Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuropathol Appl Neurobiol ; 50(3): e12977, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38680020

RESUMO

AIM: Leigh syndrome (LS), the most common paediatric presentation of genetic mitochondrial dysfunction, is a multi-system disorder characterised by severe neurologic and metabolic abnormalities. Symmetric, bilateral, progressive necrotizing lesions in the brainstem are defining features of the disease. Patients are often symptom free in early life but typically develop symptoms by about 2 years of age. The mechanisms underlying disease onset and progression in LS remain obscure. Recent studies have shown that the immune system causally drives disease in the Ndufs4(-/-) mouse model of LS: treatment of Ndufs4(-/-) mice with the macrophage-depleting Csf1r inhibitor pexidartinib prevents disease. While the precise mechanisms leading to immune activation and immune factors involved in disease progression have not yet been determined, interferon-gamma (IFNγ) and interferon gamma-induced protein 10 (IP10) were found to be significantly elevated in Ndufs4(-/-) brainstem, implicating these factors in disease. Here, we aimed to explore the role of IFNγ and IP10 in LS. METHODS: To establish the role of IFNγ and IP10 in LS, we generated IFNγ and IP10 deficient Ndufs4(-/-)/Ifng(-/-) and Ndufs4(-/-)/IP10(-/-) double knockout animals, as well as IFNγ and IP10 heterozygous, Ndufs4(-/-)/Ifng(+/-) and Ndufs4(-/-)/IP10(+/-), animals. We monitored disease onset and progression to define the impact of heterozygous or homozygous loss of IFNγ and IP10 in LS. RESULTS: Loss of IP10 does not significantly impact the onset or progression of disease in the Ndufs4(-/-) model. IFNγ loss significantly extends survival and delays disease progression in a gene dosage-dependent manner, though the benefits are modest compared to Csf1r inhibition. CONCLUSIONS: IFNγ contributes to disease onset and progression in LS. Our findings suggest that IFNγ targeting therapies may provide some benefits in genetic mitochondrial disease, but targeting IFNγ alone would likely yield only modest benefits in LS.


Assuntos
Modelos Animais de Doenças , Progressão da Doença , Complexo I de Transporte de Elétrons , Interferon gama , Doença de Leigh , Camundongos Knockout , Animais , Doença de Leigh/patologia , Doença de Leigh/genética , Interferon gama/metabolismo , Camundongos , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/deficiência , Camundongos Endogâmicos C57BL , Tronco Encefálico/patologia , Tronco Encefálico/metabolismo
2.
Anesthesiology ; 140(4): 715-728, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38147628

RESUMO

BACKGROUND: Volatile anesthetics induce hyperpolarizing potassium currents in spinal cord neurons that may contribute to their mechanism of action. They are induced at lower concentrations of isoflurane in noncholinergic neurons from mice carrying a loss-of-function mutation of the Ndufs4 gene, required for mitochondrial complex I function. The yeast NADH dehydrogenase enzyme, NDi1, can restore mitochondrial function in the absence of normal complex I activity, and gain-of-function Ndi1 transgenic mice are resistant to volatile anesthetics. The authors tested whether NDi1 would reduce the hyperpolarization caused by isoflurane in neurons from Ndufs4 and wild-type mice. Since volatile anesthetic behavioral hypersensitivity in Ndufs4 is transduced uniquely by glutamatergic neurons, it was also tested whether these currents were also unique to glutamatergic neurons in the Ndufs4 spinal cord. METHODS: Spinal cord neurons from wild-type, NDi1, and Ndufs4 mice were patch clamped to characterize isoflurane sensitive currents. Neuron types were marked using fluorescent markers for cholinergic, glutamatergic, and γ-aminobutyric acid-mediated (GABAergic) neurons. Norfluoxetine was used to identify potassium channel type. Neuron type-specific Ndufs4 knockout animals were generated using type-specific Cre-recombinase with floxed Ndufs4. RESULTS: Resting membrane potentials (RMPs) of neurons from NDi1;Ndufs4, unlike those from Ndufs4, were not hyperpolarized by 0.6% isoflurane (Ndufs4, ΔRMP -8.2 mV [-10 to -6.6]; P = 1.3e-07; Ndi1;Ndufs4, ΔRMP -2.1 mV [-7.6 to +1.4]; P = 1). Neurons from NDi1 animals in a wild-type background were not hyperpolarized by 1.8% isoflurane (wild-type, ΔRMP, -5.2 mV [-7.3 to -3.2]; P = 0.00057; Ndi1, ΔRMP, 0.6 mV [-1.7 to 3.2]; P = 0.68). In spinal cord slices from global Ndufs4 animals, holding currents (HC) were induced by 0.6% isoflurane in both GABAergic (ΔHC, 81.3 pA [61.7 to 101.4]; P = 2.6e-05) and glutamatergic (ΔHC, 101.2 pA [63.0 to 146.2]; P = 0.0076) neurons. In neuron type-specific Ndufs4 knockouts, HCs were increased in cholinergic (ΔHC, 119.5 pA [82.3 to 156.7]; P = 0.00019) and trended toward increase in glutamatergic (ΔHC, 85.5 pA [49 to 126.9]; P = 0.064) neurons but not in GABAergic neurons. CONCLUSIONS: Bypassing complex I by overexpression of NDi1 eliminates increases in potassium currents induced by isoflurane in the spinal cord. The isoflurane-induced potassium currents in glutamatergic neurons represent a potential downstream mechanism of complex I inhibition in determining minimum alveolar concentration.


Assuntos
Anestésicos Inalatórios , Isoflurano , Camundongos , Animais , Isoflurano/farmacologia , Anestésicos Inalatórios/farmacologia , Canais de Potássio , Medula Espinal , Camundongos Transgênicos , Interneurônios , Complexo I de Transporte de Elétrons/genética , Colinérgicos
3.
Nat Metab ; 5(6): 955-967, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37365290

RESUMO

Mitochondrial diseases represent a spectrum of disorders caused by impaired mitochondrial function, ranging in severity from mortality during infancy to progressive adult-onset disease. Mitochondrial dysfunction is also recognized as a molecular hallmark of the biological ageing process. Rapamycin, a drug that increases lifespan and health during normative ageing, also increases survival and reduces neurological symptoms in a mouse model of the severe mitochondrial disease Leigh syndrome. The Ndufs4 knockout (Ndufs4-/-) mouse lacks the complex I subunit NDUFS4 and shows rapid onset and progression of neurodegeneration mimicking patients with Leigh syndrome. Here we show that another drug that extends lifespan and delays normative ageing in mice, acarbose, also suppresses symptoms of disease and improves survival of Ndufs4-/- mice. Unlike rapamycin, acarbose rescues disease phenotypes independently of inhibition of the mechanistic target of rapamycin. Furthermore, rapamycin and acarbose have additive effects in delaying neurological symptoms and increasing maximum lifespan in Ndufs4-/- mice. We find that acarbose remodels the intestinal microbiome and alters the production of short-chain fatty acids. Supplementation with tributyrin, a source of butyric acid, recapitulates some effects of acarbose on lifespan and disease progression, while depletion of the endogenous microbiome in Ndufs4-/- mice appears to fully recapitulate the effects of acarbose on healthspan and lifespan in these animals. To our knowledge, this study provides the first evidence that alteration of the gut microbiome plays a significant role in severe mitochondrial disease and provides further support for the model that biological ageing and severe mitochondrial disorders share underlying common mechanisms.


Assuntos
Doença de Leigh , Doenças Mitocondriais , Camundongos , Animais , Doença de Leigh/tratamento farmacológico , Doença de Leigh/genética , Acarbose/farmacologia , Acarbose/uso terapêutico , Doenças Mitocondriais/tratamento farmacológico , Mitocôndrias/genética , Sirolimo/farmacologia , Sirolimo/uso terapêutico , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons
4.
Curr Biol ; 32(14): 3016-3032.e3, 2022 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-35688155

RESUMO

The mechanisms of volatile anesthetic action remain among the most perplexing mysteries of medicine. Across phylogeny, volatile anesthetics selectively inhibit mitochondrial complex I, and they also depress presynaptic excitatory signaling. To explore how these effects are linked, we studied isoflurane effects on presynaptic vesicle cycling and ATP levels in hippocampal cultured neurons from wild-type and complex I mutant (Ndufs4(KO)) mice. To bypass complex I, we measured isoflurane effects on anesthetic sensitivity in mice expressing NADH dehydrogenase (NDi1). Endocytosis in physiologic concentrations of glucose was delayed by effective behavioral concentrations of isoflurane in both wild-type (τ [unexposed] 44.8 ± 24.2 s; τ [exposed] 116.1 ± 28.1 s; p < 0.01) and Ndufs4(KO) cultures (τ [unexposed] 67.6 ± 16.0 s; τ [exposed] 128.4 ± 42.9 s; p = 0.028). Increasing glucose, to enhance glycolysis and increase ATP production, led to maintenance of both ATP levels and endocytosis (τ [unexposed] 28.0 ± 14.4; τ [exposed] 38.2 ± 5.7; reducing glucose worsened ATP levels and depressed endocytosis (τ [unexposed] 85.4 ± 69.3; τ [exposed] > 1,000; p < 0.001). The block in recycling occurred at the level of reuptake of synaptic vesicles into the presynaptic cell. Expression of NDi1 in wild-type mice caused behavioral resistance to isoflurane for tail clamp response (EC50 Ndi1(-) 1.27% ± 0.14%; Ndi1(+) 1.55% ± 0.13%) and halothane (EC50 Ndi1(-) 1.20% ± 0.11%; Ndi1(+) 1.46% ± 0.10%); expression of NDi1 in neurons improved hippocampal function, alleviated inhibition of presynaptic recycling, and increased ATP levels during isoflurane exposure. The clear alignment of cell culture data to in vivo phenotypes of both isoflurane-sensitive and -resistant mice indicates that inhibition of mitochondrial complex I is a primary mechanism of action of volatile anesthetics.


Assuntos
Anestésicos Inalatórios , Isoflurano , Trifosfato de Adenosina , Anestésicos Inalatórios/farmacologia , Animais , Complexo I de Transporte de Elétrons/genética , Endocitose , Glucose , Isoflurano/farmacologia , Camundongos
5.
Neurobiol Dis ; 163: 105594, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34933094

RESUMO

Genetic mitochondrial diseases are the most frequent cause of inherited metabolic disorders and one of the most prevalent causes of heritable neurological disease. Leigh syndrome is the most common clinical presentation of pediatric mitochondrial disease, typically appearing in the first few years of life, and involving severe multisystem pathologies. Clinical care for Leigh syndrome patients is difficult, complicated by the wide range of symptoms including characteristic progressive CNS lesion, metabolic sequelae, and epileptic seizures, which can be intractable to standard management. While no proven therapies yet exist for the underlying mitochondrial disease, a ketogenic diet has led to some reports of success in managing mitochondrial epilepsies, with ketosis reducing seizure risk and severity. The impact of ketosis on other aspects of disease progression in Leigh syndrome has not been studied, however, and a rigorous study of the impact of ketosis on seizures in mitochondrial disease is lacking. Conversely, preclinical efforts have identified the intracellular nutrient signaling regulator mTOR as a promising therapeutic target, with data suggesting the benefits are mediated by metabolic changes. mTOR inhibition alleviates epilepsies arising from defects in TSC, an mTOR regulator, but the therapeutic potential of mTOR inhibition in seizures related to primary mitochondrial dysfunction is unknown. Given that ketogenic diet is used clinically in the setting of mitochondrial disease, and mTOR inhibition is in clinical trials for intractable pediatric epilepsies of diverse causal origins, a direct experimental assessment of their effects is imperative. Here, we define the impact of dietary ketosis on survival and CNS disease in the Ndufs4(KO) mouse model of Leigh syndrome and the therapeutic potential of both dietary ketosis and mTOR inhibition on seizures in this model. These data provide timely insight into two important clinical interventions.


Assuntos
Dieta Cetogênica , Doença de Leigh/terapia , Sirolimo/uso terapêutico , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/genética , Doença de Leigh/dietoterapia , Doença de Leigh/tratamento farmacológico , Doença de Leigh/genética , Camundongos , Camundongos Knockout , Sirolimo/farmacologia , Resultado do Tratamento
6.
Br J Anaesth ; 128(1): 77-88, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34857359

RESUMO

BACKGROUND: If anaesthetics cause permanent cognitive deficits in some children, the implications are enormous, but the molecular causes of anaesthetic-induced neurotoxicity, and consequently possible therapies, are still debated. Anaesthetic exposure early in development can be neurotoxic in the invertebrate Caenorhabditis elegans causing endoplasmic reticulum (ER) stress and defects in chemotaxis during adulthood. We screened this model organism for compounds that alleviated neurotoxicity, and then tested these candidates for efficacy in mice. METHODS: We screened compounds for alleviation of ER stress induction by isoflurane in C. elegans assayed by induction of a green fluorescent protein (GFP) reporter. Drugs that inhibited ER stress were screened for reduction of the anaesthetic-induced chemotaxis defect. Compounds that alleviated both aspects of neurotoxicity were then blindly tested for the ability to inhibit induction of caspase-3 by isoflurane in P7 mice. RESULTS: Isoflurane increased ER stress indicated by increased GFP reporter fluorescence (240% increase, P<0.001). Nine compounds reduced induction of ER stress by isoflurane by 90-95% (P<0.001 in all cases). Of these compounds, tetraethylammonium chloride and trehalose also alleviated the isoflurane-induced defect in chemotaxis (trehalose by 44%, P=0.001; tetraethylammonium chloride by 23%, P<0.001). In mouse brain, tetraethylammonium chloride reduced isoflurane-induced caspase staining in the anterior cortical (-54%, P=0.007) and hippocampal regions (-46%, P=0.002). DISCUSSION: Tetraethylammonium chloride alleviated isoflurane-induced neurotoxicity in two widely divergent species, raising the likelihood that it may have therapeutic value. In C. elegans, ER stress predicts isoflurane-induced neurotoxicity, but is not its cause.


Assuntos
Isoflurano/toxicidade , Síndromes Neurotóxicas/prevenção & controle , Tetraetilamônio/farmacologia , Anestésicos Inalatórios/toxicidade , Animais , Caenorhabditis elegans , Caspase 3/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Camundongos , Síndromes Neurotóxicas/etiologia , Especificidade da Espécie
7.
Mol Genet Metab ; 130(2): 118-132, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32331968

RESUMO

Leigh Syndrome (LS) is a mitochondrial disorder defined by progressive focal neurodegenerative lesions in specific regions of the brain. Defects in NDUFS4, a subunit of complex I of the mitochondrial electron transport chain, cause LS in humans; the Ndufs4 knockout mouse (Ndufs4(KO)) closely resembles the human disease. Here, we probed brain region-specific molecular signatures in pre-symptomatic Ndufs4(KO) to identify factors which underlie focal neurodegeneration. Metabolomics revealed that free amino acid concentrations are broadly different by region, and glucose metabolites are increased in a manner dependent on both region and genotype. We then tested the impact of the mTOR inhibitor rapamycin, which dramatically attenuates LS in Ndufs4(KO), on region specific metabolism. Our data revealed that loss of Ndufs4 drives pathogenic changes to CNS glutamine/glutamate/α-ketoglutarate metabolism which are rescued by mTOR inhibition Finally, restriction of the Ndufs4 deletion to pre-synaptic glutamatergic neurons recapitulated the whole-body knockout. Together, our findings are consistent with mTOR inhibition alleviating disease by increasing availability of α-ketoglutarate, which is both an efficient mitochondrial complex I substrate in Ndufs4(KO) and an important metabolite related to neurotransmitter metabolism in glutamatergic neurons.


Assuntos
Encéfalo/patologia , Complexo I de Transporte de Elétrons/fisiologia , Ácido Glutâmico/metabolismo , Ácidos Cetoglutáricos/metabolismo , Doença de Leigh/patologia , Metaboloma , Doenças Mitocondriais/patologia , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Doença de Leigh/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças Mitocondriais/metabolismo , Serina-Treonina Quinases TOR/metabolismo
9.
PLoS One ; 11(1): e0148219, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26824698

RESUMO

BACKGROUND: Lack of NDUFS4, a subunit of mitochondrial complex I (NADH:ubiquinone oxidoreductase), causes Leigh syndrome (LS), a progressive encephalomyopathy. Knocking out Ndufs4, either systemically or in brain only, elicits LS in mice. In patients as well as in KO mice distinct regions of the brain degenerate while surrounding tissue survives despite systemic complex I dysfunction. For the understanding of disease etiology and ultimately for the development of rationale treatments for LS, it appears important to uncover the mechanisms that govern focal neurodegeneration. RESULTS: Here we used the Ndufs4(KO) mouse to investigate whether regional and temporal differences in respiratory capacity of the brain could be correlated with neurodegeneration. In the KO the respiratory capacity of synaptosomes from the degeneration prone regions olfactory bulb, brainstem and cerebellum was significantly decreased. The difference was measurable even before the onset of neurological symptoms. Furthermore, neither compensating nor exacerbating changes in glycolytic capacity of the synaptosomes were found. By contrast, the KO retained near normal levels of synaptosomal respiration in the degeneration-resistant/resilient "rest" of the brain. We also investigated non-synaptic mitochondria. The KO expectedly had diminished capacity for oxidative phosphorylation (state 3 respiration) with complex I dependent substrate combinations pyruvate/malate and glutamate/malate but surprisingly had normal activity with α-ketoglutarate/malate. No correlation between oxidative phosphorylation (pyruvate/malate driven state 3 respiration) and neurodegeneration was found: Notably, state 3 remained constant in the KO while in controls it tended to increase with time leading to significant differences between the genotypes in older mice in both vulnerable and resilient brain regions. Neither regional ROS damage, measured as HNE-modified protein, nor regional complex I stability, assessed by blue native gels, could explain regional neurodegeneration. CONCLUSION: Our data suggests that locally insufficient respiration capacity of the nerve terminals may drive focal neurodegeneration.


Assuntos
Tronco Encefálico/metabolismo , Cerebelo/metabolismo , Complexo I de Transporte de Elétrons/genética , Doença de Leigh/genética , Mitocôndrias/metabolismo , Bulbo Olfatório/metabolismo , Animais , Tronco Encefálico/patologia , Respiração Celular/genética , Cerebelo/patologia , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/deficiência , Expressão Gênica , Ácido Glutâmico/metabolismo , Glicólise/genética , Humanos , Ácidos Cetoglutáricos/metabolismo , Doença de Leigh/metabolismo , Doença de Leigh/patologia , Malatos/metabolismo , Camundongos , Mitocôndrias/patologia , Bulbo Olfatório/patologia , Especificidade de Órgãos , Fosforilação Oxidativa , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/patologia , Ácido Pirúvico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sinaptossomos/metabolismo , Sinaptossomos/patologia
10.
Proc Natl Acad Sci U S A ; 112(45): E6148-57, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26504246

RESUMO

Mitochondria play an important role in numerous diseases as well as normative aging. Severe reduction in mitochondrial function contributes to childhood disorders such as Leigh Syndrome, whereas mild disruption can extend the lifespan of model organisms. The Caenorhabditis elegans isp-1 gene encodes the Rieske iron-sulfur protein subunit of cytochrome c oxidoreductase (complex III of the electron transport chain). The partial loss of function allele, isp-1(qm150), leads to several pleiotropic phenotypes. To better understand the molecular mechanisms of ISP-1 function, we sought to identify genetic suppressors of the delayed development of isp-1(qm150) animals. Here we report a series of intragenic suppressors, all located within a highly conserved six amino acid tether region of ISP-1. These intragenic mutations suppress all of the evaluated isp-1(qm150) phenotypes, including developmental rate, pharyngeal pumping rate, brood size, body movement, activation of the mitochondrial unfolded protein response reporter, CO2 production, mitochondrial oxidative phosphorylation, and lifespan extension. Furthermore, analogous mutations show a similar effect when engineered into the budding yeast Rieske iron-sulfur protein Rip1, revealing remarkable conservation of the structure-function relationship of these residues across highly divergent species. The focus on a single subunit as causal both in generation and in suppression of diverse pleiotropic phenotypes points to a common underlying molecular mechanism, for which we propose a "spring-loaded" model. These observations provide insights into how gating and control processes influence the function of ISP-1 in mediating pleiotropic phenotypes including developmental rate, movement, sensitivity to stress, and longevity.


Assuntos
Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Complexo III da Cadeia de Transporte de Elétrons/química , Complexo III da Cadeia de Transporte de Elétrons/genética , Pleiotropia Genética/genética , Modelos Moleculares , Fenótipo , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/fisiologia , Tamanho da Ninhada/genética , Complexo III da Cadeia de Transporte de Elétrons/fisiologia , Crescimento e Desenvolvimento/genética , Longevidade/genética , Microscopia de Fluorescência , Movimento/fisiologia , Mutagênese , Mutação/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Engenharia de Proteínas , Proteínas de Saccharomyces cerevisiae/genética , Estresse Fisiológico/genética
11.
Dis Model Mech ; 7(10): 1165-74, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25085991

RESUMO

Mutations affecting mitochondrial complex I, a multi-subunit assembly that couples electron transfer to proton pumping, are the most frequent cause of heritable mitochondrial diseases. However, the mechanisms by which complex I dysfunction results in disease remain unclear. Here, we describe a Drosophila model of complex I deficiency caused by a homoplasmic mutation in the mitochondrial-DNA-encoded NADH dehydrogenase subunit 2 (ND2) gene. We show that ND2 mutants exhibit phenotypes that resemble symptoms of mitochondrial disease, including shortened lifespan, progressive neurodegeneration, diminished neural mitochondrial membrane potential and lower levels of neural ATP. Our biochemical studies of ND2 mutants reveal that complex I is unable to efficiently couple electron transfer to proton pumping. Thus, our study provides evidence that the ND2 subunit participates directly in the proton pumping mechanism of complex I. Together, our findings support the model that diminished respiratory chain activity, and consequent energy deficiency, are responsible for the pathogenesis of complex-I-associated neurodegeneration.


Assuntos
Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/genética , Doenças Mitocondriais/etiologia , Mutação , Bombas de Próton/metabolismo , Animais , Drosophila , Transporte de Elétrons , Doenças Mitocondriais/enzimologia , Doenças Mitocondriais/metabolismo , Fosforilação Oxidativa , Espécies Reativas de Oxigênio/metabolismo
12.
J Cereb Blood Flow Metab ; 34(3): 514-21, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24398942

RESUMO

Anesthetics used in infants and children are implicated in the development of neurocognitive disorders. Although propofol induces neuroapoptosis in developing brain, the underlying mechanisms require elucidation and may have an energetic basis. We studied substrate utilization in immature swine anesthetized with either propofol or isoflurane for 4 hours. Piglets were infused with 13-Carbon-labeled glucose and leucine in the common carotid artery to assess citric acid cycle (CAC) metabolism in the parietal cortex. The anesthetics produced similar systemic hemodynamics and cerebral oxygen saturation by near-infrared spectroscopy. Compared with isoflurane, propofol depleted ATP and glycogen stores. Propofol decreased pools of the CAC intermediates, citrate, and α-ketoglutarate, while markedly increasing succinate along with decreasing mitochondrial complex II activity. Propofol also inhibited acetyl-CoA entry into the CAC through pyruvate dehydrogenase, while promoting glycolytic flux with marked lactate accumulation. Although oxygen supply appeared similar between the anesthetic groups, propofol yielded a metabolic phenotype that resembled a hypoxic state. Propofol impairs substrate flux through the CAC in the immature cerebral cortex. These impairments occurred without systemic metabolic perturbations that typically accompany propofol infusion syndrome. These metabolic abnormalities may have a role in the neurotoxity observed with propofol in the vulnerable immature brain.


Assuntos
Anestésicos Gerais/efeitos adversos , Córtex Cerebral/efeitos dos fármacos , Isoflurano/efeitos adversos , Mitocôndrias , Propofol/efeitos adversos , Suínos/metabolismo , Administração por Inalação , Anestésicos Gerais/administração & dosagem , Animais , Animais Recém-Nascidos , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Metabolismo Energético/efeitos dos fármacos , Cromatografia Gasosa-Espectrometria de Massas , Glucose/metabolismo , Infusões Intravenosas , Isoflurano/administração & dosagem , Leucina/metabolismo , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Propofol/administração & dosagem , Suínos/crescimento & desenvolvimento
13.
Science ; 342(6165): 1524-8, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-24231806

RESUMO

Mitochondrial dysfunction contributes to numerous health problems, including neurological and muscular degeneration, cardiomyopathies, cancer, diabetes, and pathologies of aging. Severe mitochondrial defects can result in childhood disorders such as Leigh syndrome, for which there are no effective therapies. We found that rapamycin, a specific inhibitor of the mechanistic target of rapamycin (mTOR) signaling pathway, robustly enhances survival and attenuates disease progression in a mouse model of Leigh syndrome. Administration of rapamycin to these mice, which are deficient in the mitochondrial respiratory chain subunit Ndufs4 [NADH dehydrogenase (ubiquinone) Fe-S protein 4], delays onset of neurological symptoms, reduces neuroinflammation, and prevents brain lesions. Although the precise mechanism of rescue remains to be determined, rapamycin induces a metabolic shift toward amino acid catabolism and away from glycolysis, alleviating the buildup of glycolytic intermediates. This therapeutic strategy may prove relevant for a broad range of mitochondrial diseases.


Assuntos
Doença de Leigh/tratamento farmacológico , Doenças Mitocondriais/tratamento farmacológico , Terapia de Alvo Molecular , Complexos Multiproteicos/antagonistas & inibidores , Fármacos Neuroprotetores/uso terapêutico , Sirolimo/uso terapêutico , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/patologia , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Glicólise/efeitos dos fármacos , Doença de Leigh/genética , Doença de Leigh/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Camundongos Mutantes , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Doenças Mitocondriais/genética , Doenças Mitocondriais/patologia
14.
Anesth Analg ; 112(6): 1321-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21467554

RESUMO

BACKGROUND: Complex I of the electron transport chain (ETC) is a possible target of volatile anesthetics (VAs). Complex I enzymatic activities are inhibited by VAs, and dysfunction of complex I can lead to hypersensitivity to VAs in worms and in people. Mutant analysis in Caenorhabditis (C.) elegans suggests that VAs may specifically interfere with complex I function at the binding site for its substrate ubiquinone. We hypothesized that isoflurane inhibits electron transport by competing with ubiquinone for binding to complex I. METHODS: Wildtype and mutant C. elegans were used to study the effects of isoflurane on isolated mitochondria. Enzymatic activities of the ETC were assayed and dose-response curves determined using established techniques. Two-dimensional native gels of mitochondrial proteins were performed after exposure of mitochondria to isoflurane. RESULTS: Complex I is the most sensitive component of the ETC to isoflurane inhibition; however, the proximal portion of complex I (the flavoprotein) is relatively insensitive to isoflurane. Isoflurane and quinone do not compete for a common binding site on complex I. The absolute rate of complex I enzymatic activity in vitro does not predict immobilization of the animal by isoflurane. Isoflurane had no measurable effect on stability of mitochondrial supercomplexes. Reduction of ubiquinone by complex I displayed positive cooperative kinetics not disrupted by isoflurane. CONCLUSIONS: Isoflurane directly inhibits complex I at a site distal to the flavoprotein subcomplex. However, we have excluded our original hypothesis that isoflurane and ubiquinone compete for a common hydrophobic binding site on complex I. In addition, immobilization of the nematode by isoflurane is not due to limiting absolute amounts of complex I electron transport as measured in isolated mitochondria.


Assuntos
Anestésicos Inalatórios/farmacologia , Complexo I de Transporte de Elétrons/metabolismo , Isoflurano/farmacologia , Animais , Sítios de Ligação , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , Citocromos c/metabolismo , Transporte de Elétrons , Eletroforese em Gel Bidimensional/métodos , Eletroforese em Gel de Poliacrilamida , Cavalos , Interações Hidrofóbicas e Hidrofílicas , Isoflurano/química , Cinética , Mitocôndrias/metabolismo , Mutação , NADH Desidrogenase/metabolismo , Solubilidade , Ubiquinona/química , Água/química
15.
Curr Biol ; 16(16): 1641-5, 2006 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-16920626

RESUMO

Despite the widespread clinical use of volatile anesthetics, their mechanisms of action remain unknown [1-6]. An unbiased genetic screen in the nematode C. elegans for animals with altered volatile anesthetic sensitivity identified a mutant in a nuclear-encoded subunit of mitochondrial complex I [7,8]. This raised the question of whether mitochondrial dysfunction might be the primary mechanism by which volatile anesthetics act, rather than an untoward secondary effect [9,10]. We report here analysis of additional C. elegans mutations in orthologs of human genes that contribute to the formation of complex I, complex II, complex III, and coenzyme Q [11-14]. To further characterize the specific contribution of complex I, we generated four hypomorphic C. elegans mutants encoding different complex I subunits [15]. Our main finding is the identification of a clear correlation between complex I-dependent oxidative phosphorylation capacity and volatile anesthetic sensitivity. These extended data link a physiologic determinant of anesthetic action in a tractable animal model to similar clinical observations in children with mitochondrial myopathies [16]. This work is the first to specifically implicate complex I-dependent oxidative phosphorylation function as a primary mediator of volatile anesthetic effect.


Assuntos
Anestésicos/farmacologia , Caenorhabditis elegans/genética , Complexo de Proteínas da Cadeia de Transporte de Elétrons/genética , Mitocôndrias/metabolismo , Fosforilação Oxidativa/efeitos dos fármacos , Animais , Caenorhabditis elegans/efeitos dos fármacos , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Testes Genéticos , Halotano , Mutação/genética , Interferência de RNA
16.
Anesthesiology ; 101(2): 365-72, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15277919

RESUMO

BACKGROUND: : The gene gas-1 encodes a subunit of complex I of the mitochondrial electron transport chain in Caenorhabditis elegans. A mutation in gas-1 profoundly increases sensitivity of C. elegans to volatile anesthetics. It is unclear which aspects of mitochondrial function account for the hypersensitivity of the mutant. METHODS: : Oxidative phosphorylation was determined by measuring mitochondrial oxygen consumption using electron donors specific for either complex I or complex II. Adenosine triphosphate concentrations were determined by measuring luciferase activity. Oxidative damage to mitochondrial proteins was identified using specific antibodies. RESULTS: : Halothane inhibited oxidative phosphorylation in isolated wild-type mitochondria within a concentration range that immobilizes intact worms. At equal halothane concentrations, complex I activity but not complex II activity was lower in mitochondria from mutant (gas-1) animals than from wild-type (N2) animals. The halothane concentrations needed to immobilize 50% of N2 or gas-1 animals, respectively, did not reduce oxidative phosphorylation to identical rates in the two strains. In air, adenosine triphosphate concentrations were similar for N2 and gas-1 but were decreased in the presence of halothane only in gas-1 animals. Oxygen tension changed the sensitivity of both strains to halothane. When nematodes were raised in room air, oxidative damage to mitochondrial proteins was increased in the mutant animal compared with the wild type. CONCLUSIONS: : Rates of oxidative phosphorylation and changes in adenosine triphosphate concentrations by themselves do not control anesthetic-induced immobility of wild-type C. elegans. However, they may contribute to the increased sensitivity to volatile anesthetics of the gas-1 mutant. Oxidative damage to proteins may be an important contributor to sensitivity to volatile anesthetics in C. elegans.


Assuntos
Anestésicos Inalatórios/farmacologia , Caenorhabditis elegans/fisiologia , Complexo I de Transporte de Elétrons/efeitos dos fármacos , Halotano/farmacologia , Difosfato de Adenosina/metabolismo , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Western Blotting , Proteínas de Caenorhabditis elegans/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Metabolismo Energético/efeitos dos fármacos , Técnicas In Vitro , Cinética , Malatos/metabolismo , Oxidantes/toxicidade , Oxirredução , Fosforilação Oxidativa/efeitos dos fármacos , Succinatos/metabolismo
17.
J Biol Chem ; 279(52): 54479-86, 2004 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-15269213

RESUMO

The long-lived mutant of Caenorhabditis elegans, clk-1, is unable to synthesize ubiquinone, CoQ(9). Instead, the mutant accumulates demethoxyubiquinone(9) and small amounts of rhodoquinone(9) as well as dietary CoQ(8). We found a profound defect in oxidative phosphorylation, a test of integrated mitochondrial function, in clk-1 mitochondria fueled by NADH-linked electron donors, i.e. complex I-dependent substrates. Electron transfer from complex I to complex III, which requires quinones, is severely depressed, whereas the individual complexes are fully active. In contrast, oxidative phosphorylation initiated through complex II, which also requires quinones, is completely normal. Here we show that complexes I and II differ in their ability to use the quinone pool in clk-1. This is the first direct demonstration of a differential interaction of complex I and complex II with the endogenous quinone pool. This study uses the combined power of molecular genetics and biochemistry to highlight the role of quinones in mitochondrial function and aging.


Assuntos
Caenorhabditis elegans/crescimento & desenvolvimento , Mitocôndrias/metabolismo , Mutação , Fosforilação Oxidativa , Ubiquinona/análogos & derivados , Animais , Ácido Ascórbico/metabolismo , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/ultraestrutura , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/genética , Complexo II de Transporte de Elétrons/metabolismo , Ácido Glutâmico/metabolismo , Hidroquinonas/metabolismo , Malatos/metabolismo , Ácido Pirúvico/metabolismo , Quinonas/metabolismo , Especificidade por Substrato , Tetrametilfenilenodiamina/metabolismo , Fatores de Tempo , Ubiquinona/genética , Ubiquinona/metabolismo
18.
Mech Ageing Dev ; 125(6): 455-64, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15178135

RESUMO

A mutation in a subunit of complex I of the mitochondrial electron transport chain (gas-1) causes Caenorhabditis elegans to be hypersensitive to volatile anesthetics and oxygen as well as shortening lifespan. We hypothesized that changes in mitochondrial respiration or reactive oxygen species production cause these changes. Therefore, we compared gas-1 to other mitochondrial mutants to identify the relative importance of these two aspects of mitochondrial function in determining longevity. Lifespans of gas-1 and mev-1 were decreased compared with N2, while that of clk-1 was increased. Rates of oxidative phosphorylation were decreased in all three mutants, but the ROS damage was decreased only in clk-1. Suppressors of gas-1 increased rates of oxidative phosphorylation, decreased oxidative damage to mitochondrial proteins and increased lifespan. Two strains containing combinations of mutations predicted to have very decreased complex I function, had unexpectedly long lifespans. We conclude that mitochondrial changes in lifespan appear to be mediated primarily by changes in oxidative damage rather than by changes in rates of oxidative phosphorylation. In contrast, the effects of mitochondrial changes on anesthetic sensitivity appear to be mediated by both altered respiration and oxidative damage.


Assuntos
Anestésicos Inalatórios/farmacologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Complexo I de Transporte de Elétrons/metabolismo , NADH Desidrogenase/genética , NADH Desidrogenase/metabolismo , Estresse Oxidativo/fisiologia , Animais , Relação Dose-Resposta a Droga , Transporte de Elétrons , Complexo I de Transporte de Elétrons/genética , Expressão Gênica/efeitos dos fármacos , Longevidade/fisiologia , Mitocôndrias/metabolismo , Mutação/genética , Fosforilação Oxidativa , Oxigênio/toxicidade , Consumo de Oxigênio/efeitos dos fármacos , Consumo de Oxigênio/fisiologia , Análise de Sobrevida
19.
Alcohol Clin Exp Res ; 27(4): 584-92, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12711920

RESUMO

BACKGROUND: The gene gas-1 encodes the 49-kDa subunit of complex I of the mitochondrial electron transport chain in Caenorhabditis elegans. A mutation in gas-1 profoundly increases sensitivity to ethanol and decreases complex I-dependent metabolism in mitochondria. METHODS: Mitochondria were isolated from wild-type and gas-1 strains of C. elegans. The effects of ethanol on complex I-, II-, and III-dependent oxidative phosphorylation were measured for mitochondria from each strain. Reversibility of the effects of ethanol was determined by measuring oxidative phosphorylation after removal of mitochondria from 1.5 M ethanol. The effects of ethanol on mitochondrial structure were visualized with electron microscopy. RESULTS: We found that ethanol inhibited complex I-, II-, and III-dependent oxidative phosphorylation in isolated wild-type mitochondria at concentrations that immobilize intact worms. It is important to note that the inhibitory effects of ethanol on mitochondria from either C. elegans or rat skeletal muscle were reversible even at molar concentrations. Complex I activity was lower in mitochondria from gas-1 animals than in mitochondria from wild-type animals at equal ethanol concentrations. Complex II activity was higher in gas-1 than in wild-type mitochondria at all concentrations of ethanol. No difference was seen between the strains in the sensitivity of complex III to ethanol. CONCLUSIONS: The difference in ethanol sensitivities between gas-1 and wild-type nematodes results solely from altered complex I function. At the respective concentrations of ethanol that immobilize whole animals, mitochondria from each strain of worms displayed identical rates of complex I-dependent state 3 respiration. We conclude that a threshold value of complex I activity controls the transition from mobility to immobility of C. elegans.


Assuntos
Caenorhabditis elegans/efeitos dos fármacos , Complexo I de Transporte de Elétrons/genética , Etanol/farmacologia , Mitocôndrias/genética , Mutação/efeitos dos fármacos , Animais , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/genética , Relação Dose-Resposta a Droga , Complexo I de Transporte de Elétrons/fisiologia , Imobilização/fisiologia , Mitocôndrias/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA