Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters











Publication year range
1.
EMBO J ; 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261663

ABSTRACT

The mitochondrial calcium uniporter channel (MCUC) mediates mitochondrial calcium entry, regulating energy metabolism and cell death. Although several MCUC components have been identified, the molecular basis of mitochondrial calcium signaling networks and their remodeling upon changes in uniporter activity have not been assessed. Here, we map the MCUC interactome under resting conditions and upon chronic loss or gain of mitochondrial calcium uptake. We identify 89 high-confidence interactors that link MCUC to several mitochondrial complexes and pathways, half of which are associated with human disease. As a proof-of-concept, we validate the mitochondrial intermembrane space protein EFHD1 as a binding partner of the MCUC subunits MCU, EMRE, and MCUB. We further show a MICU1-dependent inhibitory effect of EFHD1 on calcium uptake. Next, we systematically survey compensatory mechanisms and functional consequences of mitochondrial calcium dyshomeostasis by analyzing the MCU interactome upon EMRE, MCUB, MICU1, or MICU2 knockdown. While silencing EMRE reduces MCU interconnectivity, MCUB loss-of-function leads to a wider interaction network. Our study provides a comprehensive and high-confidence resource to gain insights into players and mechanisms regulating mitochondrial calcium signaling and their relevance in human diseases.

2.
Sci Adv ; 10(32): eadp6182, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39121218

ABSTRACT

Endothelial cells (ECs) are highly plastic, capable of differentiating into various cell types. Endothelial-to-mesenchymal transition (EndMT) is crucial during embryonic development and contributes substantially to vascular dysfunction in many cardiovascular diseases (CVDs). While targeting EndMT holds therapeutic promise, understanding its mechanisms and modulating its pathways remain challenging. Using single-cell RNA sequencing on three in vitro EndMT models, we identified conserved gene signatures. We validated original regulators in vitro and in vivo during embryonic heart development and peripheral artery disease. EndMT induction led to global expression changes in all EC subtypes rather than in mesenchymal clusters. We identified mitochondrial calcium uptake as a key driver of EndMT; inhibiting mitochondrial calcium uniporter (MCU) prevented EndMT in vitro, and conditional Mcu deletion in ECs blocked mesenchymal activation in a hind limb ischemia model. Tissues from patients with critical limb ischemia with EndMT features exhibited significantly elevated endothelial MCU. These findings highlight MCU as a regulator of EndMT and a potential therapeutic target.


Subject(s)
Calcium Signaling , Endothelial Cells , Epithelial-Mesenchymal Transition , Mitochondria , RNA-Seq , Single-Cell Analysis , Animals , Humans , Mitochondria/metabolism , RNA-Seq/methods , Mice , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Calcium Channels/metabolism , Calcium Channels/genetics , Ischemia/metabolism , Ischemia/pathology , Calcium/metabolism , Single-Cell Gene Expression Analysis
3.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38843936

ABSTRACT

Lipid composition is conserved within sub-cellular compartments to maintain cell function. Lipidomic analyses of liver, muscle, white and brown adipose tissue (BAT) mitochondria revealed substantial differences in their glycerophospholipid (GPL) and free cholesterol (FC) contents. The GPL to FC ratio was 50-fold higher in brown than white adipose tissue mitochondria. Their purity was verified by comparison of proteomes with ER and mitochondria-associated membranes. A lipid signature containing PC and FC, calculated from the lipidomic profiles, allowed differentiation of mitochondria from BAT of mice housed at different temperatures. Elevating FC in BAT mitochondria prevented uncoupling protein (UCP) 1 function, whereas increasing GPL boosted it. Similarly, STARD3 overexpression facilitating mitochondrial FC import inhibited UCP1 function in primary brown adipocytes, whereas a knockdown promoted it. We conclude that the mitochondrial GPL/FC ratio is key for BAT function and propose that targeting it might be a promising strategy to promote UCP1 activity.


Subject(s)
Adipose Tissue, Brown , Cholesterol , Lipidomics , Mitochondria , Uncoupling Protein 1 , Animals , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Mice , Adipose Tissue, Brown/metabolism , Cholesterol/metabolism , Mitochondria/metabolism , Lipidomics/methods , Organ Specificity , Mice, Inbred C57BL , Adipose Tissue, White/metabolism , Glycerophospholipids/metabolism , Male , Lipid Metabolism
4.
Science ; 384(6700): 1111-1117, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843333

ABSTRACT

Brown adipose tissue (BAT) is a heater organ that expresses thermogenic uncoupling protein 1 (UCP1) to maintain high body temperatures during cold stress. BAT thermogenesis is considered an overarching mammalian trait, but its evolutionary origin is unknown. We show that adipose tissue of marsupials, which diverged from eutherian mammals ~150 million years ago, expresses a nonthermogenic UCP1 variant governed by a partial transcriptomic BAT signature similar to that found in eutherian beige adipose tissue. We found that the reconstructed UCP1 sequence of the common eutherian ancestor displayed typical thermogenic activity, whereas therian ancestor UCP1 is nonthermogenic. Thus, mammalian adipose tissue thermogenesis may have evolved in two distinct stages, with a prethermogenic stage in the common therian ancestor linking UCP1 expression to adipose tissue and thermal stress. We propose that in a second stage, UCP1 acquired its thermogenic function specifically in eutherians, such that the onset of mammalian BAT thermogenesis occurred only after the divergence from marsupials.


Subject(s)
Adipose Tissue, Brown , Biological Evolution , Marsupialia , Thermogenesis , Uncoupling Protein 1 , Animals , Humans , Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/metabolism , Eutheria/genetics , Eutheria/physiology , Evolution, Molecular , Marsupialia/genetics , Marsupialia/physiology , Phylogeny , Thermogenesis/genetics , Transcriptome , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
5.
Nat Metab ; 5(8): 1364-1381, 2023 08.
Article in English | MEDLINE | ID: mdl-37430025

ABSTRACT

Inflammation in the central nervous system can impair the function of neuronal mitochondria and contributes to axon degeneration in the common neuroinflammatory disease multiple sclerosis (MS). Here we combine cell-type-specific mitochondrial proteomics with in vivo biosensor imaging to dissect how inflammation alters the molecular composition and functional capacity of neuronal mitochondria. We show that neuroinflammatory lesions in the mouse spinal cord cause widespread and persisting axonal ATP deficiency, which precedes mitochondrial oxidation and calcium overload. This axonal energy deficiency is associated with impaired electron transport chain function, but also an upstream imbalance of tricarboxylic acid (TCA) cycle enzymes, with several, including key rate-limiting, enzymes being depleted in neuronal mitochondria in experimental models and in MS lesions. Notably, viral overexpression of individual TCA enzymes can ameliorate the axonal energy deficits in neuroinflammatory lesions, suggesting that TCA cycle dysfunction in MS may be amendable to therapy.


Subject(s)
Multiple Sclerosis , Neuroinflammatory Diseases , Animals , Mice , Axons/pathology , Multiple Sclerosis/pathology , Neurons/pathology , Inflammation/pathology
6.
Nat Protoc ; 18(7): 2181-2220, 2023 07.
Article in English | MEDLINE | ID: mdl-37328604

ABSTRACT

Mitochondria are key bioenergetic organelles involved in many biosynthetic and signaling pathways. However, their differential contribution to specific functions of cells within complex tissues is difficult to dissect with current methods. The present protocol addresses this need by enabling the ex vivo immunocapture of cell-type-specific mitochondria directly from their tissue context through a MitoTag reporter mouse. While other available methods were developed for bulk mitochondria isolation or more abundant cell-type-specific mitochondria, this protocol was optimized for the selective isolation of functional mitochondria from medium-to-low-abundant cell types in a heterogeneous tissue, such as the central nervous system. The protocol has three major parts: First, mitochondria of a cell type of interest are tagged via an outer mitochondrial membrane eGFP by crossing MitoTag mice to a cell-type-specific Cre-driver line or by delivery of viral vectors for Cre expression. Second, homogenates are prepared from relevant tissues by nitrogen cavitation, from which tagged organelles are immunocaptured using magnetic microbeads. Third, immunocaptured mitochondria are used for downstream assays, e.g., to probe respiratory capacity or calcium handling, revealing cell-type-specific mitochondrial diversity in molecular composition and function. The MitoTag approach enables the identification of marker proteins to label cell-type-specific organelle populations in situ, elucidates cell-type-enriched mitochondrial metabolic and signaling pathways, and reveals functional mitochondrial diversity between adjacent cell types in complex tissues, such as the brain. Apart from establishing the mouse colony (6-8 weeks without import), the immunocapture protocol takes 2 h and functional assays require 1-2 h.


Subject(s)
Brain , Mitochondria , Mice , Animals , Mitochondria/metabolism , Cell Line , Brain/metabolism , Magnetics , Energy Metabolism
7.
Front Cell Dev Biol ; 10: 965382, 2022.
Article in English | MEDLINE | ID: mdl-36393849

ABSTRACT

Aging is a complex process characterized by several molecular and cellular imbalances. The composition and stability of the neuronal cytoskeleton is essential for the maintenance of homeostasis, especially in long neurites. Using human skin biopsies containing sensory axons from a cohort of healthy individuals, we investigate alterations in cytoskeletal content and sensory axon caliber during aging via quantitative immunostainings. Cytoskeletal components show an increase with aging in both sexes, while elevation in axon diameter is only evident in males. Transcriptomic data from aging males illustrate various patterns in gene expression during aging. Together, the data suggest gender-specific changes during aging in peripheral sensory axons, possibly influencing cytoskeletal functionality and axonal caliber. These changes may cumulatively increase susceptibility of aged individuals to neurodegenerative diseases.

8.
Neuron ; 110(9): 1516-1531.e9, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35216662

ABSTRACT

PTEN-induced kinase 1 (PINK1) is a short-lived protein required for the removal of damaged mitochondria through Parkin translocation and mitophagy. Because the short half-life of PINK1 limits its ability to be trafficked into neurites, local translation is required for this mitophagy pathway to be active far from the soma. The Pink1 transcript is associated and cotransported with neuronal mitochondria. In concert with translation, the mitochondrial outer membrane proteins synaptojanin 2 binding protein (SYNJ2BP) and synaptojanin 2 (SYNJ2) are required for tethering Pink1 mRNA to mitochondria via an RNA-binding domain in SYNJ2. This neuron-specific adaptation for the local translation of PINK1 provides distal mitochondria with a continuous supply of PINK1 for the activation of mitophagy.


Subject(s)
Mitophagy , Protein Kinases , Mitochondria/metabolism , Mitophagy/genetics , Nerve Tissue Proteins , Neurons/metabolism , Phosphoric Monoester Hydrolases , Protein Kinases/genetics , RNA, Messenger/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
9.
Br J Pharmacol ; 178(22): 4518-4532, 2021 11.
Article in English | MEDLINE | ID: mdl-34287836

ABSTRACT

BACKGROUND AND PURPOSE: Treatment of cardiac arrhythmia remains challenging due to severe side effects of common anti-arrhythmic drugs. We previously demonstrated that mitochondrial Ca2+ uptake in cardiomyocytes represents a promising new candidate structure for safer drug therapy. However, druggable agonists of mitochondrial Ca2+ uptake suitable for preclinical and clinical studies are still missing. EXPERIMENTAL APPROACH: Herewe screened 727 compounds with a history of use in human clinical trials in a three-step screening approach. As a primary screening platform we used a permeabilized HeLa cell-based mitochondrial Ca2+ uptake assay. Hits were validated in cultured HL-1 cardiomyocytes and finally tested for anti-arrhythmic efficacy in three translational models: a Ca2+ overload zebrafish model and cardiomyocytes of both a mouse model for catecholaminergic polymorphic ventricular tachycardia (CPVT) and induced pluripotent stem cell derived cardiomyocytes from a CPVT patient. KEY RESULTS: We identifiedtwo candidate compounds, the clinically approved drugs ezetimibe and disulfiram, which stimulate SR-mitochondria Ca2+ transfer at nanomolar concentrations. This is significantly lower compared to the previously described mitochondrial Ca2+ uptake enhancers (MiCUps) efsevin, a gating modifier of the voltage-dependent anion channel 2, and kaempferol, an agonist of the mitochondrial Ca2+ uniporter. Both substances restored rhythmic cardiac contractions in a zebrafish cardiac arrhythmia model and significantly suppressed arrhythmogenesis in freshly isolated ventricular cardiomyocytes from a CPVT mouse model as well as induced pluripotent stem cell derived cardiomyocytes from a CPVT patient. CONCLUSION AND IMPLICATIONS: Taken together we identified ezetimibe and disulfiram as novel MiCUps and efficient suppressors of arrhythmogenesis and as such as, promising candidates for future preclinical and clinical studies.


Subject(s)
Pharmaceutical Preparations , Tachycardia, Ventricular , Animals , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , Calcium/metabolism , Calcium Signaling , Disulfiram/metabolism , Disulfiram/pharmacology , Ezetimibe/metabolism , HeLa Cells , Humans , Mice , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Pharmaceutical Preparations/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Tachycardia, Ventricular/metabolism , Zebrafish/metabolism
10.
Methods Mol Biol ; 2277: 69-89, 2021.
Article in English | MEDLINE | ID: mdl-34080145

ABSTRACT

The mitochondrial calcium uniporter (MCU ) is an essential protein of the inner mitochondrial membrane that mediates the uptake of calcium into mitochondria of virtually all mammalian tissues, regulating cell metabolism, signaling, and death. MCU-mediated calcium uptake has been shown to play a pathophysiological role in diverse human disease contexts, which qualifies this channel as a druggable target for therapeutic intervention.Here, we present a protocol to perform drug screens to identify effective and specific MCU-targeting inhibitors. The methodology is based on the use of cryopreserved mitochondria that are isolated from a yeast strain engineered to express the human MCU and its essential regulator EMRE together with the luminescence calcium sensor aequorin. Yeast mitochondria with a functionally reconstituted MCU-mediated calcium uptake are then employed as a ready-to-use screening reagent. False discovery rate is further minimized by energizing mitochondria with D-lactate in a mannitol/sucrose-based medium, which provides a mean to discriminate between direct and secondary effects of drugs on mitochondrial calcium uptake. This screening assay is sensitive and robust and can be easily implemented in any laboratory.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Drug Evaluation, Preclinical/methods , Mitochondria/drug effects , Aequorin/pharmacology , Calcium/metabolism , Calcium Channels/genetics , Drug Discovery/methods , Humans , Lactic Acid/pharmacology , Mitochondria/metabolism , Mitoxantrone/pharmacology , Saccharomyces cerevisiae/cytology
12.
Commun Biol ; 4(1): 666, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34079053

ABSTRACT

Calcium dynamics control synaptic transmission. Calcium triggers synaptic vesicle fusion, determines release probability, modulates vesicle recycling, participates in long-term plasticity and regulates cellular metabolism. Mitochondria, the main source of cellular energy, serve as calcium signaling hubs. Mitochondrial calcium transients are primarily determined by the balance between calcium influx, mediated by the mitochondrial calcium uniporter (MCU), and calcium efflux through the sodium/lithium/calcium exchanger (NCLX). We identified a human recessive missense SLC8B1 variant that impairs NCLX activity and is associated with severe mental retardation. On this basis, we examined the effect of deleting NCLX in mice on mitochondrial and synaptic calcium homeostasis, synaptic activity, and plasticity. Neuronal mitochondria exhibited basal calcium overload, membrane depolarization, and a reduction in the amplitude and rate of calcium influx and efflux. We observed smaller cytoplasmic calcium transients in the presynaptic terminals of NCLX-KO neurons, leading to a lower probability of release and weaker transmission. In agreement, synaptic facilitation in NCLX-KO hippocampal slices was enhanced. Importantly, deletion of NCLX abolished long term potentiation of Schaffer collateral synapses. Our results show that NCLX controls presynaptic calcium transients that are crucial for defining synaptic strength as well as short- and long-term plasticity, key elements of learning and memory processes.


Subject(s)
Intellectual Disability/genetics , Intellectual Disability/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Calcium Signaling , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Female , Hippocampus/metabolism , Humans , In Vitro Techniques , Long-Term Potentiation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/deficiency , Neuronal Plasticity , Neurons/metabolism , Pedigree , Point Mutation , Presynaptic Terminals/metabolism , Sodium-Calcium Exchanger/chemistry , Synaptic Transmission/genetics , Synaptic Transmission/physiology
13.
Cell Calcium ; 95: 102364, 2021 05.
Article in English | MEDLINE | ID: mdl-33601101

ABSTRACT

Human mitochondria are complex and highly dynamic biological systems, comprised of over a thousand parts and evolved to fully integrate into the specialized intracellular signaling networks and metabolic requirements of each cell and organ. Over the last two decades, several complementary, top-down computational and experimental approaches have been developed to identify, characterize and modulate the human mitochondrial system, demonstrating the power of integrating classical reductionist and discovery-driven analyses in order to de-orphanize hitherto unknown molecular components of mitochondrial machineries and pathways. To this goal, systematic, multiomics-based surveys of proteome composition, protein networks, and phenotype-to-pathway associations at the tissue, cell and organellar level have been largely exploited to predict the full complement of mitochondrial proteins and their functional interactions, therefore catalyzing data-driven hypotheses. Collectively, these multidisciplinary and integrative research approaches hold the potential to propel our understanding of mitochondrial biology and provide a systems-level framework to unraveling mitochondria-mediated and disease-spanning pathomechanisms.


Subject(s)
Mitochondria/metabolism , Proteome/metabolism , Proteomics/methods , Systems Biology/methods , Animals , Humans , Mitochondria/chemistry , Oxidative Stress/physiology , Proteome/chemistry
14.
Cell Rep ; 32(8): 108059, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32846138

ABSTRACT

The proteasome is the main proteolytic system for targeted protein degradation in the cell and is fine-tuned according to cellular needs. Here, we demonstrate that mitochondrial dysfunction and concomitant metabolic reprogramming of the tricarboxylic acid (TCA) cycle reduce the assembly and activity of the 26S proteasome. Both mitochondrial mutations in respiratory complex I and treatment with the anti-diabetic drug metformin impair 26S proteasome activity. Defective 26S assembly is reversible and can be overcome by supplementation of aspartate or pyruvate. This metabolic regulation of 26S activity involves specific regulation of proteasome assembly factors via the mTORC1 pathway. Of note, reducing 26S activity by metformin confers increased resistance toward the proteasome inhibitor bortezomib, which is reversible upon pyruvate supplementation. Our study uncovers unexpected consequences of defective mitochondrial metabolism for proteasomal protein degradation in the cell, which has important pathophysiological and therapeutic implications.


Subject(s)
Mitochondria/metabolism , Proteasome Endopeptidase Complex/metabolism , Humans
15.
Nat Commun ; 11(1): 4031, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32788582

ABSTRACT

Calcium (Ca2+) influx into mitochondria occurs through a Ca2+-selective uniporter channel, which regulates essential cellular processes in eukaryotic organisms. Previous evolutionary analyses of its pore-forming subunits MCU and EMRE, and gatekeeper MICU1, pinpointed an evolutionary paradox: the presence of MCU homologs in fungal species devoid of any other uniporter components and of mt-Ca2+ uptake. Here, we trace the mt-Ca2+ uniporter evolution across 1,156 fully-sequenced eukaryotes and show that animal and fungal MCUs represent two distinct paralogous subfamilies originating from an ancestral duplication. Accordingly, we find EMRE orthologs outside Holoza and uncover the existence of an animal-like uniporter within chytrid fungi, which enables mt-Ca2+ uptake when reconstituted in vivo in the yeast Saccharomyces cerevisiae. Our study represents the most comprehensive phylogenomic analysis of the mt-Ca2+ uptake system and demonstrates that MCU, EMRE, and MICU formed the core of the ancestral opisthokont uniporter, with major implications for comparative structural and functional studies.


Subject(s)
Calcium Channels/genetics , Evolution, Molecular , Fungal Proteins/genetics , Amino Acid Sequence , Calcium/metabolism , Calcium Channels/chemistry , Chytridiomycota/genetics , Fungal Proteins/chemistry , HeLa Cells , Humans , Likelihood Functions , Phylogeny , Species Specificity
16.
J Hepatol ; 73(6): 1347-1359, 2020 12.
Article in English | MEDLINE | ID: mdl-32598967

ABSTRACT

BACKGROUND & AIMS: Selective elimination of virus-infected hepatocytes occurs through virus-specific CD8 T cells recognizing peptide-loaded MHC molecules. Herein, we report that virus-infected hepatocytes are also selectively eliminated through a cell-autonomous mechanism. METHODS: We generated recombinant adenoviruses and genetically modified mouse models to identify the molecular mechanisms determining TNF-induced hepatocyte apoptosis in vivo and used in vivo bioluminescence imaging, immunohistochemistry, immunoblot analysis, RNAseq/proteome/phosphoproteome analyses, bioinformatic analyses, mitochondrial function tests. RESULTS: We found that TNF precisely eliminated only virus-infected hepatocytes independently of local inflammation and activation of immune sensory receptors. TNF receptor I was equally relevant for NF-kB activation in healthy and infected hepatocytes, but selectively mediated apoptosis in infected hepatocytes. Caspase 8 activation downstream of TNF receptor signaling was dispensable for apoptosis in virus-infected hepatocytes, indicating an unknown non-canonical cell-intrinsic pathway promoting apoptosis in hepatocytes. We identified a unique state of mitochondrial vulnerability in virus-infected hepatocytes as the cause for this non-canonical induction of apoptosis through TNF. Mitochondria from virus-infected hepatocytes showed normal biophysical and bioenergetic functions but were characterized by reduced resilience to calcium challenge. In the presence of unchanged TNF-induced signaling, reactive oxygen species-mediated calcium release from the endoplasmic reticulum caused mitochondrial permeability transition and apoptosis, which identified a link between extrinsic death receptor signaling and cell-intrinsic mitochondrial-mediated caspase activation. CONCLUSION: Our findings reveal a novel concept in immune surveillance by identifying a cell-autonomous defense mechanism that selectively eliminates virus-infected hepatocytes through mitochondrial permeability transition. LAY SUMMARY: The liver is known for its unique immune functions. Herein, we identify a novel mechanism by which virus-infected hepatocytes can selectively eliminate themselves through reduced mitochondrial resilience to calcium challenge.


Subject(s)
Caspase 8/metabolism , Hepatocytes , Mitochondria, Liver , Receptors, Tumor Necrosis Factor, Type I/metabolism , Animals , Apoptosis/immunology , Calcium Signaling , Cells, Cultured , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Mice , Mitochondria, Liver/immunology , Mitochondria, Liver/metabolism , Mitochondrial Transmembrane Permeability-Driven Necrosis , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
17.
Nucleic Acids Res ; 48(2): 605-632, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31799603

ABSTRACT

Mitochondria participate in metabolism and signaling. They adapt to the requirements of various cell types. Publicly available expression data permit to study expression dynamics of genes with mitochondrial function (mito-genes) in various cell types, conditions and organisms. Yet, we lack an easy way of extracting these data for mito-genes. Here, we introduce the visual data mining platform mitoXplorer, which integrates expression and mutation data of mito-genes with a manually curated mitochondrial interactome containing ∼1200 genes grouped in 38 mitochondrial processes. User-friendly analysis and visualization tools allow to mine mitochondrial expression dynamics and mutations across various datasets from four model species including human. To test the predictive power of mitoXplorer, we quantify mito-gene expression dynamics in trisomy 21 cells, as mitochondrial defects are frequent in trisomy 21. We uncover remarkable differences in the regulation of the mitochondrial transcriptome and proteome in one of the trisomy 21 cell lines, caused by dysregulation of the mitochondrial ribosome and resulting in severe defects in oxidative phosphorylation. With the newly developed Fiji plugin mitoMorph, we identify mild changes in mitochondrial morphology in trisomy 21. Taken together, mitoXplorer (http://mitoxplorer.ibdm.univ-mrs.fr) is a user-friendly, web-based and freely accessible software, aiding experimental scientists to quantify mitochondrial expression dynamics.


Subject(s)
Computational Biology , Data Mining , Mitochondria/genetics , Software , Gene Expression Regulation/genetics , Humans , Mutation/genetics , Oxidative Phosphorylation , Proteome/genetics , Transcriptome/genetics
18.
Nat Neurosci ; 22(10): 1731-1742, 2019 10.
Article in English | MEDLINE | ID: mdl-31501572

ABSTRACT

Mitochondria vary in morphology and function in different tissues; however, little is known about their molecular diversity among cell types. Here we engineered MitoTag mice, which express a Cre recombinase-dependent green fluorescent protein targeted to the outer mitochondrial membrane, and developed an isolation approach to profile tagged mitochondria from defined cell types. We determined the mitochondrial proteome of the three major cerebellar cell types (Purkinje cells, granule cells and astrocytes) and identified hundreds of mitochondrial proteins that are differentially regulated. Thus, we provide markers of cell-type-specific mitochondria for the healthy and diseased mouse and human central nervous systems, including in amyotrophic lateral sclerosis and Alzheimer's disease. Based on proteomic predictions, we demonstrate that astrocytic mitochondria metabolize long-chain fatty acids more efficiently than neuronal mitochondria. We also characterize cell-type differences in mitochondrial calcium buffering via the mitochondrial calcium uniporter (Mcu) and identify regulator of microtubule dynamics protein 3 (Rmdn3) as a determinant of endoplasmic reticulum-mitochondria proximity in Purkinje cells. Our approach enables exploring mitochondrial diversity in many in vivo contexts.


Subject(s)
Brain/cytology , Mitochondria/metabolism , Neurons/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Astrocytes/metabolism , Calcium Signaling/genetics , Calcium Signaling/physiology , Cells, Cultured , Cerebellum/cytology , Fatty Acids/metabolism , Humans , Mice , Mice, Transgenic , Mitochondrial Membranes/metabolism , Proteomics , Purkinje Cells/metabolism
19.
Methods Mol Biol ; 1925: 197-222, 2019.
Article in English | MEDLINE | ID: mdl-30674029

ABSTRACT

The development of fluorescence-based oxygen sensors coupled with microplate-based assays for quantitative bioenergetics analyses enables screening multiple experimental conditions at once with small biological material and in a timely manner. In this chapter, we outline detailed protocols and practical tips to design and perform controlled measurements of (a) respiratory and glycolytic metabolism of intact cells, (b) substrate-dependent respiration in permeabilized cells and isolated mitochondria, and (c) calcium-dependent regulation of mitochondrial bioenergetics with Seahorse XF Flux Analyzers.


Subject(s)
Biosensing Techniques/instrumentation , Calcium/metabolism , Cell Culture Techniques/instrumentation , Energy Metabolism , Mitochondria/metabolism , Oxygen/metabolism , Animals , Buffers , Cell Respiration , Equipment Design , Humans , Mice , Oxygen Consumption , Permeability
20.
Cell Rep ; 25(6): 1425-1435.e7, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30403999

ABSTRACT

The mitochondrial calcium uniporter is a highly selective ion channel composed of species- and tissue-specific subunits. However, the functional role of each component still remains unclear. Here, we establish a synthetic biology approach to dissect the interdependence between the pore-forming subunit MCU and the calcium-sensing regulator MICU1. Correlated evolutionary patterns across 247 eukaryotes indicate that their co-occurrence may have conferred a positive fitness advantage. We find that, while the heterologous reconstitution of MCU and EMRE in vivo in yeast enhances manganese stress, this is prevented by co-expression of MICU1. Accordingly, MICU1 deletion sensitizes human cells to manganese-dependent cell death by disinhibiting MCU-mediated manganese uptake. As a result, manganese overload increases oxidative stress, which can be effectively prevented by NAC treatment. Our study identifies a critical contribution of MICU1 to the uniporter selectivity, with important implications for patients with MICU1 deficiency, as well as neurological disorders arising upon chronic manganese exposure.


Subject(s)
Calcium Channels/metabolism , Calcium-Binding Proteins/metabolism , Cation Transport Proteins/metabolism , Cytoprotection , Manganese/toxicity , Mitochondrial Membrane Transport Proteins/metabolism , Apoptosis/drug effects , Cytoprotection/drug effects , Eukaryota , Evolution, Molecular , HEK293 Cells , HeLa Cells , Humans , Iron/toxicity , Mitochondria/metabolism , Phylogeny , Saccharomyces cerevisiae/metabolism , Stress, Physiological/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL