Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Med ; 29(6): 1487-1499, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37291212

ABSTRACT

Cannabis use disorder (CUD) is widespread, and there is no pharmacotherapy to facilitate its treatment. AEF0117, the first of a new pharmacological class, is a signaling-specific inhibitor of the cannabinoid receptor 1 (CB1-SSi). AEF0117 selectively inhibits a subset of intracellular effects resulting from Δ9-tetrahydrocannabinol (THC) binding without modifying behavior per se. In mice and non-human primates, AEF0117 decreased cannabinoid self-administration and THC-related behavioral impairment without producing significant adverse effects. In single-ascending-dose (0.2 mg, 0.6 mg, 2 mg and 6 mg; n = 40) and multiple-ascending-dose (0.6 mg, 2 mg and 6 mg; n = 24) phase 1 trials, healthy volunteers were randomized to ascending-dose cohorts (n = 8 per cohort; 6:2 AEF0117 to placebo randomization). In both studies, AEF0117 was safe and well tolerated (primary outcome measurements). In a double-blind, placebo-controlled, crossover phase 2a trial, volunteers with CUD were randomized to two ascending-dose cohorts (0.06 mg, n = 14; 1 mg, n = 15). AEF0117 significantly reduced cannabis' positive subjective effects (primary outcome measurement, assessed by visual analog scales) by 19% (0.06 mg) and 38% (1 mg) compared to placebo (P < 0.04). AEF0117 (1 mg) also reduced cannabis self-administration (P < 0.05). In volunteers with CUD, AEF0117 was well tolerated and did not precipitate cannabis withdrawal. These data suggest that AEF0117 is a safe and potentially efficacious treatment for CUD.ClinicalTrials.gov identifiers: NCT03325595 , NCT03443895 and NCT03717272 .


Subject(s)
Cannabis , Hallucinogens , Marijuana Abuse , Substance Withdrawal Syndrome , Animals , Mice , Double-Blind Method , Dronabinol/adverse effects , Hallucinogens/therapeutic use , Randomized Controlled Trials as Topic , Substance Withdrawal Syndrome/drug therapy
2.
Neuron ; 111(12): 1887-1897.e6, 2023 06 21.
Article in English | MEDLINE | ID: mdl-37098353

ABSTRACT

Corticosteroid-mediated stress responses require the activation of complex brain circuits involving mitochondrial activity, but the underlying cellular and molecular mechanisms are scantly known. The endocannabinoid system is implicated in stress coping, and it can directly regulate brain mitochondrial functions via type 1 cannabinoid (CB1) receptors associated with mitochondrial membranes (mtCB1). In this study, we show that the impairing effect of corticosterone in the novel object recognition (NOR) task in mice requires mtCB1 receptors and the regulation of mitochondrial calcium levels in neurons. Different brain circuits are modulated by this mechanism to mediate the impact of corticosterone during specific phases of the task. Thus, whereas corticosterone recruits mtCB1 receptors in noradrenergic neurons to impair NOR consolidation, mtCB1 receptors in local hippocampal GABAergic interneurons are required to inhibit NOR retrieval. These data reveal unforeseen mechanisms mediating the effects of corticosteroids during different phases of NOR, involving mitochondrial calcium alterations in different brain circuits.


Subject(s)
Adrenergic Neurons , Corticosterone , Mice , Animals , Corticosterone/pharmacology , Receptors, Cannabinoid , Calcium , Mitochondria , Endocannabinoids , Receptor, Cannabinoid, CB1 , Hippocampus/physiology
4.
J Neurosci Methods ; 373: 109560, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35320763

ABSTRACT

Mitochondrial calcium handling is a particularly active research area in the neuroscience field, as it plays key roles in the regulation of several functions of the central nervous system, such as synaptic transmission and plasticity, astrocyte calcium signaling, neuronal activity… In the last few decades, a panel of techniques have been developed to measure mitochondrial calcium dynamics, relying mostly on photonic microscopy, and including synthetic sensors, hybrid sensors and genetically encoded calcium sensors. The goal of this review is to endow the reader with a deep knowledge of the historical and latest tools to monitor mitochondrial calcium events in the brain, as well as a comprehensive overview of the current state of the art in brain mitochondrial calcium signaling. We will discuss the main calcium probes used in the field, their mitochondrial targeting strategies, their key properties and major drawbacks. In addition, we will detail the main roles of mitochondrial calcium handling in neuronal tissues through an extended report of the recent studies using mitochondrial targeted calcium sensors in neuronal and astroglial cells, in vitro and in vivo.


Subject(s)
Calcium Signaling , Calcium , Brain/diagnostic imaging , Brain/metabolism , Calcium/metabolism , Mitochondria/metabolism , Neurons/metabolism
5.
Cell Rep ; 37(12): 110133, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34936875

ABSTRACT

Intracellular calcium signaling underlies the astroglial control of synaptic transmission and plasticity. Mitochondria-endoplasmic reticulum contacts (MERCs) are key determinants of calcium dynamics, but their functional impact on astroglial regulation of brain information processing is unexplored. We found that the activation of astrocyte mitochondrial-associated type-1 cannabinoid (mtCB1) receptors determines MERC-dependent intracellular calcium signaling and synaptic integration. The stimulation of mtCB1 receptors promotes calcium transfer from the endoplasmic reticulum to mitochondria through a specific molecular cascade, involving the mitochondrial calcium uniporter (MCU). Physiologically, mtCB1-dependent mitochondrial calcium uptake determines the dynamics of cytosolic calcium events in astrocytes upon endocannabinoid mobilization. Accordingly, electrophysiological recordings in hippocampal slices showed that conditional genetic exclusion of mtCB1 receptors or dominant-negative MCU expression in astrocytes blocks lateral synaptic potentiation, through which astrocytes integrate the activity of distant synapses. Altogether, these data reveal an endocannabinoid link between astroglial MERCs and the regulation of brain network functions.


Subject(s)
Astrocytes/metabolism , Calcium/metabolism , Cannabinoids/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Receptors, Cannabinoid/physiology , Synapses/physiology , Animals , Astrocytes/cytology , Calcium Channels/physiology , Calcium Signaling , Cells, Cultured , Hippocampus/metabolism , Homeostasis , Mice , Mice, Inbred C57BL , Neuronal Plasticity , Synaptic Transmission
6.
Neuropharmacology ; 195: 108678, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34157362

ABSTRACT

The endocannabinoid system (ECS) is involved in a variety of brain functions, mainly through the activation of the type-1 cannabinoid receptors (CB1R). CB1R are highly expressed throughout the brain at different structural, cellular and subcellular locations and its activity and expression levels have a direct impact in synaptic activity and behavior. In the last few decades, astrocytes have arisen as active players of brain physiology through their participation in the tripartite synapse and through their metabolic interaction with neurons. Here, we discuss some of the mechanisms by which astroglial CB1R at different subcellular locations, regulate astrocyte calcium signals and have an impact on gliotransmission and metabolic regulation. In addition, we discuss evidence pointing at astrocytes as potential important sources of endocannabinoid synthesis and release. Thus, we summarize recent findings that add further complexity and establish that the ECS is a fundamental effector of astrocyte functions in the brain. This article is part of the special issue on 'Cannabinoids'.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Endocannabinoids/metabolism , Neurons/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Synapses/metabolism , Synaptic Transmission/physiology
7.
Cancers (Basel) ; 13(5)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807672

ABSTRACT

ARMCX3 is encoded by a member of the Armcx gene family and is known to be involved in nervous system development and function. We found that ARMCX3 is markedly upregulated in mouse liver in response to high lipid availability, and that hepatic ARMCX3 is upregulated in patients with NAFLD and hepatocellular carcinoma (HCC). Mice were subjected to ARMCX3 invalidation (inducible ARMCX3 knockout) and then exposed to a high-fat diet and diethylnitrosamine-induced hepatocarcinogenesis. The effects of experimental ARMCX3 knockdown or overexpression in HCC cell lines were also analyzed. ARMCX3 invalidation protected mice against high-fat-diet-induced NAFLD and chemically induced hepatocarcinogenesis. ARMCX3 invalidation promoted apoptotic cell death and macrophage infiltration in livers of diethylnitrosamine-treated mice maintained on a high-fat diet. ARMCX3 downregulation reduced the viability, clonality and migration of HCC cell lines, whereas ARMCX3 overexpression caused the reciprocal effects. SOX9 was found to mediate the effects of ARMCX3 in hepatic cells, with the SOX9 interaction required for the effects of ARMCX3 on hepatic cell proliferation. In conclusion, ARMCX3 is identified as a novel molecular actor in liver physiopathology and carcinogenesis. ARMCX3 downregulation appears to protect against hepatocarcinogenesis, especially under conditions of high dietary lipid-mediated hepatic insult.

8.
Nature ; 583(7817): 603-608, 2020 07.
Article in English | MEDLINE | ID: mdl-32641832

ABSTRACT

Astrocytes take up glucose from the bloodstream to provide energy to the brain, thereby allowing neuronal activity and behavioural responses1-5. By contrast, astrocytes are under neuronal control through specific neurotransmitter receptors5-7. However, whether the activation of astroglial receptors can directly regulate cellular glucose metabolism to eventually modulate behavioural responses is unclear. Here we show that activation of mouse astroglial type-1 cannabinoid receptors associated with mitochondrial membranes (mtCB1) hampers the metabolism of glucose and the production of lactate in the brain, resulting in altered neuronal functions and, in turn, impaired behavioural responses in social interaction assays. Specifically, activation of astroglial mtCB1 receptors reduces the phosphorylation of the mitochondrial complex I subunit NDUFS4, which decreases the stability and activity of complex I. This leads to a reduction in the generation of reactive oxygen species by astrocytes and affects the glycolytic production of lactate through the hypoxia-inducible factor 1 pathway, eventually resulting in neuronal redox stress and impairment of behavioural responses in social interaction assays. Genetic and pharmacological correction of each of these effects abolishes the effect of cannabinoid treatment on the observed behaviour. These findings suggest that mtCB1 receptor signalling can directly regulate astroglial glucose metabolism to fine-tune neuronal activity and behaviour in mice.


Subject(s)
Astrocytes/metabolism , Energy Metabolism , Glucose/metabolism , Mitochondria/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cannabinoid Receptor Agonists/pharmacology , Cells, Cultured , Dronabinol/pharmacology , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Energy Metabolism/drug effects , Glycolysis/drug effects , Humans , Hypoxia-Inducible Factor 1/metabolism , Lactic Acid/metabolism , Male , Mice , Mitochondria/drug effects , Mitochondrial Membranes/metabolism , Oxidation-Reduction , Phosphorylation , Reactive Oxygen Species/metabolism , Receptor, Cannabinoid, CB1/agonists , Social Behavior
9.
Methods Enzymol ; 593: 143-174, 2017.
Article in English | MEDLINE | ID: mdl-28750801

ABSTRACT

Recent evidence indicates that, besides its canonical localization at cell plasma membranes, the type-1 cannabinoid receptor, CB1 is functionally present at brain and muscle mitochondrial membranes (mtCB1). Through mtCB1 receptors, cannabinoids can directly regulate intramitochondrial signaling and respiration. This new and surprising discovery paves the way to new potential fields of research, dealing with the direct impact of G protein-coupled receptors on bioenergetic processes and its functional implications. In this chapter, we summarize some key experimental approaches established in our laboratories to identify anatomical, biochemical, and functional features of mtCB1 receptors in the brain. In particular, we describe the procedures to obtain reliable and controlled detection of mtCB1 receptors by immunogold electromicroscopy and by immunoblotting methods. Then, we address the study of direct cannabinoid effects on the electron transport system and oxidative phosphorylation. Finally, we present a functional example of the impact of mtCB1 receptors on mitochondrial mobility in cultured neurons. Considering the youth of the field, these methodological approaches will very likely be improved and refined in the future, but this chapter aims at presenting the methods that are currently used and, in particular, at underlining the need of rigorous controls to obtain reliable results. We hope that this chapter might help scientists becoming interested in this new and exciting field of research.


Subject(s)
Brain/metabolism , Mitochondria/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Blotting, Western , Brain/ultrastructure , Cell Respiration , Electron Transport Complex I/metabolism , Immunohistochemistry , Immunoprecipitation , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Oxygen Consumption , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/isolation & purification
10.
Hum Mol Genet ; 26(4): 674-685, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28007911

ABSTRACT

Hereditary spastic paraplegia, SPG31, is a rare neurological disorder caused by mutations in REEP1 gene encoding the microtubule-interacting protein, REEP1. The mechanism by which REEP1-dependent processes are linked with the disease is unclear. REEP1 regulates the morphology and trafficking of various organelles via interaction with the microtubules. In this study, we collected primary fibroblasts from SPG31 patients to investigate their mitochondrial morphology. We observed that the mitochondrial morphology in patient cells was highly tubular compared with control cells. We provide evidence that these morphological alterations are caused by the inhibition of mitochondrial fission protein, DRP1, due to the hyperphosphorylation of its serine 637 residue. This hyperphosphorylation is caused by impaired interactions between REEP1 and mitochondrial phosphatase PGAM5. Genetically or pharmacologically induced decrease of DRP1-S637 phosphorylation restores mitochondrial morphology in patient cells. Furthermore, ectopic expression of REEP1 carrying pathological mutations in primary neuronal culture targets REEP1 to the mitochondria. Mutated REEP1 proteins sequester mitochondria to the perinuclear region of the neurons and therefore, hamper mitochondrial transport along the axon. Considering the established role of mitochondrial distribution and morphology in neuronal health, our results support the involvement of a mitochondrial dysfunction in SPG31 pathology.


Subject(s)
Cell Nucleus , GTP Phosphohydrolases , Microtubule-Associated Proteins , Mitochondria , Mitochondrial Proteins , Neurons/metabolism , Spastic Paraplegia, Hereditary , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cells, Cultured , Dynamins , Female , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neurons/pathology , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Phosphorylation/genetics , Spastic Paraplegia, Hereditary/metabolism , Spastic Paraplegia, Hereditary/pathology
11.
Nature ; 539(7630): 555-559, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27828947

ABSTRACT

Cellular activity in the brain depends on the high energetic support provided by mitochondria, the cell organelles which use energy sources to generate ATP. Acute cannabinoid intoxication induces amnesia in humans and animals, and the activation of type-1 cannabinoid receptors present at brain mitochondria membranes (mtCB1) can directly alter mitochondrial energetic activity. Although the pathological impact of chronic mitochondrial dysfunctions in the brain is well established, the involvement of acute modulation of mitochondrial activity in high brain functions, including learning and memory, is unknown. Here, we show that acute cannabinoid-induced memory impairment in mice requires activation of hippocampal mtCB1 receptors. Genetic exclusion of CB1 receptors from hippocampal mitochondria prevents cannabinoid-induced reduction of mitochondrial mobility, synaptic transmission and memory formation. mtCB1 receptors signal through intra-mitochondrial Gαi protein activation and consequent inhibition of soluble-adenylyl cyclase (sAC). The resulting inhibition of protein kinase A (PKA)-dependent phosphorylation of specific subunits of the mitochondrial electron transport system eventually leads to decreased cellular respiration. Hippocampal inhibition of sAC activity or manipulation of intra-mitochondrial PKA signalling or phosphorylation of the Complex I subunit NDUFS2 inhibit bioenergetic and amnesic effects of cannabinoids. Thus, the G protein-coupled mtCB1 receptors regulate memory processes via modulation of mitochondrial energy metabolism. By directly linking mitochondrial activity to memory formation, these data reveal that bioenergetic processes are primary acute regulators of cognitive functions.


Subject(s)
Cannabinoids/adverse effects , Memory Disorders/chemically induced , Memory/drug effects , Memory/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Adenylyl Cyclases/metabolism , Animals , Cannabinoids/metabolism , Cell Respiration/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Electron Transport/drug effects , Energy Metabolism/drug effects , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Memory Disorders/enzymology , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/enzymology , Mitochondrial Membranes/metabolism , NADH Dehydrogenase/metabolism , Oxidative Phosphorylation/drug effects , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/drug effects , Synaptic Transmission/drug effects
12.
Toxicol Sci ; 139(2): 407-20, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24595818

ABSTRACT

3,4-Methylenedioxymethamphetamine (MDMA; "ecstasy") is a potentially neurotoxic recreational drug of abuse. Though the mechanisms involved are still not completely understood, formation of reactive metabolites and mitochondrial dysfunction contribute to MDMA-related neurotoxicity. Neuronal mitochondrial trafficking, and their targeting to synapses, is essential for proper neuronal function and survival, rendering neurons particularly vulnerable to mitochondrial dysfunction. Indeed, MDMA-associated disruption of Ca(2+) homeostasis and ATP depletion have been described in neurons, thus suggesting possible MDMA interference on mitochondrial dynamics. In this study, we performed real-time functional experiments of mitochondrial trafficking to explore the role of in situ mitochondrial dysfunction in MDMA's neurotoxic actions. We show that the mixture of MDMA and six of its major in vivo metabolites, each compound at 10µM, impaired mitochondrial trafficking and increased the fragmentation of axonal mitochondria in cultured hippocampal neurons. Furthermore, the overexpression of mitofusin 2 (Mfn2) or dynamin-related protein 1 (Drp1) K38A constructs almost completely rescued the trafficking deficits caused by this mixture. Finally, in hippocampal neurons overexpressing a Mfn2 mutant, Mfn2 R94Q, with impaired fusion and transport properties, it was confirmed that a dysregulation of mitochondrial fission/fusion events greatly contributed to the reported trafficking phenotype. In conclusion, our study demonstrated, for the first time, that the mixture of MDMA and its metabolites, at concentrations relevant to the in vivo scenario, impaired mitochondrial trafficking and increased mitochondrial fragmentation in hippocampal neurons, thus providing a new insight in the context of "ecstasy"-induced neuronal injury.


Subject(s)
Axonal Transport/drug effects , Hippocampus/drug effects , Mitochondrial Dynamics/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/metabolism , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Neurons/drug effects , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , GTP Phosphohydrolases/metabolism , Hippocampus/metabolism , Mice , Neurons/metabolism , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Rats
13.
Arch Toxicol ; 88(8): 1561-72, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24522274

ABSTRACT

Identification of the mechanisms by which drugs of abuse cause neuronal dysfunction is essential for understanding the biological bases of their acute and long-lasting effects in the brain. Here, we performed real-time functional experiments of axonal transport of mitochondria to explore the role of in situ mitochondrial dysfunction in 3,4-methylenedioxymethamphetamine (MDMA; "ecstasy")-related brain actions. We showed that MDMA dramatically reduced mitochondrial trafficking in hippocampal neurons in a Tau-dependent manner, in which glycogen synthase kinase 3ß activity was implicated. Furthermore, we found that these trafficking abnormalities were rescued by over-expression of Mitofusin2 and dynamin-related protein 1, but not of Miro1. Given the relevance of mitochondrial targeting for neuronal function and neurotransmission, our data underscore a novel mechanism of action of MDMA that may contribute to our understanding of how this drug of abuse alters neuronal functioning.


Subject(s)
Dynamins/metabolism , GTP Phosphohydrolases/metabolism , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Neurons/drug effects , tau Proteins/metabolism , Animals , Axonal Transport/drug effects , Calcium/metabolism , Cells, Cultured , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/embryology , Mice, Inbred C57BL , Mitochondria/metabolism , Neurons/metabolism , Phosphorylation
14.
PLoS One ; 8(7): e67773, 2013.
Article in English | MEDLINE | ID: mdl-23844091

ABSTRACT

The regulation of mitochondrial dynamics is vital in complex cell types, such as neurons, that transport and localize mitochondria in high energy-demanding cell domains. The Armcx3 gene encodes a mitochondrial-targeted protein (Alex3) that contains several arm-like domains. In a previous study we showed that Alex3 protein regulates mitochondrial aggregation and trafficking. Here we studied the contribution of Wnt proteins to the mitochondrial aggregation and dynamics regulated by Alex3. Overexpression of Alex3 in HEK293 cells caused a marked aggregation of mitochondria, which was attenuated by treatment with several Wnts. We also found that this decrease was caused by Alex3 degradation induced by Wnts. While the Wnt canonical pathway did not alter the pattern of mitochondrial aggregation induced by Alex3, we observed that the Wnt/PKC non-canonical pathway regulated both mitochondrial aggregation and Alex3 protein levels, thereby rendering a mitochondrial phenotype and distribution similar to control patterns. Our data suggest that the Wnt pathway regulates mitochondrial distribution and dynamics through Alex3 protein degradation.


Subject(s)
Armadillo Domain Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/metabolism , Protein Kinase C/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Amino Acid Motifs , Armadillo Domain Proteins/genetics , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Mitochondria/genetics , Mitochondrial Proteins/genetics , Molecular Sequence Data , Naphthalenes/pharmacology , Protein Kinase C/genetics , Protein Kinase Inhibitors/pharmacology , Protein Stability , Protein Structure, Tertiary , Proteolysis , Wnt Proteins/genetics
15.
Nat Commun ; 3: 814, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22569362

ABSTRACT

Brain function requires neuronal activity-dependent energy consumption. Neuronal energy supply is controlled by molecular mechanisms that regulate mitochondrial dynamics, including Kinesin motors and Mitofusins, Miro1-2 and Trak2 proteins. Here we show a new protein family that localizes to the mitochondria and controls mitochondrial dynamics. This family of proteins is encoded by an array of armadillo (Arm) repeat-containing genes located on the X chromosome. The Armcx cluster is unique to Eutherian mammals and evolved from a single ancestor gene (Armc10). We show that these genes are highly expressed in the developing and adult nervous system. Furthermore, we demonstrate that Armcx3 expression levels regulate mitochondrial dynamics and trafficking in neurons, and that Alex3 interacts with the Kinesin/Miro/Trak2 complex in a Ca(2+)-dependent manner. Our data provide evidence of a new Eutherian-specific family of mitochondrial proteins that controls mitochondrial dynamics and indicate that this key process is differentially regulated in the brain of higher vertebrates.


Subject(s)
Armadillo Domain Proteins/metabolism , Carrier Proteins/metabolism , Evolution, Molecular , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Armadillo Domain Proteins/genetics , Carrier Proteins/genetics , Cell Line , Humans , Mitochondria/genetics , Mitochondrial Proteins/genetics , Multigene Family , Nerve Tissue Proteins/genetics , Protein Binding , Protein Transport , rho GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...