Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Neurosci ; 41(31): 6596-6616, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34261699

ABSTRACT

Eukaryotic cells maintain proteostasis through mechanisms that require cytoplasmic and mitochondrial translation. Genetic defects affecting cytoplasmic translation perturb synapse development, neurotransmission, and are causative of neurodevelopmental disorders, such as Fragile X syndrome. In contrast, there is little indication that mitochondrial proteostasis, either in the form of mitochondrial protein translation and/or degradation, is required for synapse development and function. Here we focus on two genes deleted in a recurrent copy number variation causing neurodevelopmental disorders, the 22q11.2 microdeletion syndrome. We demonstrate that SLC25A1 and MRPL40, two genes present in the microdeleted segment and whose products localize to mitochondria, interact and are necessary for mitochondrial ribosomal integrity and proteostasis. Our Drosophila studies show that mitochondrial ribosome function is necessary for synapse neurodevelopment, function, and behavior. We propose that mitochondrial proteostasis perturbations, either by genetic or environmental factors, are a pathogenic mechanism for neurodevelopmental disorders.SIGNIFICANCE STATEMENT The balance between cytoplasmic protein synthesis and degradation, or cytoplasmic proteostasis, is required for normal synapse function and neurodevelopment. Cytoplasmic and mitochondrial ribosomes are necessary for two compartmentalized, yet interdependent, forms of proteostasis. Proteostasis dependent on cytoplasmic ribosomes is a well-established target of genetic defects that cause neurodevelopmental disorders, such as autism. Here we show that the mitochondrial ribosome is a neurodevelopmentally regulated organelle whose function is required for synapse development and function. We propose that defective mitochondrial proteostasis is a mechanism with the potential to contribute to neurodevelopmental disease.


Subject(s)
Developmental Disabilities , Mitochondria/physiology , Mitochondrial Proteins/genetics , Organic Anion Transporters/genetics , Proteostasis/genetics , Ribonucleoproteins/genetics , Ribosomal Proteins/genetics , Animals , Cell Line , Developmental Disabilities/genetics , Developmental Disabilities/metabolism , Developmental Disabilities/physiopathology , Drosophila , Gene Expression Regulation/genetics , Humans , Neurogenesis/physiology , Protein Biosynthesis/genetics , Rats , Rats, Sprague-Dawley , Ribosomes/physiology
2.
J Neurosci ; 41(2): 215-233, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33208468

ABSTRACT

Rare genetic diseases preponderantly affect the nervous system causing neurodegeneration to neurodevelopmental disorders. This is the case for both Menkes and Wilson disease, arising from mutations in ATP7A and ATP7B, respectively. The ATP7A and ATP7B proteins localize to the Golgi and regulate copper homeostasis. We demonstrate genetic and biochemical interactions between ATP7 paralogs with the conserved oligomeric Golgi (COG) complex, a Golgi apparatus vesicular tether. Disruption of Drosophila copper homeostasis by ATP7 tissue-specific transgenic expression caused alterations in epidermis, aminergic, sensory, and motor neurons. Prominent among neuronal phenotypes was a decreased mitochondrial content at synapses, a phenotype that paralleled with alterations of synaptic morphology, transmission, and plasticity. These neuronal and synaptic phenotypes caused by transgenic expression of ATP7 were rescued by downregulation of COG complex subunits. We conclude that the integrity of Golgi-dependent copper homeostasis mechanisms, requiring ATP7 and COG, are necessary to maintain mitochondria functional integrity and localization to synapses.SIGNIFICANCE STATEMENT Menkes and Wilson disease affect copper homeostasis and characteristically afflict the nervous system. However, their molecular neuropathology mechanisms remain mostly unexplored. We demonstrate that copper homeostasis in neurons is maintained by two factors that localize to the Golgi apparatus, ATP7 and the conserved oligomeric Golgi (COG) complex. Disruption of these mechanisms affect mitochondrial function and localization to synapses as well as neurotransmission and synaptic plasticity. These findings suggest communication between the Golgi apparatus and mitochondria through homeostatically controlled cellular copper levels and copper-dependent enzymatic activities in both organelles.


Subject(s)
Copper/physiology , Golgi Apparatus/physiology , Homeostasis/physiology , Organelle Biogenesis , Synapses/physiology , Adenosine Triphosphatases/metabolism , Animals , Animals, Genetically Modified , Cell Line , Copper/toxicity , Copper-Transporting ATPases/genetics , Drosophila , Electric Stimulation , Extracellular Space/metabolism , Female , Humans , Male , RNA, Small Interfering , Synapses/ultrastructure
3.
Am J Physiol Regul Integr Comp Physiol ; 319(6): R611-R616, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32966119

ABSTRACT

Posttraumatic stress disorder (PTSD) is an independent risk factor for the development of hypertension and cardiovascular disease. Patients with PTSD have heightened blood pressure and sympathetic nervous system reactivity; however, it is unclear if patients with PTSD have exaggerated vasoconstriction in response to sympathetic nerve activation that could also contribute to increased blood pressure reactivity. Therefore, we hypothesized that patients with PTSD have increased sensitivity of vascular α1-adrenergic receptors (α1ARs), the major mediators of vasoconstriction in response to release of norepinephrine at sympathetic nerve terminals. To assess vascular α1AR sensitivity, we measured the degree of venoconstriction in a dorsal hand vein in response to exponentially increasing doses of the selective α1AR agonist, phenylephrine (PE), in 9 patients with PTSD (age = 59 ± 2 yr) and 10 age-matched controls (age = 60 ± 1 yr). Individual dose-response curves were generated to determine the dose of PE that induces 50% of maximal venoconstriction (i.e., PE ED50) reflective of vascular α1AR sensitivity. In support of our hypothesis, PE ED50 values were lower in PTSD compared with controls (245 ± 54 ng/min vs. 1,995 ± 459 ng/min, P = 0.012), indicating increased vascular α1AR sensitivity in PTSD. The PTSD group also had an increase in slope of rise in venoconstriction, indicative of an altered venoconstrictive reactivity to PE compared with controls (19.8% ± 1.2% vs. 15.1% ± 1.2%, P = 0.009). Heightened vascular α1AR sensitivity in PTSD may contribute to augmented vasoconstriction and blood pressure reactivity to sympathoexcitation and to increased cardiovascular disease risk in this patient population.


Subject(s)
Aging/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Stress Disorders, Post-Traumatic/metabolism , Sympathetic Nervous System/metabolism , Vasoconstriction , Adrenergic alpha-1 Receptor Agonists/administration & dosage , Age Factors , Blood Pressure , Case-Control Studies , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Phenylephrine/administration & dosage , Receptors, Adrenergic, alpha-1/drug effects , Signal Transduction , Stress Disorders, Post-Traumatic/physiopathology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiopathology , Vasoconstriction/drug effects
4.
Curr Opin Cell Biol ; 59: 24-33, 2019 08.
Article in English | MEDLINE | ID: mdl-30928671

ABSTRACT

Copper is an essential micronutrient required for oxygen-dependent enzymes, yet excess of the metal is a toxicant. The tug-of-war between these copper activities is balanced by chaperones and membrane transporters, which control copper distribution and availability. The P-type ATPase transporters, ATP7A and ATP7B, regulate cytoplasmic copper by pumping copper out of cells or into the endomembrane system. Mutations in ATP7A and ATP7B cause diseases that share neuropsychiatric phenotypes, which are similar to phenotypes observed in mutations affecting cytoplasmic trafficking complexes required for ATP7A/B dynamics. Here, we discuss evidence indicating that phenotypes associated to genetic defects in trafficking complexes, such as retromer and the adaptor complex AP-1, result in part from copper dyshomeostasis due to mislocalized ATP7A and ATP7B.


Subject(s)
Copper Transport Proteins/metabolism , Copper-Transporting ATPases/metabolism , Animals , Humans , Mitochondria/metabolism , Mutation/genetics , Parkinson Disease/genetics , Protein Transport
5.
J Neurosci ; 39(18): 3561-3581, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30833507

ABSTRACT

Neurodevelopmental disorders offer insight into synaptic mechanisms. To unbiasedly uncover these mechanisms, we studied the 22q11.2 syndrome, a recurrent copy number variant, which is the highest schizophrenia genetic risk factor. We quantified the proteomes of 22q11.2 mutant human fibroblasts from both sexes and mouse brains carrying a 22q11.2-like defect, Df(16)A+/- Molecular ontologies defined mitochondrial compartments and pathways as some of top ranked categories. In particular, we identified perturbations in the SLC25A1-SLC25A4 mitochondrial transporter interactome as associated with the 22q11.2 genetic defect. Expression of SLC25A1-SLC25A4 interactome components was affected in neuronal cells from schizophrenia patients. Furthermore, hemideficiency of the Drosophila SLC25A1 or SLC25A4 orthologues, dSLC25A1-sea and dSLC25A4-sesB, affected synapse morphology, neurotransmission, plasticity, and sleep patterns. Our findings indicate that synapses are sensitive to partial loss of function of mitochondrial solute transporters. We propose that mitoproteomes regulate synapse development and function in normal and pathological conditions in a cell-specific manner.SIGNIFICANCE STATEMENT We address the central question of how to comprehensively define molecular mechanisms of the most prevalent and penetrant microdeletion associated with neurodevelopmental disorders, the 22q11.2 microdeletion syndrome. This complex mutation reduces gene dosage of ∼63 genes in humans. We describe a disruption of the mitoproteome in 22q11.2 patients and brains of a 22q11.2 mouse model. In particular, we identify a network of inner mitochondrial membrane transporters as a hub required for synapse function. Our findings suggest that mitochondrial composition and function modulate the risk of neurodevelopmental disorders, such as schizophrenia.


Subject(s)
22q11 Deletion Syndrome/metabolism , Brain/metabolism , Mitochondria/metabolism , Neurons/metabolism , Synapses/metabolism , Adenine Nucleotide Translocator 1/metabolism , Animals , Behavior, Animal , Cell Line , Chromosome Deletion , Chromosomes, Human, Pair 22/metabolism , Drosophila , Female , Fibroblasts/metabolism , Humans , Male , Mitochondrial Proteins/metabolism , Organic Anion Transporters/metabolism , Proteome , Schizophrenia/metabolism
6.
Cell Syst ; 6(3): 368-380.e6, 2018 Mar 28.
Article in English | MEDLINE | ID: mdl-29397366

ABSTRACT

Rare neurological diseases shed light onto universal neurobiological processes. However, molecular mechanisms connecting genetic defects to their disease phenotypes are elusive. Here, we obtain mechanistic information by comparing proteomes of cells from individuals with rare disorders with proteomes from their disease-free consanguineous relatives. We use triple-SILAC mass spectrometry to quantify proteomes from human pedigrees affected by mutations in ATP7A, which cause Menkes disease, a rare neurodegenerative and neurodevelopmental disorder stemming from systemic copper depletion. We identified 214 proteins whose expression was altered in ATP7A-/y fibroblasts. Bioinformatic analysis of ATP7A-mutant proteomes identified known phenotypes and processes affected in rare genetic diseases causing copper dyshomeostasis, including altered mitochondrial function. We found connections between copper dyshomeostasis and the UCHL1/PARK5 pathway of Parkinson disease, which we validated with mitochondrial respiration and Drosophila genetics assays. We propose that our genealogical "omics" strategy can be broadly applied to identify mechanisms linking a genomic locus to its phenotypes.


Subject(s)
Copper/metabolism , Ubiquitin Thiolesterase/genetics , Adenosine Triphosphatases/genetics , Animals , Cation Transport Proteins/genetics , Computational Biology/methods , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Disease Models, Animal , Drosophila , Female , Fibroblasts/metabolism , Homeostasis/genetics , Humans , Male , Menkes Kinky Hair Syndrome/genetics , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Mutation , Pedigree , Phenotype , Proteomics/methods , Rare Diseases/metabolism , Ubiquitin Thiolesterase/metabolism
7.
Dev Neurobiol ; 78(3): 311-330, 2018 03.
Article in English | MEDLINE | ID: mdl-28986965

ABSTRACT

The biogenesis of lysosome-related organelles complex-1 (BLOC-1) and the bloc-one-related complex (BORC) are the cytosolic protein complexes required for specialized membrane protein traffic along the endocytic route and the spatial distribution of endosome-derived compartments, respectively. BLOC-1 and BORC complex subunits and components of their interactomes have been associated with the risk and/or pathomechanisms of neurodevelopmental disorders. Thus, cellular processes requiring BLOC-1 and BORC interactomes have the potential to offer novel insight into mechanisms underlying behavioral defects. We focus on interactions between BLOC-1 or BORC subunits with the actin and microtubule cytoskeleton, membrane tethers, and SNAREs. These interactions highlight requirements for BLOC-1 and BORC in membrane movement by motors, control of actin polymerization, and targeting of membrane proteins to specialized cellular domains such as the nerve terminal and the primary cilium. We propose that the endosome-primary cilia pathway is an underappreciated hub in the genesis and mechanisms of neurodevelopmental disorders. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 311-330, 2018.


Subject(s)
Cilia/metabolism , Endosomes/metabolism , Nerve Tissue Proteins/metabolism , Neurodevelopmental Disorders/metabolism , Animals , Humans
8.
Elife ; 62017 03 29.
Article in English | MEDLINE | ID: mdl-28355134

ABSTRACT

Genetic and environmental factors, such as metals, interact to determine neurological traits. We reasoned that interactomes of molecules handling metals in neurons should include novel metal homeostasis pathways. We focused on copper and its transporter ATP7A because ATP7A null mutations cause neurodegeneration. We performed ATP7A immunoaffinity chromatography and identified 541 proteins co-isolating with ATP7A. The ATP7A interactome concentrated gene products implicated in neurodegeneration and neurodevelopmental disorders, including subunits of the Golgi-localized conserved oligomeric Golgi (COG) complex. COG null cells possess altered content and subcellular localization of ATP7A and CTR1 (SLC31A1), the transporter required for copper uptake, as well as decreased total cellular copper, and impaired copper-dependent metabolic responses. Changes in the expression of ATP7A and COG subunits in Drosophila neurons altered synapse development in larvae and copper-induced mortality of adult flies. We conclude that the ATP7A interactome encompasses a novel COG-dependent mechanism to specify neuronal development and survival.


Subject(s)
Copper-Transporting ATPases/metabolism , Copper/metabolism , Neurons/physiology , Protein Interaction Maps , Animals , Cell Line , Cell Survival , Drosophila , Humans
9.
J Neurosci ; 36(49): 12393-12411, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27927957

ABSTRACT

Proteome modifications downstream of monogenic or polygenic disorders have the potential to uncover novel molecular mechanisms participating in pathogenesis and/or extragenic modification of phenotypic expression. We tested this idea by determining the proteome sensitive to genetic defects in a locus encoding dysbindin, a protein required for synapse biology and implicated in schizophrenia risk. We applied quantitative mass spectrometry to identify proteins expressed in neuronal cells the abundance of which was altered after downregulation of the schizophrenia susceptibility factor dysbindin (Bloc1s8) or two other dysbindin-interacting polypeptides, which assemble into the octameric biogenesis of lysosome-related organelles complex 1 (BLOC-1). We found 491 proteins sensitive to dysbindin and BLOC-1 loss of function. Gene ontology of these 491 proteins singled out the actin cytoskeleton and the actin polymerization factor, the Arp2/3 complex, as top statistical molecular pathways contained within the BLOC-1-sensitive proteome. Subunits of the Arp2/3 complex were downregulated by BLOC-1 loss of function, thus affecting actin dynamics in early endosomes of BLOC-1-deficient cells. Furthermore, we demonstrated that Arp2/3, dysbindin, and subunits of the BLOC-1 complex biochemically and genetically interact, modulating Drosophila melanogaster synapse morphology and homeostatic synaptic plasticity. Our results indicate that ontologically prioritized proteomics identifies novel pathways that modify synaptic phenotypes associated with neurodevelopmental disorder gene defects. SIGNIFICANCE STATEMENT: The mechanisms associated with schizophrenia are mostly unknown despite the increasing number of genetic loci identified that increase disease risk. We present an experimental strategy that impartially and comprehensively interrogates the proteome of neurons to identify effects of genetic mutations in a schizophrenia risk factor, dysbindin. We find that the expression of the actin polymerization complex Arp2/3 is reduced in dysbindin-deficient cells, thus affecting actin-dependent phenotypes in two cellular compartments where dysbindin resides, endosomes and presynapses. Our studies indicate that a central cellular structure affected by schizophrenia susceptibility loci is the actin cytoskeleton, an organelle necessary for synaptic function in the presynaptic and postsynaptic compartment.


Subject(s)
Actin-Related Protein 3/genetics , Angiopoietins/genetics , Carrier Proteins/genetics , Dystrophin-Associated Proteins/genetics , Lectins/genetics , Schizophrenia/genetics , Synapses , Actins/genetics , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Animals , Cells, Cultured , Cytoskeleton/genetics , Drosophila melanogaster , Dysbindin , Female , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Polymerization , Proteome
10.
Dev Neurobiol ; 68(10): 1225-42, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18548486

ABSTRACT

During learning and memory formation, information flow through networks is regulated significantly through structural alterations in neurons. Dendrites, sites of signal integration, are key targets of activity-mediated modifications. Although local mechanisms of dendritic growth ensure synapse-specific changes, global mechanisms linking neural activity to nuclear gene expression may have profound influences on neural function. Fos, being an immediate-early gene, is ideally suited to be an initial transducer of neural activity, but a precise role for the AP-1 transcription factor in dendrite growth remains to be elucidated. Here we measure changes in the dendritic fields of identified Drosophila motor neurons in vivo and in primary culture to investigate the role of the immediate-early transcription factor AP-1 in regulating endogenous and activity-induced dendrite growth. Our data indicate that (a) increased neural excitability or depolarization stimulates dendrite growth, (b) AP-1 (a Fos, Jun hetero-dimer) is required for normal motor neuron dendritic growth during development and in response to activity induction, and (c) neuronal Fos protein levels are rapidly but transiently induced in motor neurons following neural activity. Taken together, these results show that AP-1 mediated transcription is important for dendrite growth, and that neural activity influences global dendritic growth through a gene-expression dependent mechanism gated by AP-1.


Subject(s)
Cell Differentiation/genetics , Central Nervous System/growth & development , Dendrites/metabolism , Drosophila melanogaster/growth & development , Motor Neurons/physiology , Transcription Factor AP-1/metabolism , Animals , Cells, Cultured , Central Nervous System/cytology , Dendrites/ultrastructure , Drosophila melanogaster/cytology , Gene Expression Regulation, Developmental/genetics , Genes, Immediate-Early/genetics , Motor Neurons/cytology , Motor Neurons/metabolism , Neuronal Plasticity/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Synaptic Transmission/genetics , Transcription Factor AP-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...