Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Mol Brain ; 15(1): 22, 2022 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-35248135

RESUMEN

The accumulation of neurotoxic proteins characteristic of age-related neurodegenerative pathologies such as Alzheimer's and Parkinson's diseases is associated with the perturbation of metabolism, bioenergetics, and mitochondrial quality control. One approach to exploit these interactions therapeutically is to target the pathways that regulate metabolism. In this respect, the nutrient-sensing hexosamine biosynthesis pathway is of particular interest since it introduces a protein post-translational modification known as O-GlcNAcylation, which modifies different proteins in control versus neurodegenerative disease postmortem brains. A potent inhibitor of the O-GlcNAcase enzyme that removes the modification from proteins, Thiamet G (TG), has been proposed to have potential benefits in Alzheimer's disease. We tested whether key factors in the O-GlcNAcylation are correlated with mitochondrial electron transport and proteins related to the autophagy/lysosomal pathways in the cortex of male and female mice with and without exposure to TG (10 mg/kg i.p.). Mitochondrial complex activities were measured in the protein homogenates, and a panel of metabolic, autophagy/lysosomal proteins and O-GlcNAcylation enzymes were assessed by either enzyme activity assay or by western blot analysis. We found that the networks associated with O-GlcNAcylation enzymes and activities with mitochondrial parameters, autophagy-related proteins as well as neurodegenerative disease-related proteins exhibited sex and TG dependent differences. Taken together, these studies provide a framework of interconnectivity for multiple O-GlcNAc-dependent pathways in mouse brain of relevance to aging and sex/age-dependent neurodegenerative pathogenesis and response to potential therapies.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Enfermedad de Alzheimer/metabolismo , Animales , Autofagia , Metabolismo Energético , Femenino , Masculino , Ratones , Procesamiento Proteico-Postraduccional
2.
Acta Pharm Sin B ; 12(2): 511-531, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35256932

RESUMEN

Aging is by far the most prominent risk factor for Alzheimer's disease (AD), and both aging and AD are associated with apparent metabolic alterations. As developing effective therapeutic interventions to treat AD is clearly in urgent need, the impact of modulating whole-body and intracellular metabolism in preclinical models and in human patients, on disease pathogenesis, have been explored. There is also an increasing awareness of differential risk and potential targeting strategies related to biological sex, microbiome, and circadian regulation. As a major part of intracellular metabolism, mitochondrial bioenergetics, mitochondrial quality-control mechanisms, and mitochondria-linked inflammatory responses have been considered for AD therapeutic interventions. This review summarizes and highlights these efforts.

3.
Am J Transplant ; 21(9): 2964-2977, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33724664

RESUMEN

Calcineurin inhibitors (CNIs) are potent immunosuppressive agents, universally used following solid organ transplantation to prevent rejection. Although effective, the long-term use of CNIs is associated with nephrotoxicity. The etiology of this adverse effect is complex, and effective therapeutic interventions remain to be determined. Using a combination of in vitro techniques and a mouse model of CNI-mediated nephrotoxicity, we found that the CNIs, cyclosporine A (CsA), and tacrolimus (TAC) share a similar mechanism of tubular epithelial kidney cell injury, including mitochondrial dysfunction and release of High-Mobility Group Box I (HMGB1). CNIs promote bioenergetic reprogramming due to mitochondrial dysfunction and a shift toward glycolytic metabolism. These events were accompanied by diminished cell-to-cell adhesion, loss of the epithelial cell phenotype, and release of HMGB1. Notably, Erk1/2 inhibitors effectively diminished HMGB1 release, and similar inhibitor was observed on inclusion of pan-caspase inhibitor zVAD-FMK. In vivo, while CNIs activate tissue proremodeling signaling pathways, MAPK/Erk1/2 inhibitor prevented nephrotoxicity, including diminished HMGB1 release from kidney epithelial cells and accumulation in urine. In summary, HMGB1 is an early indicator and marker of progressive nephrotoxicity induced by CNIs. We suggest that proremodeling signaling pathway and loss of mitochondrial redox/bioenergetics homeostasis are crucial therapeutic targets to ameliorate CNI-mediated nephrotoxicity.


Asunto(s)
Inhibidores de la Calcineurina , Proteína HMGB1 , Animales , Inhibidores de la Calcineurina/efectos adversos , Ciclosporina/efectos adversos , Metabolismo Energético , Inmunosupresores/efectos adversos , Ratones , Tacrolimus/toxicidad
4.
Redox Biol ; 28: 101311, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31546171

RESUMEN

Non-invasive measures of the response of individual patients to cancer therapeutics is an emerging strategy in precision medicine. Platelets offer a potential dynamic marker for metabolism and bioenergetic responses in individual patients since they have active glycolysis and mitochondrial oxidative phosphorylation and can be easily isolated from a small blood sample. We have recently shown how the bioenergetic-metabolite interactome can be defined in platelets isolated from human subjects by measuring metabolites and bioenergetics in the same sample. In the present study, we used a model system to assess test the hypothesis that this interactome is modified by xenobiotics using exposure to the anti-cancer drug doxorubicin (Dox) in individual donors. We found that unsupervised analysis of the metabolome showed clear differentiation between the control and Dox treated group. Dox treatment resulted in a concentration-dependent decrease in bioenergetic parameters with maximal respiration being most sensitive and this was associated with significant changes in over 166 features. A metabolome-wide association study of Dox was also conducted, and Dox was found to have associations with metabolites in the glycolytic and TCA cycle pathways. Lastly, network analysis showed the impact of Dox on the bioenergetic-metabolite interactome and revealed profound changes in the regulation of reserve capacity. Taken together, these data support the conclusion that platelets are a suitable platform to predict and monitor therapeutic efficacy as well as anticipate susceptibility to toxicity in the context of precision medicine.


Asunto(s)
Plaquetas/efectos de los fármacos , Doxorrubicina/efectos adversos , Metabolismo Energético/efectos de los fármacos , Metaboloma/efectos de los fármacos , Plaquetas/metabolismo , Estudios de Casos y Controles , Ciclo del Ácido Cítrico/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Glucólisis/efectos de los fármacos , Humanos , Metabolómica/métodos , Medicina de Precisión , Aprendizaje Automático no Supervisado
5.
Mol Genet Metab Rep ; 19: 100451, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30740306

RESUMEN

Porphyria is a group of metabolic disorders due to altered enzyme activities within the heme biosynthetic pathway. It is a systemic disease with multiple potential contributions to mitochondrial dysfunction and oxidative stress. Recently, it has become possible to measure mitochondrial function from cells isolated from peripheral blood (cellular bioenergetics) using the XF96 analyzer (Seahorse Bioscience). Mitochondrial respiration in these cells is measured with the addition of activators and inhibitors of respiration. The output is measured as the O2 consumption rate (OCR) at basal conditions, ATP linked, proton leak, maximal, reserve capacity, non-mitochondrial, and oxidative burst. We performed cellular bioenergetics on 22 porphyria (12 porphyria cutanea tarda (PCT), seven acute hepatic porphyria (AHP), and three erythropoietic protoporphyria (EPP)) patients and 18 age and gender matched healthy controls. Of porphyria cases, eight were active (2 PCT, 1 EPP, and 5 AHP) and 14 in biochemical remission. The OCR were decreased in patients compared to healthy controls. The bioenergetic profile was significantly lower when measuring proton leak and the non-mitochondrial associated OCR in the eight active porphyria patients when compared to 18 healthy controls. In conclusion, we demonstrate that the bioenergetic profile and mitochondrial activities assessed in porphyria patients and is different than in healthy control individuals. Further, our novel preliminary findings suggest the existence of a mitochondrial dysfunction in porphyria and this may be used as potential non-invasive biomarker for disease activity. This needs to be assessed with a systematic examination in a larger patient cohort. Studies are also suggested to examine mitochondrial metabolism as basis to understand mechanisms of these findings and deriving mitochondrial based therapies for porphyria.

6.
Autophagy ; 13(11): 1828-1840, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28837411

RESUMEN

The production of reactive species contributes to the age-dependent accumulation of dysfunctional mitochondria and protein aggregates, all of which are associated with neurodegeneration. A putative mediator of these effects is the lipid peroxidation product 4-hydroxynonenal (4-HNE), which has been shown to inhibit mitochondrial function, and accumulate in the postmortem brains of patients with neurodegenerative diseases. This deterioration in mitochondrial quality could be due to direct effects on mitochondrial proteins, or through perturbation of the macroautophagy/autophagy pathway, which plays an essential role in removing damaged mitochondria. Here, we use a click chemistry-based approach to demonstrate that alkyne-4-HNE can adduct to specific mitochondrial and autophagy-related proteins. Furthermore, we found that at lower concentrations (5-10 µM), 4-HNE activates autophagy, whereas at higher concentrations (15 µM), autophagic flux is inhibited, correlating with the modification of key autophagy proteins at higher concentrations of alkyne-4-HNE. Increasing concentrations of 4-HNE also cause mitochondrial dysfunction by targeting complex V (the ATP synthase) in the electron transport chain, and induce significant changes in mitochondrial fission and fusion protein levels, which results in alterations to mitochondrial network length. Finally, inhibition of autophagy initiation using 3-methyladenine (3MA) also results in a significant decrease in mitochondrial function and network length. These data show that both the mitochondria and autophagy are critical targets of 4-HNE, and that the proteins targeted by 4-HNE may change based on its concentration, persistently driving cellular dysfunction.


Asunto(s)
Aldehídos/metabolismo , Autofagia/fisiología , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Neuronas/fisiología , Estrés Oxidativo , Adenina/análogos & derivados , Adenina/farmacología , Aldehídos/análisis , Aldehídos/farmacología , Animales , Autofagia/efectos de los fármacos , Células Cultivadas , Metabolismo Energético , Dinámicas Mitocondriales , Neuronas/citología , Neuronas/efectos de los fármacos , Cultivo Primario de Células , Ratas
7.
Redox Biol ; 12: 648-656, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28395172

RESUMEN

Women living with HIV may present with high levels of body fat that are associated with altered bioenergetic function. Excess body fat may therefore exacerbate the bioenergetic dysfunction observed with HIV infection. To determine if body fat is associated with bioenergetic function in HIV, we conducted a cross-sectional study of 42 women with HIV who were virologically suppressed on antiretroviral therapy. Body composition was determined via dual-energy x-ray absorptiometry. Oxygen consumption rate (OCR) of monocytes was sorted from peripheral blood mononuclear cells obtained from participants in the fasting state. Differences in bioenergetic function, as measured by OCR, was assessed using Kruskal-Wallis tests and Spearman correlations adjusted for age, race, and smoking status. Participants were 86% Black, 45.5 years old, 48% current smokers, and 57% were obese (body mass index ≥30). Nearly all women (93%) had >30% total fat mass, while 12% had >50% total fat mass. Elevated levels of total fat mass, trunk fat, and leg fat were inversely correlated with measures of bioenergetic health as evidenced by lower maximal and reserve capacity OCR, and Bioenergetic Health Index. Measures of extracellular acidification (ECAR) in the absence (basal) or maximal (with oligomycin) were positively correlated with measures of bioenergetics, except proton leak, and were negatively correlated with fat mass. Despite virological suppression, women with HIV present with extremely high levels of adiposity that correlate with impaired bioenergetic health. Without effective interventions, this syndemic of HIV infection and obesity will likely have devastating consequences for our patients, potentially mediated through altered mitochondrial and glycolytic function.


Asunto(s)
Tejido Adiposo/diagnóstico por imagen , Infecciones por VIH/diagnóstico por imagen , Infecciones por VIH/metabolismo , Monocitos/fisiología , Obesidad/diagnóstico por imagen , Absorciometría de Fotón , Fármacos Anti-VIH/uso terapéutico , Composición Corporal , Estudios Transversales , Metabolismo Energético , Femenino , Infecciones por VIH/tratamiento farmacológico , Humanos , Persona de Mediana Edad , Consumo de Oxígeno
8.
Redox Biol ; 11: 73-81, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27889640

RESUMEN

Autophagy is an important cell recycling program responsible for the clearance of damaged or long-lived proteins and organelles. Pharmacological modulators of this pathway have been extensively utilized in a wide range of basic research and pre-clinical studies. Bafilomycin A1 and chloroquine are commonly used compounds that inhibit autophagy by targeting the lysosomes but through distinct mechanisms. Since it is now clear that mitochondrial quality control, particularly in neurons, is dependent on autophagy, it is important to determine whether these compounds modify cellular bioenergetics. To address this, we cultured primary rat cortical neurons from E18 embryos and used the Seahorse XF96 analyzer and a targeted metabolomics approach to measure the effects of bafilomycin A1 and chloroquine on bioenergetics and metabolism. We found that both bafilomycin and chloroquine could significantly increase the autophagosome marker LC3-II and inhibit key parameters of mitochondrial function, and increase mtDNA damage. Furthermore, we observed significant alterations in TCA cycle intermediates, particularly those downstream of citrate synthase and those linked to glutaminolysis. Taken together, these data demonstrate a significant impact of bafilomycin and chloroquine on cellular bioenergetics and metabolism consistent with decreased mitochondrial quality associated with inhibition of autophagy.


Asunto(s)
Autofagia/genética , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias/metabolismo , Neuronas/metabolismo , Animales , Cloroquina/farmacología , Daño del ADN/efectos de los fármacos , ADN Mitocondrial/efectos de los fármacos , Metabolismo Energético/genética , Lisosomas/efectos de los fármacos , Lisosomas/genética , Macrólidos/farmacología , Metabolómica/métodos , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/efectos de los fármacos , Ratas
9.
Cell Metab ; 23(5): 930-44, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27133131

RESUMEN

Human platelets acutely increase mitochondrial energy generation following stimulation. Herein, a lipidomic circuit was uncovered whereby the substrates for this are exclusively provided by cPLA2, including multiple fatty acids and oxidized species that support energy generation via ß-oxidation. This indicates that acute lipid membrane remodeling is required to support energetic demands during platelet activation. Phospholipase activity is linked to energy metabolism, revealing cPLA2 as a central regulator of both lipidomics and energy flux. Using a lipidomic approach (LipidArrays), we also estimated the total number of lipids in resting, thrombin-activated, and aspirinized platelets. Significant diversity between genetically unrelated individuals and a wealth of species was revealed. Resting platelets demonstrated ∼5,600 unique species, with only ∼50% being putatively identified. Thrombin elevated ∼900 lipids >2-fold with 86% newly appearing and 45% inhibited by aspirin supplementation, indicating COX-1 is required for major activation-dependent lipidomic fluxes. Many lipids were structurally identified. With ∼50% of the lipids being absent from databases, a major opportunity for mining lipids relevant to human health and disease is presented.


Asunto(s)
Plaquetas/metabolismo , Metabolismo Energético , Metaboloma , Mitocondrias/metabolismo , Fosfolipasas A2 Citosólicas/metabolismo , Aspirina/farmacología , Plaquetas/efectos de los fármacos , Eicosanoides/metabolismo , Metabolismo Energético/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Ácidos Grasos/metabolismo , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Metaboloma/efectos de los fármacos , Oxidación-Reducción , Fosfolípidos/metabolismo , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato/efectos de los fármacos , Trombina/farmacología , Factores de Tiempo
10.
Aging Dis ; 7(2): 150-62, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27114848

RESUMEN

Redox and proteotoxic stress contributes to age-dependent accumulation of dysfunctional mitochondria and protein aggregates, and is associated with neurodegeneration. The free radical theory of aging inspired many studies using reactive species scavengers such as alpha-tocopherol, ascorbate and coenzyme Q to suppress the initiation of oxidative stress. However, clinical trials have had limited success in the treatment of neurodegenerative diseases. We ascribe this to the emerging literature which suggests that the oxidative stress hypothesis does not encompass the role of reactive species in cell signaling and therefore the interception with reactive species with antioxidant supplementation may result in disruption of redox signaling. In addition, the accumulation of redox modified proteins or organelles cannot be reversed by oxidant intercepting antioxidants and must then be removed by alternative mechanisms. We have proposed that autophagy serves this essential function in removing damaged or dysfunctional proteins and organelles thus preserving neuronal function and survival. In this review, we will highlight observations regarding the impact of autophagy regulation on cellular bioenergetics and survival in response to reactive species or reactive species generating compounds, and in response to proteotoxic stress.

11.
Redox Biol ; 8: 43-50, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26748041

RESUMEN

Metabolic and bioenergetic dysfunction are associated with oxidative stress and thought to be a common underlying mechanism of chronic diseases such as atherosclerosis, diabetes, and neurodegeneration. Recent findings support an emerging concept that circulating leukocytes and platelets can act as sensors or biomarkers of mitochondrial function in patients subjected to metabolic diseases. It is proposed that systemic stress-induced alterations in leukocyte bioenergetics are the consequence of several factors including reactive oxygen species. This suggests that oxidative stress mediated changes in leukocyte mitochondrial function could be used as an indicator of bioenergetic health in individuals. To test this concept, we investigated the effect of the redox cycling agent, 2,3 dimethoxynaphthoquinone (DMNQ) on the bioenergetic profiles of monocytes isolated from healthy human subjects using the extracellular flux analyzer. In addition, we tested the hypothesis that the bioenergetic health index (BHI), a single value that represents the bioenergetic health of individuals, is dynamically sensitive to oxidative stress in human monocytes. DMNQ decreased monocyte ATP-linked respiration, maximal respiration, and reserve capacity and caused an increase in proton leak and non-mitochondrial respiration compared to monocytes not treated with DMNQ. The BHI was a more sensitive indicator of the DMNQ-dependent changes in bioenergetics than any individual parameter. These data suggest that monocytes are susceptible to oxidative stress mediated by DMNQ and this can be accurately assessed by the BHI. Taken together, our findings suggest that the BHI has the potential to act as a functional biomarker of the impact of systemic oxidative stress in patients with metabolic disorders.


Asunto(s)
Metabolismo Energético , Monocitos/metabolismo , Estrés Oxidativo , Adulto , Biomarcadores , Metabolismo Energético/efectos de los fármacos , Femenino , Humanos , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Monocitos/efectos de los fármacos , Naftoquinonas/farmacología , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
12.
Biosci Rep ; 35(4)2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26181371

RESUMEN

Monitoring the bioenergetics of leucocytes is now emerging as an important approach in translational research to detect mitochondrial dysfunction in blood or other patient samples. Using the mitochondrial stress test, which involves the sequential addition of mitochondrial inhibitors to adherent leucocytes, we have calculated a single value, the Bioenergetic Health Index (BHI), which represents the mitochondrial function in cells isolated from patients. In the present report, we assess the BHI of monocytes isolated from the post-operative blood and post-operative pericardial fluid (PO-PCF) from patients undergoing cardiac surgery. Analysis of the bioenergetics of monocytes isolated from patients' PO-PCF revealed a profound decrease in mitochondrial function compared with monocytes isolated from their blood or from healthy controls. Further, patient blood monocytes showed no significant difference in the individual energetic parameters from the mitochondrial stress test but, when integrated into the BHI evaluation, there was a significant decrease in BHI compared with healthy control monocytes. These data support the utility of BHI measurements in integrating the individual parameters from the mitochondrial stress test into a single value. Supporting our previous finding that the PO-PCF is pro-oxidant, we found that exposure of rat cardiomyocytes to PO-PCF caused a significant loss of mitochondrial membrane potential and increased reactive oxygen species (ROS). These findings support the hypothesis that integrated measures of bioenergetic health could have prognostic and diagnostic value in translational bioenergetics.


Asunto(s)
Metabolismo Energético , Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Monocitos/metabolismo , Isquemia Miocárdica/metabolismo , Líquido Pericárdico/metabolismo , Animales , Procedimientos Quirúrgicos Cardíacos , Femenino , Humanos , Masculino , Mitocondrias/patología , Monocitos/patología , Isquemia Miocárdica/patología , Isquemia Miocárdica/cirugía , Ratas
13.
Biochem J ; 467(3): 517-27, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25742174

RESUMEN

The apoA-I (apolipoprotein A-I) mimetic peptide 4F favours the differentiation of human monocytes to an alternatively activated M2 phenotype. The goal of the present study was to test whether the 4F-mediated differentiation of MDMs (monocyte-derived macrophages) requires the induction of an oxidative metabolic programme. 4F treatment induced several genes in MDMs that play an important role in lipid metabolism, including PPARγ (peroxisome-proliferator-activated receptor γ) and CD36. Addition of 4F was associated with a significant increase in FA (fatty acid) uptake and oxidation compared with vehicle treatment. Mitochondrial respiration was assessed by measurement of the OCR (oxygen-consumption rate). 4F increased basal and ATP-linked OCR as well as maximal uncoupled mitochondrial respiration. These changes were associated with a significant increase in ΔΨm (mitochondrial membrane potential). The increase in metabolic activity in 4F-treated MDMs was attenuated by etomoxir, an inhibitor of mitochondrial FA uptake. Finally, addition of the PPARγ antagonist T0070907 to 4F-treated MDMs reduced the expression of CD163 and CD36, cell-surface markers for M2 macrophages, and reduced basal and ATP-linked OCR. These results support our hypothesis that the 4F-mediated differentiation of MDMs to an anti-inflammatory phenotype is due, in part, to an increase in FA uptake and mitochondrial oxidative metabolism.


Asunto(s)
Apolipoproteína A-I/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Péptidos/farmacología , Antiinflamatorios/farmacología , Benzamidas/farmacología , Materiales Biomiméticos/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Metabolismo Energético , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Macrófagos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Consumo de Oxígeno , PPAR gamma/antagonistas & inhibidores , Piridinas/farmacología
14.
J Neurochem ; 131(5): 625-33, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25081478

RESUMEN

Parkinson's disease is the second most common neurodegenerative disorder with both mitochondrial dysfunction and insufficient autophagy playing a key role in its pathogenesis. Among the risk factors, exposure to the environmental neurotoxin rotenone increases the probability of developing Parkinson's disease. We previously reported that in differentiated SH-SY5Y cells, rotenone-induced cell death is directly related to inhibition of mitochondrial function. How rotenone at nM concentrations inhibits mitochondrial function, and whether it can engage the autophagy pathway necessary to remove damaged proteins and organelles, is unknown. We tested the hypothesis that autophagy plays a protective role against rotenone toxicity in primary neurons. We found that rotenone (10-100 nM) immediately inhibited cellular bioenergetics. Concentrations that decreased mitochondrial function at 2 h, caused cell death at 24 h with an LD50 of 10 nM. Overall, autophagic flux was decreased by 10 nM rotenone at both 2 and 24 h, but surprisingly mitophagy, or autophagy of the mitochondria, was increased at 24 h, suggesting that a mitochondrial-specific lysosomal degradation pathway may be activated. Up-regulation of autophagy by rapamycin protected against cell death while inhibition of autophagy by 3-methyladenine exacerbated cell death. Interestingly, while 3-methyladenine exacerbated the rotenone-dependent effects on bioenergetics, rapamycin did not prevent rotenone-induced mitochondrial dysfunction, but caused reprogramming of mitochondrial substrate usage associated with both complex I and complex II activities. Taken together, these data demonstrate that autophagy can play a protective role in primary neuron survival in response to rotenone; moreover, surviving neurons exhibit bioenergetic adaptations to this metabolic stressor.


Asunto(s)
Autofagia/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Insecticidas/farmacología , Rotenona/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Daño del ADN/efectos de los fármacos , ADN Mitocondrial/antagonistas & inhibidores , ADN Mitocondrial/genética , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Lactosilceramidos/farmacología , Neuronas/efectos de los fármacos , Oligomicinas/farmacología , Consumo de Oxígeno/efectos de los fármacos , Ratas , Sirolimus/farmacología
15.
Clin Sci (Lond) ; 127(6): 367-73, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24895057

RESUMEN

Bioenergetics has become central to our understanding of pathological mechanisms, the development of new therapeutic strategies and as a biomarker for disease progression in neurodegeneration, diabetes, cancer and cardiovascular disease. A key concept is that the mitochondrion can act as the 'canary in the coal mine' by serving as an early warning of bioenergetic crisis in patient populations. We propose that new clinical tests to monitor changes in bioenergetics in patient populations are needed to take advantage of the early and sensitive ability of bioenergetics to determine severity and progression in complex and multifactorial diseases. With the recent development of high-throughput assays to measure cellular energetic function in the small number of cells that can be isolated from human blood these clinical tests are now feasible. We have shown that the sequential addition of well-characterized inhibitors of oxidative phosphorylation allows a bioenergetic profile to be measured in cells isolated from normal or pathological samples. From these data we propose that a single value-the Bioenergetic Health Index (BHI)-can be calculated to represent the patient's composite mitochondrial profile for a selected cell type. In the present Hypothesis paper, we discuss how BHI could serve as a dynamic index of bioenergetic health and how it can be measured in platelets and leucocytes. We propose that, ultimately, BHI has the potential to be a new biomarker for assessing patient health with both prognostic and diagnostic value.


Asunto(s)
Metabolismo Energético , Mitocondrias/metabolismo , Investigación Biomédica Traslacional , Animales , Biomarcadores/metabolismo , Humanos , Estrés Oxidativo/fisiología
16.
J Vis Exp ; (85)2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24747339

RESUMEN

Mitochondrial dysfunction is known to play a significant role in a number of pathological conditions such as atherosclerosis, diabetes, septic shock, and neurodegenerative diseases but assessing changes in bioenergetic function in patients is challenging. Although diseases such as diabetes or atherosclerosis present clinically with specific organ impairment, the systemic components of the pathology, such as hyperglycemia or inflammation, can alter bioenergetic function in circulating leukocytes or platelets. This concept has been recognized for some time but its widespread application has been constrained by the large number of primary cells needed for bioenergetic analysis. This technical limitation has been overcome by combining the specificity of the magnetic bead isolation techniques, cell adhesion techniques, which allow cells to be attached without activation to microplates, and the sensitivity of new technologies designed for high throughput microplate respirometry. An example of this equipment is the extracellular flux analyzer. Such instrumentation typically uses oxygen and pH sensitive probes to measure rates of change in these parameters in adherent cells, which can then be related to metabolism. Here we detail the methods for the isolation and plating of monocytes, lymphocytes, neutrophils and platelets, without activation, from human blood and the analysis of mitochondrial bioenergetic function in these cells. In addition, we demonstrate how the oxidative burst in monocytes and neutrophils can also be measured in the same samples. Since these methods use only 8-20 ml human blood they have potential for monitoring reactive oxygen species generation and bioenergetics in a clinical setting.


Asunto(s)
Plaquetas/citología , Plaquetas/metabolismo , Técnicas Citológicas/métodos , Leucocitos/citología , Leucocitos/metabolismo , Estallido Respiratorio/fisiología , Metabolismo Energético , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Linfocitos/citología , Linfocitos/metabolismo , Mitocondrias/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo , Especies Reactivas de Oxígeno/metabolismo
17.
Redox Biol ; 2: 82-90, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24494187

RESUMEN

Oxidative stress including DNA damage, increased lipid and protein oxidation, are important features of aging and neurodegeneration suggesting that endogenous antioxidant protective pathways are inadequate or overwhelmed. Importantly, oxidative protein damage contributes to age-dependent accumulation of dysfunctional mitochondria or protein aggregates. In addition, environmental toxins such as rotenone and paraquat, which are risk factors for the pathogenesis of neurodegenerative diseases, also promote protein oxidation. The obvious approach of supplementing the primary antioxidant systems designed to suppress the initiation of oxidative stress has been tested in animal models and positive results were obtained. However, these findings have not been effectively translated to treating human patients, and clinical trials for antioxidant therapies using radical scavenging molecules such as α-tocopherol, ascorbate and coenzyme Q have met with limited success, highlighting several limitations to this approach. These could include: (1) radical scavenging antioxidants cannot reverse established damage to proteins and organelles; (2) radical scavenging antioxidants are oxidant specific, and can only be effective if the specific mechanism for neurodegeneration involves the reactive species to which they are targeted and (3) since reactive species play an important role in physiological signaling, suppression of endogenous oxidants maybe deleterious. Therefore, alternative approaches that can circumvent these limitations are needed. While not previously considered an antioxidant system we propose that the autophagy-lysosomal activities, may serve this essential function in neurodegenerative diseases by removing damaged or dysfunctional proteins and organelles.


Asunto(s)
Antioxidantes/fisiología , Autofagia , Lisosomas/fisiología , Enfermedades Neurodegenerativas/metabolismo , Envejecimiento/metabolismo , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Antiparkinsonianos/farmacología , Antiparkinsonianos/uso terapéutico , Encéfalo/metabolismo , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Peroxidación de Lípido , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/patología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Oxidación-Reducción , Estrés Oxidativo , Oxidorreductasas/fisiología , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Ácido Peroxinitroso/metabolismo
18.
Redox Biol ; 2: 206-10, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24494194

RESUMEN

The assessment of metabolic function in cells isolated from human blood for treatment and diagnosis of disease is a new and important area of translational research. It is now becoming clear that a broad range of pathologies which present clinically with symptoms predominantly in one organ, such as the brain or kidney, also modulate mitochondrial energetics in platelets and leukocytes allowing these cells to serve as "the canary in the coal mine" for bioenergetic dysfunction. This opens up the possibility that circulating platelets and leukocytes can sense metabolic stress in patients and serve as biomarkers of mitochondrial dysfunction in human pathologies such as diabetes, neurodegeneration and cardiovascular disease. In this overview we will describe how the utilization of glycolysis and oxidative phosphorylation differs in platelets and leukocytes and discuss how they can be used in patient populations. Since it is clear that the metabolic programs between leukocytes and platelets are fundamentally distinct the measurement of mitochondrial function in distinct cell populations is necessary for translational research.


Asunto(s)
Plaquetas/metabolismo , Metabolismo Energético , Glucólisis/fisiología , Inflamación/sangre , Leucocitos/metabolismo , Mitocondrias/fisiología , Adenosina Trifosfato/biosíntesis , Animales , Aterosclerosis/sangre , Biomarcadores , Plaquetas/ultraestructura , Predicción , Humanos , Leucocitos/ultraestructura , Síndrome Metabólico/sangre , Neoplasias/sangre , Fosforilación Oxidativa , Estrés Oxidativo
19.
Free Radic Biol Med ; 65: 1215-1228, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24056030

RESUMEN

Excessive nitric oxide (NO) production is known to damage mitochondrial proteins and the autophagy repair pathway and so can potentially contribute to neurotoxicity. Accordingly, we hypothesized that protection against protein damage from reactive oxygen and nitrogen species under conditions of low oxygen by the autophagy pathway in neurons would be impaired by NO and enhance bioenergetic dysfunction. Rat primary cortical neurons had the same basal cellular respiration in hypoxia as in normoxia, whereas NO-exposed cells exhibited a gradual decrease in mitochondrial respiration in hypoxia. Upon reoxygenation, the respiration in NO-treated cells did not recover to prehypoxic levels. Hypoxia-reoxygenation in the presence of NO was associated with inhibition of autophagy, and the inability to recover during reoxygenation was exacerbated by an inhibitor of autophagy, 3-methyladenine. The effects of hypoxia could be recapitulated by inhibiting glycolytic flux under normoxic conditions. Under both normoxic and hypoxic conditions NO exposure induced immediate stimulation of glycolysis, but prolonged NO exposure, associated with irreversible inhibition of mitochondrial respiration in hypoxia, inhibited glycolysis. Importantly, we found that NO inhibited basal respiration under normoxic conditions only when glucose was absent from the medium or glycolysis was inhibited by 2-deoxy-d-glucose, revealing a novel NO-dependent mechanism for the inhibition of mitochondrial respiration that is modulated by glycolysis. Taken together these data suggest an oxygen-dependent interaction between mitochondrial respiration, glycolysis, and autophagy in protecting neuronal cells exposed to NO. Importantly, they indicate that mitochondrial dysfunction is intimately linked to a failure of glycolytic flux induced by exposure to NO. In addition, these studies provide new insights into the understanding of how autophagy and NO may play interactive roles in neuroinflammation-induced cellular damage, which is pertinent to our understanding of the pathology of neurodegenerative diseases in which excessive NO is generated.


Asunto(s)
Autofagia/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Glucólisis/efectos de los fármacos , Neuronas/metabolismo , Óxido Nítrico/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Hipoxia de la Célula , Supervivencia Celular , Células Cultivadas , Desoxiglucosa/farmacología , Células Endoteliales/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Ratas , Ratas Sprague-Dawley , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo
20.
PLoS One ; 8(6): e68348, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840849

RESUMEN

Solid tumors are characterized by regions of low oxygen tension (OT), which play a central role in tumor progression and resistance to therapy. Low OT affects mitochondrial function and for the cells to survive, mitochondria must functionally adapt to low OT to maintain the cellular bioenergetics. In this study, a novel experimental approach was developed to examine the real-time bioenergetic changes in breast cancer cells (BCCs) during adaptation to OT (from 20% to <1% oxygen) using sensitive extracellular flux technology. Oxygen was gradually removed from the medium, and the bioenergetics of metastatic BCCs (MDA-MB-231 and MCF10CA clones) was compared with non-tumorigenic (MCF10A) cells. BCCs, but not MCF10A, rapidly responded to low OT by stabilizing HIF-1α and increasing HIF-1α responsive gene expression and glucose uptake. BCCs also increased extracellular acidification rate (ECAR), which was markedly lower in MCF10A. Interestingly, BCCs exhibited a biphasic response in basal respiration as the OT was reduced from 20% to <1%. The initial stimulation of oxygen consumption is found to be due to increased mitochondrial respiration. This effect was HIF-1α-dependent, as silencing HIF-1α abolished the biphasic response. During hypoxia and reoxygenation, BCCs also maintained oxygen consumption rates at specific OT; however, HIF-1α silenced BCC were less responsive to changes in OT. Our results suggest that HIF-1α provides a high degree of bioenergetic flexibility under different OT which may confer an adaptive advantage for BCC survival in the tumor microenvironment and during invasion and metastasis. This study thus provides direct evidence for the cross-talk between HIF-1α and mitochondria during adaptation to low OT by BCCs and may be useful in identifying novel therapeutic agents that target the bioenergetics of BCCs in response to low OT.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Metabolismo Energético/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Mitocondrias/patología , Mitocondrias/fisiología , Metástasis de la Neoplasia/fisiopatología , Oxígeno/metabolismo , Adaptación Biológica/genética , Adaptación Biológica/fisiología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Respiración de la Célula/genética , Respiración de la Célula/fisiología , Metabolismo Energético/genética , Femenino , Glucosa/metabolismo , Glucólisis/genética , Glucólisis/fisiología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Metástasis de la Neoplasia/genética , Consumo de Oxígeno/genética , Consumo de Oxígeno/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA