Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cell Chem Biol ; 31(3): 593-606.e9, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38039968

ABSTRACT

Cerebral dopamine neurotrophic factor (CDNF) is an unconventional neurotropic factor that modulates unfolded protein response (UPR) pathway signaling and alleviates endoplasmic reticulum (ER) stress providing cytoprotective effects in different models of neurodegenerative disorders. Here, we developed a brain-penetrating peptidomimetic compound based on human CDNF. This compound called HER-096 shows similar potency and mechanism of action as CDNF, and promotes dopamine neuron survival, reduces α-synuclein aggregation and modulates UPR signaling in in vitro models. HER-096 is metabolically stable and able to penetrate to cerebrospinal (CSF) and brain interstitial fluids (ISF) after subcutaneous administration, with an extended CSF and brain ISF half-life compared to plasma. Subcutaneously administered HER-096 modulated UPR pathway activity, protected dopamine neurons, and reduced α-synuclein aggregates and neuroinflammation in substantia nigra of aged mice with synucleinopathy. Peptidomimetic HER-096 is a candidate for development of a disease-modifying therapy for Parkinson's disease with a patient-friendly route of administration.


Subject(s)
Parkinson Disease , Peptidomimetics , Synucleinopathies , Humans , Mice , Animals , Parkinson Disease/drug therapy , Dopaminergic Neurons , alpha-Synuclein , Peptidomimetics/pharmacology , Peptidomimetics/therapeutic use , Brain , Nerve Growth Factors
2.
JCI Insight ; 5(4)2020 02 27.
Article in English | MEDLINE | ID: mdl-32017711

ABSTRACT

The mitochondrial calcium uniporter is widely accepted as the primary route of rapid calcium entry into mitochondria, where increases in matrix calcium contribute to bioenergetics but also mitochondrial permeability and cell death. Hence, regulation of uniporter activity is critical to mitochondrial homeostasis. The uniporter subunit EMRE is known to be an essential regulator of the channel-forming protein MCU in cell culture, but EMRE's impact on organismal physiology is less understood. Here we characterize a mouse model of EMRE deletion and show that EMRE is indeed required for mitochondrial calcium uniporter function in vivo. EMRE-/- mice are born less frequently; however, the mice that are born are viable, healthy, and do not manifest overt metabolic impairment, at rest or with exercise. Finally, to investigate the role of EMRE in disease processes, we examine the effects of EMRE deletion in a muscular dystrophy model associated with mitochondrial calcium overload.


Subject(s)
Calcium Channels/physiology , Mitochondrial Membrane Transport Proteins/physiology , Animals , Calcium/metabolism , Disease Models, Animal , Heart/physiopathology , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Myocardial Reperfusion Injury/metabolism
3.
Cardiovasc Res ; 115(2): 385-394, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30165576

ABSTRACT

Aims: Knockout (KO) of the mitochondrial Ca2+ uniporter (MCU) in mice abrogates mitochondrial Ca2+ uptake and permeability transition pore (PTP) opening. However, hearts from global MCU-KO mice are not protected from ischaemic injury. We aimed to investigate whether adaptive alterations occur in cell death signalling pathways in the hearts of global MCU-KO mice. Methods and results: First, we examined whether cell death may occur via an upregulation in necroptosis in MCU-KO mice. However, our results show that neither RIP1 inhibition nor RIP3 knockout afford protection against ischaemia-reperfusion injury in MCU-KO as in wildtype (WT) hearts, indicating that the lack of protection cannot be explained by upregulation of necroptosis. Instead, we have identified alterations in cyclophilin D (CypD) signalling in MCU-KO hearts. In the presence of a calcium ionophore, MCU-KO mitochondria take up calcium and do undergo PTP opening. Furthermore, PTP opening in MCU-KO mitochondria has a lower calcium retention capacity (CRC), suggesting that the calcium sensitivity of PTP is higher. Phosphoproteomics identified an increase in phosphorylation of CypD-S42 in MCU-KO. We investigated the interaction of CypD with the putative PTP component ATP synthase and identified an approximately 50% increase in this interaction in MCU-KO cardiac mitochondria. Mutation of the novel CypD phosphorylation site S42 to a phosphomimic reduced CRC, increased CypD-ATP synthase interaction by approximately 50%, and increased cell death in comparison to a phospho-resistant mutant. Conclusion: Taken together these data suggest that MCU-KO mitochondria exhibit an increase in phosphorylation of CypD-S42 which decreases PTP calcium sensitivity thus allowing activation of PTP in the absence of an MCU-mediated increase in matrix calcium.


Subject(s)
Calcium Channels/deficiency , Calcium/metabolism , Cyclophilins/metabolism , Mitochondria, Heart/enzymology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/deficiency , Myocardial Infarction/enzymology , Myocardial Reperfusion Injury/enzymology , Myocardium/enzymology , Animals , Calcium Channels/genetics , Cyclophilins/genetics , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/metabolism , Mice, Knockout , Mitochondria, Heart/drug effects , Mitochondria, Heart/pathology , Mitochondrial Permeability Transition Pore , Mitochondrial Proteins/genetics , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction
4.
Dis Model Mech ; 10(2): 163-171, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28067626

ABSTRACT

Plants and many lower organisms, but not mammals, express alternative oxidases (AOXs) that branch the mitochondrial respiratory chain, transferring electrons directly from ubiquinol to oxygen without proton pumping. Thus, they maintain electron flow under conditions when the classical respiratory chain is impaired, limiting excess production of oxygen radicals and supporting redox and metabolic homeostasis. AOX from Ciona intestinalis has been used to study and mitigate mitochondrial impairments in mammalian cell lines, Drosophila disease models and, most recently, in the mouse, where multiple lentivector-AOX transgenes conferred substantial expression in specific tissues. Here, we describe a genetically tractable mouse model in which Ciona AOX has been targeted to the Rosa26 locus for ubiquitous expression. The AOXRosa26 mouse exhibited only subtle phenotypic effects on respiratory complex formation, oxygen consumption or the global metabolome, and showed an essentially normal physiology. AOX conferred robust resistance to inhibitors of the respiratory chain in organello; moreover, animals exposed to a systemically applied LD50 dose of cyanide did not succumb. The AOXRosa26 mouse is a useful tool to investigate respiratory control mechanisms and to decipher mitochondrial disease aetiology in vivo.


Subject(s)
Mitochondrial Proteins/metabolism , Oxidoreductases/metabolism , Physiological Phenomena , Plant Proteins/metabolism , Animals , Ciona intestinalis/enzymology , Cyanides/administration & dosage , Cyanides/toxicity , Mice, Transgenic , Mitochondria/metabolism , Protective Agents/metabolism , RNA, Untranslated/genetics
5.
J Biol Chem ; 291(44): 23343-23362, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27637331

ABSTRACT

Control of myocardial energetics by Ca2+ signal propagation to the mitochondrial matrix includes local Ca2+ delivery from sarcoplasmic reticulum (SR) ryanodine receptors (RyR2) to the inner mitochondrial membrane (IMM) Ca2+ uniporter (mtCU). mtCU activity in cardiac mitochondria is relatively low, whereas the IMM surface is large, due to extensive cristae folding. Hence, stochastically distributed mtCU may not suffice to support local Ca2+ transfer. We hypothesized that mtCU concentrated at mitochondria-SR associations would promote the effective Ca2+ transfer. mtCU distribution was determined by tracking MCU and EMRE, the proteins essential for channel formation. Both proteins were enriched in the IMM-outer mitochondrial membrane (OMM) contact point submitochondrial fraction and, as super-resolution microscopy revealed, located more to the mitochondrial periphery (inner boundary membrane) than inside the cristae, indicating high accessibility to cytosol-derived Ca2+ inputs. Furthermore, MCU immunofluorescence distribution was biased toward the mitochondria-SR interface (RyR2), and this bias was promoted by Ca2+ signaling activity in intact cardiomyocytes. The SR fraction of heart homogenate contains mitochondria with extensive SR associations, and these mitochondria are highly enriched in EMRE. Size exclusion chromatography suggested for EMRE- and MCU-containing complexes a wide size range and also revealed MCU-containing complexes devoid of EMRE (thus disabled) in the mitochondrial but not the SR fraction. Functional measurements suggested more effective mtCU-mediated Ca2+ uptake activity by the mitochondria of the SR than of the mitochondrial fraction. Thus, mtCU "hot spots" can be formed at the cardiac muscle mitochondria-SR associations via localization and assembly bias, serving local Ca2+ signaling and the excitation-energetics coupling.


Subject(s)
Calcium Channels/metabolism , Myocardium/metabolism , Animals , Calcium/metabolism , Calcium Channels/genetics , Calcium Signaling , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Mitochondrial Membranes/metabolism , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum/genetics , Sarcoplasmic Reticulum/metabolism
6.
Cell Rep ; 16(6): 1561-1573, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27477272

ABSTRACT

MICU1 is a component of the mitochondrial calcium uniporter, a multiprotein complex that also includes MICU2, MCU, and EMRE. Here, we describe a mouse model of MICU1 deficiency. MICU1(-/-) mitochondria demonstrate altered calcium uptake, and deletion of MICU1 results in significant, but not complete, perinatal mortality. Similar to afflicted patients, viable MICU1(-/-) mice manifest marked ataxia and muscle weakness. Early in life, these animals display a range of biochemical abnormalities, including increased resting mitochondrial calcium levels, altered mitochondrial morphology, and reduced ATP. Older MICU1(-/-) mice show marked, spontaneous improvement coincident with improved mitochondrial calcium handling and an age-dependent reduction in EMRE expression. Remarkably, deleting one allele of EMRE helps normalize calcium uptake while simultaneously rescuing the high perinatal mortality observed in young MICU1(-/-) mice. Together, these results demonstrate that MICU1 serves as a molecular gatekeeper preventing calcium overload and suggests that modulating the calcium uniporter could have widespread therapeutic benefits.


Subject(s)
Calcium Channels/metabolism , Calcium-Binding Proteins/metabolism , Calcium/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Animals , Calcium-Binding Proteins/genetics , Cation Transport Proteins/metabolism , Membrane Potential, Mitochondrial/physiology , Mice, Knockout , Mitochondrial Membrane Transport Proteins/genetics
7.
Curr Opin Toxicol ; 1: 80-91, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28066829

ABSTRACT

The transcription factor nuclear factor erythroid 2 p45-related factor 2 (Nrf2) is the master regulator of the cellular redox homeostasis. Nrf2 target genes comprise of a large network of antioxidant enzymes, proteins involved in xenobiotic detoxification, repair and removal of damaged proteins, inhibition of inflammation, as well as other transcription factors. In recent years it has emerged that as part of its role as a regulator of cytoprotective gene expression, Nrf2 impacts mitochondrial function. Increased Nrf2 activity defends against mitochondrial toxins. Reduced glutathione, the principal small molecule antioxidant in the mammalian cell and a product of several of the downstream target genes of Nrf2, counterbalances mitochondrial ROS production. The function of Nrf2 is suppressed in mitochondria-related disorders, such as Parkinson's disease and Friedrich's ataxia. Studies using isolated mitochondria and cultured cells have demonstrated that Nrf2 deficiency leads to impaired mitochondrial fatty acid oxidation, respiration and ATP production. Small molecule activators of Nrf2 support mitochondrial integrity by promoting mitophagy and conferring resistance to oxidative stress-mediated permeability transition. Excitingly, recent studies have shown that Nrf2 also affects mitochondrial function in stem cells with implications for stem cell self-renewal, cardiomyocyte regeneration, and neural stem/progenitor cell survival.

8.
Mol Cell ; 60(4): 685-96, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26549682

ABSTRACT

Alterations in mitophagy have been increasingly linked to aging and age-related diseases. There are, however, no convenient methods to analyze mitophagy in vivo. Here, we describe a transgenic mouse model in which we expressed a mitochondrial-targeted form of the fluorescent reporter Keima (mt-Keima). Keima is a coral-derived protein that exhibits both pH-dependent excitation and resistance to lysosomal proteases. Comparison of a wide range of primary cells and tissues generated from the mt-Keima mouse revealed significant variations in basal mitophagy. In addition, we have employed the mt-Keima mice to analyze how mitophagy is altered by conditions including diet, oxygen availability, Huntingtin transgene expression, the absence of macroautophagy (ATG5 or ATG7 expression), an increase in mitochondrial mutational load, the presence of metastatic tumors, and normal aging. The ability to assess mitophagy under a host of varying environmental and genetic perturbations suggests that the mt-Keima mouse should be a valuable resource.


Subject(s)
Luminescent Proteins/metabolism , Mice, Transgenic , Mitophagy , Aging/physiology , Animals , Luminescent Proteins/genetics , Mice , Organ Specificity , Oxygen/metabolism
9.
Biochem Soc Trans ; 43(4): 602-10, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26551700

ABSTRACT

The Kelch-like ECH associated protein 1 (Keap1)-NF-E2 p45-related factor 2 (Nrf2) pathway regulates networks of proteins that protect against the cumulative damage of oxidants, electrophiles and misfolded proteins. The interaction between transcription factor Nrf2 and its main negative cytoplasmic regulator Keap1 follows a cycle whereby the protein complex sequentially adopts two conformations: 'open', in which Nrf2 binds to one monomer of Keap1, followed by 'closed', in which Nrf2 interacts with both members of the Keap1 dimer. Electrophiles and oxidants (inducers) are recognized by cysteine sensors within Keap1, disrupting its ability to target Nrf2 for ubiquitination and degradation. Consequently, the protein complex accumulates in the 'closed' conformation, free Keap1 is not regenerated and newly synthesized Nrf2 is stabilized to activate target-gene transcription. The prevailing view of the Keap1-Nrf2 pathway, for which there exists a wealth of experimental evidence, is that it lies at the heart of cellular defence, playing crucial roles in adaptation and survival under conditions of stress. More recently, the significance of Nrf2 in intermediary metabolism and mitochondrial physiology has also been recognized, adding another layer of cytoprotection to the repertoire of functions of Nrf2. One way by which Nrf2 influences mitochondrial activity is through increasing the availability of substrates (NADH and FADH2) for respiration. Another way is through accelerating fatty acid oxidation (FAO). These findings reinforce the reciprocal relationship between oxidative phosphorylation and the cellular redox state, and highlight the key role of Nrf2 in regulating this balance.


Subject(s)
Energy Metabolism , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Kelch-Like ECH-Associated Protein 1 , Mitochondria/metabolism , Models, Molecular , NF-E2-Related Factor 2/chemistry , Oxidation-Reduction , Signal Transduction
10.
J Mol Cell Cardiol ; 85: 178-82, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26057074

ABSTRACT

Mitochondrial calcium is thought to play an important role in the regulation of cardiac bioenergetics and function. The entry of calcium into the mitochondrial matrix requires that the divalent cation pass through the inner mitochondrial membrane via a specialized pore known as the mitochondrial calcium uniporter (MCU). Here, we use mice deficient of MCU expression to rigorously assess the role of mitochondrial calcium in cardiac function. Mitochondria isolated from MCU(-/-) mice have reduced matrix calcium levels, impaired calcium uptake and a defect in calcium-stimulated respiration. Nonetheless, we find that the absence of MCU expression does not affect basal cardiac function at either 12 or 20months of age. Moreover, the physiological response of MCU(-/-) mice to isoproterenol challenge or transverse aortic constriction appears similar to control mice. Thus, while mitochondria derived from MCU(-/-) mice have markedly impaired mitochondrial calcium handling, the hearts of these animals surprisingly appear to function relatively normally under basal conditions and during stress.


Subject(s)
Calcium Channels/genetics , Animals , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling , Female , Mice, Knockout , Mitochondria, Heart/metabolism , Stroke Volume
11.
Circ Res ; 116(11): 1810-9, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25999421

ABSTRACT

Calcium is thought to play an important role in regulating mitochondrial function. Evidence suggests that an increase in mitochondrial calcium can augment ATP production by altering the activity of calcium-sensitive mitochondrial matrix enzymes. In contrast, the entry of large amounts of mitochondrial calcium in the setting of ischemia-reperfusion injury is thought to be a critical event in triggering cellular necrosis. For many decades, the details of how calcium entered the mitochondria remained a biological mystery. In the past few years, significant progress has been made in identifying the molecular components of the mitochondrial calcium uniporter complex. Here, we review how calcium enters and leaves the mitochondria, the growing insight into the topology, stoichiometry and function of the uniporter complex, and the early lessons learned from some initial mouse models that genetically perturb mitochondrial calcium homeostasis.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Humans , Ion Transport , Models, Biological
12.
Biochim Biophys Acta ; 1850(4): 794-801, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25484314

ABSTRACT

BACKGROUND: Nuclear factor (erythroid-derived 2) factor 2 (Nrf2) is a crucial transcription factor mediating protection against oxidants. Nrf2 is negatively regulated by cytoplasmic Kelch-like ECH associated protein 1 (Keap1) thereby providing inducible antioxidant defence. Antioxidant properties of Nrf2 are thought to be mainly exerted by stimulating transcription of antioxidant proteins, whereas its effects on ROS production within the cell are uncertain. METHODS: Live cell imaging and qPCR in brain hippocampal glio-neuronal cultures and explants slice cultures with graded expression of Nrf2, i.e. Nrf2-knockout (Nrf2-KO), wild-type (WT), and Keap1-knockdown (Keap1-KD). RESULTS: We here show that ROS production in Nrf2-KO cells and tissues is increased compared to their WT counterparts. Mitochondrial ROS production is regulated by the Keap1-Nrf2 pathway by controlling mitochondrial bioenergetics. Surprisingly, Keap1-KD cells and tissues also showed higher rates of ROS production when compared to WT, although with a smaller magnitude. Analysis of the mRNA expression levels of the two NOX isoforms implicated in brain pathology showed, that NOX2 is dramatically upregulated under conditions of Nrf2 deficiency, whereas NOX4 is upregulated when Nrf2 is constitutively activated (Keap1-KD) to a degree which paralleled the increases in ROS production. CONCLUSIONS: These observations suggest that the Keap1-Nrf2 pathway regulates both mitochondrial and cytosolic ROS production through NADPH oxidase. GENERAL SIGNIFICANCE: Findings supports a key role of the Keap1-Nrf2 pathway in redox homeostasis within the cell.


Subject(s)
Mitochondria/metabolism , NADPH Oxidases/physiology , NF-E2-Related Factor 2/physiology , Reactive Oxygen Species/metabolism , Animals , Cytosol/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Kelch-Like ECH-Associated Protein 1 , Mice , Mice, Hairless
13.
Nat Rev Mol Cell Biol ; 15(6): 411-21, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24854789

ABSTRACT

Reactive oxygen species (ROS), which were originally characterized in terms of their harmful effects on cells and invading microorganisms, are increasingly implicated in various cell fate decisions and signal transduction pathways. The mechanism involved in ROS-dependent signalling involves the reversible oxidation and reduction of specific amino acids, with crucial reactive Cys residues being the most frequent target. In this Review, we discuss the sources of ROS within cells and what is known regarding how intracellular oxidant levels are regulated. We further discuss the recent observations that reduction-oxidation (redox)-dependent regulation has a crucial role in an ever-widening range of biological activities - from immune function to stem cell self-renewal, and from tumorigenesis to ageing.


Subject(s)
Aging/pathology , Cell Physiological Phenomena , Neoplasms/physiopathology , Reactive Oxygen Species/metabolism , Signal Transduction , Animals , Humans , Oxidation-Reduction
14.
Biochem Biophys Res Commun ; 449(4): 384-5, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24792186

ABSTRACT

Entry of mitochondrial calcium is believed to play an essential role in regulating bioenergetics and initiating cell death pathways. We have recently described a mouse model lacking MCU expression. Surprisingly, these mice are viable and the cells and tissues from these animals do not exhibit any marked protection from cell death. Here, we discuss our findings as well as potential explanations for some of the more unexpected results.


Subject(s)
Calcium Channels/deficiency , Calcium Channels/genetics , Calcium/metabolism , Mitochondria/metabolism , Animals , Cell Death , Cell Line , Mice , Mitochondrial Membrane Transport Proteins/metabolism
15.
Biochemistry ; 53(15): 2442-53, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24717093

ABSTRACT

Oligomers of the amyloid-ß (Aß) peptide have been implicated in the neurotoxicity associated with Alzheimer's disease. We have used single-molecule techniques to examine quantitatively the cellular effects of adding well characterized Aß oligomers to primary hippocampal cells and hence determine the initial pathway of damage. We found that even picomolar concentrations of Aß (1-40) and Aß (1-42) oligomers can, within minutes of addition, increase the levels of intracellular calcium in astrocytes but not in neurons, and this effect is saturated at a concentration of about 10 nM of oligomers. Both Aß (1-40) and Aß (1-42) oligomers have comparable effects. The rise in intracellular calcium is followed by an increase in the rate of ROS production by NADPH oxidase in both neurons and astrocytes. The increase in ROS production then triggers caspase-3 activation resulting in the inhibition of long-term potentiation. Our quantitative approach also reveals that only a small fraction of the oligomers are damaging and that an individual rare oligomer binding to an astrocyte can initiate the aforementioned cascade of responses, making it unlikely to be due to any specific interaction. Preincubating the Aß oligomers with an extracellular chaperone, clusterin, sequesters the oligomers in long-lived complexes and inhibits all of the physiological damage, even at a ratio of 100:1, total Aß to clusterin. To explain how Aß oligomers are so damaging but that it takes decades to develop Alzheimer's disease, we suggest a model for disease progression where small amounts of neuronal damage from individual unsequestered oligomers can accumulate over time leading to widespread tissue-level dysfunction.


Subject(s)
Amyloid beta-Peptides/physiology , Astrocytes/cytology , Biopolymers/physiology , Neurons/cytology , Animals , Astrocytes/enzymology , Astrocytes/metabolism , Calcium/metabolism , Caspase 3/metabolism , Enzyme Activation , Neurons/enzymology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
16.
Biol Open ; 2(8): 761-70, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23951401

ABSTRACT

Transcription factor Nrf2 and its repressor Keap1 regulate a network of cytoprotective genes involving more than 1% of the genome, their best known targets being drug-metabolizing and antioxidant genes. Here we demonstrate a novel role for this pathway in directly regulating mitochondrial bioenergetics in murine neurons and embryonic fibroblasts. Loss of Nrf2 leads to mitochondrial depolarisation, decreased ATP levels and impaired respiration, whereas genetic activation of Nrf2 increases the mitochondrial membrane potential and ATP levels, the rate of respiration and the efficiency of oxidative phosphorylation. We further show that Nrf2-deficient cells have increased production of ATP in glycolysis, which is then used by the F1Fo-ATPase for maintenance of the mitochondrial membrane potential. While the levels and in vitro activities of the respiratory complexes are unaffected by Nrf2 deletion, their activities in isolated mitochondria and intact live cells are substantially impaired. In addition, the rate of regeneration of NADH after inhibition of respiration is much slower in Nrf2-knockout cells than in their wild-type counterparts. Taken together, these results show that Nrf2 directly regulates cellular energy metabolism through modulating the availability of substrates for mitochondrial respiration. Our findings highlight the importance of efficient energy metabolism in Nrf2-mediated cytoprotection.

17.
Int J Biochem Cell Biol ; 45(4): 899-907, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23353645

ABSTRACT

Aggregated amyloid-ß causes pathological changes in mixed cultures of neurons and astrocytes such as sporadic cytoplasmic intracellular Ca(2+)-signalling, increase in reactive oxygen species production and cell death. Some of the toxic effects of amyloid-ß are mediated through the interaction of the peptide with α7-type nicotinic acetylcholine receptors at the cell surface. Here we demonstrated that affinity purified antibodies to synthetic fragment 173-193 of the α7-subunit of the nAChR are able to protect cells from amyloid-ß induced cell death. The antibodies had no effect on the amyloid-ß induced calcium signal in astrocytes. However, they significantly reduced amyloid-ß induced and NADPH oxidase mediated ROS production. Modulation of the NADPH oxidase activity by either the antibodies, the receptor agonist acetylcholine or the antagonist of the α7-type nicotinic acetylcholine receptors α-bungarotoxin was vital in inhibiting both amyloid-ß induced ROS production, caspase 3 cleavage as well as cell death. The uncovered details of the mechanism underlying the action of antibodies to α7-type nicotinic acetylcholine receptors gives additional insight into the involvement of this receptor in Alzheimer's disease pathology and provides a new approach to anti-Alzheimer's disease vaccine design.


Subject(s)
Acetylcholine/pharmacology , Amyloid beta-Peptides/toxicity , Antibodies/immunology , Astrocytes/drug effects , Neurons/drug effects , Peptide Fragments/toxicity , Receptors, Nicotinic/immunology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Calcium Signaling/drug effects , Caspase 3/metabolism , Cell Death/drug effects , Enzyme Activation/drug effects , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , alpha7 Nicotinic Acetylcholine Receptor
18.
Cell Calcium ; 53(4): 256-63, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23340218

ABSTRACT

Glucocorticoids are steroid hormones which act through the glucocorticoid receptor. They regulate a wide variety of biological processes. Two glucocorticoids, the naturally occurring corticosterone and chemically produced dexamethasone, have been used to investigate the effect of glucocorticoids on Ca(2+)-signalling in cortical co-cultures of neurons and astrocytes. Dexamethasone and to a lesser degree corticosterone both induced a decrease in cytosolic Ca(2+) concentration in neurons and astrocytes. The effect of both compounds can be blocked by inhibition of the plasmamembrane ATPase, calmodulin and by application of a glucocorticoid receptor antagonist, while inhibition of NMDA receptors or the endoplasmic reticulum calcium pump had no effect. Glucocorticoid treatment further protects against detrimental calcium signalling and cell death by modulating the delayed calcium deregulation in response to glutamate toxicity. At the concentrations used dexamethasone and corticosterone did not show cell toxicity of their own. Thus, these results indicate that dexamethasone and corticosterone might be used for protection of the cells from calcium overload.


Subject(s)
Calcium/metabolism , Glucocorticoids/pharmacology , Glutamic Acid/toxicity , Neurons/drug effects , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Calcium/analysis , Calcium Signaling/drug effects , Cells, Cultured , Cytosol/chemistry , Cytosol/drug effects , Cytosol/metabolism , Dexamethasone/pharmacology , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley
19.
Nat Commun ; 4: 1362, 2013.
Article in English | MEDLINE | ID: mdl-23322050

ABSTRACT

Inorganic polyphosphate is known to be present in the mammalian brain at micromolar concentrations. Here we show that polyphosphate may act as a gliotransmitter, mediating communication between astrocytes. It is released by astrocytes in a calcium-dependent manner and signals to neighbouring astrocytes through P2Y(1) purinergic receptors, activation of phospholipase C and release of calcium from the intracellular stores. In primary neuroglial cultures, application of polyP triggers release of endogenous polyphosphate from astrocytes while neurons take it up. In vivo, central actions of polyphosphate at the level of the brainstem include profound increases in key homeostatic physiological activities, such as breathing, central sympathetic outflow and the arterial blood pressure. Together, these results suggest a role for polyphosphate as a mediator of astroglial signal transmission in the mammalian brain.


Subject(s)
Brain/metabolism , Mammals/metabolism , Polyphosphates/metabolism , Signal Transduction , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Brain/drug effects , Brain Stem/drug effects , Brain Stem/physiology , Calcium/pharmacology , Calcium Signaling/drug effects , Cells, Cultured , Coculture Techniques , Heart/drug effects , Heart/physiology , Male , Polyphosphates/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2Y1/metabolism , Respiration/drug effects , Signal Transduction/drug effects
20.
Am J Hum Genet ; 91(6): 1041-50, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23200863

ABSTRACT

In this study, we combined linkage analysis with whole-exome sequencing of two individuals to identify candidate causal variants in a moderately-sized UK kindred exhibiting autosomal-dominant inheritance of craniocervical dystonia. Subsequent screening of these candidate causal variants in a large number of familial and sporadic cases of cervical dystonia led to the identification of a total of six putatively pathogenic mutations in ANO3, a gene encoding a predicted Ca(2+)-gated chloride channel that we show to be highly expressed in the striatum. Functional studies using Ca(2+) imaging in case and control fibroblasts demonstrated clear abnormalities in endoplasmic-reticulum-dependent Ca(2+) signaling. We conclude that mutations in ANO3 are a cause of autosomal-dominant craniocervical dystonia. The locus DYT23 has been reserved as a synonym for this gene. The implication of an ion channel in the pathogenesis of dystonia provides insights into an alternative mechanism that opens fresh avenues for further research.


Subject(s)
Chloride Channels/genetics , Genes, Dominant , Mutation , Torticollis/genetics , Amino Acid Sequence , Anoctamins , Base Sequence , Calcium Signaling , Chloride Channels/metabolism , Corpus Striatum/metabolism , Dystonia , Endoplasmic Reticulum/metabolism , Exome , Female , Fibroblasts , Gene Expression Regulation , Genetic Linkage , High-Throughput Nucleotide Sequencing , Humans , Ion Channels/genetics , Male , Molecular Sequence Data , Pedigree , Phenotype , Sequence Alignment , Torticollis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...