanced by the accumulation of mtDNA deletions. Consistent with our results, (i) SCO2 deficiency and/or CCO activity defects have been reported in patients with learning disabilities including autism and (ii) mutated proteins in ASD have been found associated with p53-signaling pathways.


Asunto(s)
Haploinsuficiencia/genética , Mitocondrias/genética , Fosfohidrolasa PTEN/genética , Trastorno de la Conducta Social/genética , Proteína p53 Supresora de Tumor/genética , Animales , Conducta Animal , Corteza Cerebelosa/metabolismo , Cerebelo/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Células HCT116 , Hipocampo/metabolismo , Humanos , Masculino , Megalencefalia/genética , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Neuronas/metabolismo , Fosfohidrolasa PTEN/metabolismo , Trastorno de la Conducta Social/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
17.
Am J Physiol Gastrointest Liver Physiol ; 296(5): G1130-9, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19228885

RESUMEN

Diets deficient in an indispensable amino acid are known to suppress food intake in rats. Few studies were focused at understanding how amino acid-deficient diets may elicit biochemical changes at the mitochondrial level. The goal of this study was to evaluate mitochondrial function in rats fed diets with 0.00, 0.18, 0.36, and 0.88% threonine (Thr) (set at 0, 30, 60, and 140% of Thr requirement for growth). Here, it is described for the first time that Thr-deficient diets induce a specific uncoupling of mitochondria in liver, especially with NADH-linked substrates, not observed in heart (except for Thr-devoid diet). The advantage of this situation would be to provide ATP to support growth and maintenance when high-quality protein food (or wealth of high-quality food in general) is available, whereas Thr-deficient diets (or deficient-quality protein food) promote the opposite, increasing mitochondrial uncoupling in liver. The uncoupling with NADH substrates would favor the use of nutrients as energy sources with higher FADH-to-NADH ratios, such as fat, minimizing the first irreversible NADH-dependent catabolism of many amino acids, including Thr, thus enhancing the use of the limiting amino acid for protein synthesis when a low quality protein source is available.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales , Proteínas en la Dieta/metabolismo , Metabolismo Energético , Hígado/metabolismo , Mitocondrias Hepáticas/metabolismo , Deficiencia de Proteína/metabolismo , Treonina/deficiencia , Adenosina Trifosfato/metabolismo , Animales , Peso Corporal , Proteínas en la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Ingestión de Alimentos , Flavina-Adenina Dinucleótido/metabolismo , Masculino , Mitocondrias Cardíacas/metabolismo , Miocardio/metabolismo , NAD/metabolismo , Fosforilación Oxidativa , Deficiencia de Proteína/fisiopatología , Ratas , Ratas Sprague-Dawley , Treonina/administración & dosificación , Factores de Tiempo
18.
Science ; 307(5716): 1776-8, 2005 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-15774759

RESUMEN

Recognizing a deficiency of indispensable amino acids (IAAs) for protein synthesis is vital for dietary selection in metazoans, including humans. Cells in the brain's anterior piriform cortex (APC) are sensitive to IAA deficiency, signaling diet rejection and foraging for complementary IAA sources, but the mechanism is unknown. Here we report that the mechanism for recognizing IAA-deficient foods follows the conserved general control (GC) system, wherein uncharged transfer RNA induces phosphorylation of eukaryotic initiation factor 2 (eIF2) via the GC nonderepressing 2 (GCN2) kinase. Thus, a basic mechanism of nutritional stress management functions in mammalian brain to guide food selection for survival.


Asunto(s)
Aminoácidos Esenciales/administración & dosificación , Aminoácidos Esenciales/deficiencia , Factor 2 Eucariótico de Iniciación/metabolismo , Alimentos , Leucina/análogos & derivados , Vías Olfatorias/metabolismo , Proteínas Quinasas/metabolismo , ARN de Transferencia/metabolismo , Acilación , Aminoácidos Esenciales/análisis , Animales , Dieta , Ingestión de Alimentos , Preferencias Alimentarias , Leucina/administración & dosificación , Leucina/farmacología , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas Serina-Treonina Quinasas , Ratas , Estereoisomerismo , Treonina/administración & dosificación , eIF-2 Quinasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA