Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Mol Cell Proteomics ; 23(8): 100809, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38936775

RESUMO

Microglia are resident immune cells of the brain and regulate its inflammatory state. In neurodegenerative diseases, microglia transition from a homeostatic state to a state referred to as disease-associated microglia (DAM). DAM express higher levels of proinflammatory signaling molecules, like STAT1 and TLR2, and show transitions in mitochondrial activity toward a more glycolytic response. Inhibition of Kv1.3 decreases the proinflammatory signature of DAM, though how Kv1.3 influences the response is unknown. Our goal was to identify the potential proteins interacting with Kv1.3 during transition to DAM. We utilized TurboID, a biotin ligase, fused to Kv1.3 to evaluate potential interacting proteins with Kv1.3 via mass spectrometry in BV-2 microglia following TLR4-mediated activation. Electrophysiology, Western blotting, and flow cytometry were used to evaluate Kv1.3 channel presence and TurboID biotinylation activity. We hypothesized that Kv1.3 contains domain-specific interactors that vary during a TLR4-induced inflammatory response, some of which are dependent on the PDZ-binding domain on the C terminus. We determined that the N terminus of Kv1.3 is responsible for trafficking Kv1.3 to the cell surface and mitochondria (e.g., NUDC, TIMM50). Whereas, the C terminus interacts with immune signaling proteins in a lipopolysaccharide-induced inflammatory response (e.g., STAT1, TLR2, and C3). There are 70 proteins that rely on the C-terminal PDZ-binding domain to interact with Kv1.3 (e.g., ND3, Snx3, and Sun1). Furthermore, we used Kv1.3 blockade to verify functional coupling between Kv1.3 and interferon-mediated STAT1 activation. Overall, we highlight that the Kv1.3 potassium channel functions beyond conducting the outward flux of potassium ions in an inflammatory context and that Kv1.3 modulates the activity of key immune signaling proteins, such as STAT1 and C3.

2.
Hum Mol Genet ; 33(1): 12-32, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37712894

RESUMO

Genes mutated in monogenic neurodevelopmental disorders are broadly expressed. This observation supports the concept that monogenic neurodevelopmental disorders are systemic diseases that profoundly impact neurodevelopment. We tested the systemic disease model focusing on Rett syndrome, which is caused by mutations in MECP2. Transcriptomes and proteomes of organs and brain regions from Mecp2-null mice as well as diverse MECP2-null male and female human cells were assessed. Widespread changes in the steady-state transcriptome and proteome were identified in brain regions and organs of presymptomatic Mecp2-null male mice as well as mutant human cell lines. The extent of these transcriptome and proteome modifications was similar in cortex, liver, kidney, and skeletal muscle and more pronounced than in the hippocampus and striatum. In particular, Mecp2- and MECP2-sensitive proteomes were enriched in synaptic and metabolic annotated gene products, the latter encompassing lipid metabolism and mitochondrial pathways. MECP2 mutations altered pyruvate-dependent mitochondrial respiration while maintaining the capacity to use glutamine as a mitochondrial carbon source. We conclude that mutations in Mecp2/MECP2 perturb lipid and mitochondrial metabolism systemically limiting cellular flexibility to utilize mitochondrial fuels.


Assuntos
Proteoma , Síndrome de Rett , Animais , Feminino , Humanos , Masculino , Camundongos , Encéfalo/metabolismo , Modelos Animais de Doenças , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Proteoma/genética , Proteoma/metabolismo , Síndrome de Rett/genética , Síndrome de Rett/metabolismo
3.
J Neurosci ; 41(31): 6596-6616, 2021 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-34261699

RESUMO

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.


Assuntos
Deficiências do Desenvolvimento , Mitocôndrias/fisiologia , Proteínas Mitocondriais/genética , Transportadores de Ânions Orgânicos/genética , Proteostase/genética , Ribonucleoproteínas/genética , Proteínas Ribossômicas/genética , Animais , Linhagem Celular , Deficiências do Desenvolvimento/genética , Deficiências do Desenvolvimento/metabolismo , Deficiências do Desenvolvimento/fisiopatologia , Drosophila , Regulação da Expressão Gênica/genética , Humanos , Neurogênese/fisiologia , Biossíntese de Proteínas/genética , Ratos , Ratos Sprague-Dawley , Ribossomos/fisiologia
4.
J Neurosci ; 41(2): 215-233, 2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33208468

RESUMO

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.


Assuntos
Cobre/fisiologia , Complexo de Golgi/fisiologia , Homeostase/fisiologia , Biogênese de Organelas , Sinapses/fisiologia , Adenosina Trifosfatases/metabolismo , Animais , Animais Geneticamente Modificados , Linhagem Celular , Cobre/toxicidade , ATPases Transportadoras de Cobre/genética , Drosophila , Estimulação Elétrica , Espaço Extracelular/metabolismo , Feminino , Humanos , Masculino , RNA Interferente Pequeno , Sinapses/ultraestrutura
5.
Proc Natl Acad Sci U S A ; 116(25): 12167-12172, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31160463

RESUMO

Copper is controlled by a sophisticated network of transport and storage proteins within mammalian cells, yet its uptake and efflux occur with rapid kinetics. Present as Cu(I) within the reducing intracellular environment, the nature of this labile copper pool remains elusive. While glutathione is involved in copper homeostasis and has been assumed to buffer intracellular copper, we demonstrate with a ratiometric fluorescent indicator, crisp-17, that cytosolic Cu(I) levels are buffered to the vicinity of 1 aM, where negligible complexation by glutathione is expected. Enabled by our phosphine sulfide-stabilized phosphine (PSP) ligand design strategy, crisp-17 offers a Cu(I) dissociation constant of 8 aM, thus exceeding the binding affinities of previous synthetic Cu(I) probes by four to six orders of magnitude. Two-photon excitation microscopy with crisp-17 revealed rapid, reversible increases in intracellular Cu(I) availability upon addition of the ionophoric complex CuGTSM or the thiol-selective oxidant 2,2'-dithiodipyridine (DTDP). While the latter effect was dramatically enhanced in 3T3 cells grown in the presence of supplemental copper and in cultured Menkes mutant fibroblasts exhibiting impaired copper efflux, basal Cu(I) availability in these cells showed little difference from controls, despite large increases in total copper content. Intracellular copper is thus tightly buffered by endogenous thiol ligands with significantly higher affinity than glutathione. The dual utility of crisp-17 to detect normal intracellular buffered Cu(I) levels as well as to probe the depth of the labile copper pool in conjunction with DTDP provides a promising strategy to characterize perturbations of cellular copper homeostasis.


Assuntos
Cobre/metabolismo , Síndrome dos Cabelos Torcidos/metabolismo , Soluções Tampão , Fibroblastos/metabolismo , Corantes Fluorescentes , Glutationa/metabolismo , Ligantes , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Mutação , Fosfinas/metabolismo
6.
Am J Physiol Cell Physiol ; 321(3): C519-C534, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34319827

RESUMO

Mitochondria are recognized as signaling organelles, because under stress, mitochondria can trigger various signaling pathways to coordinate the cell's response. The specific pathway(s) engaged by mitochondria in response to mitochondrial energy defects in vivo and in high-energy tissues like the heart are not fully understood. Here, we investigated cardiac pathways activated in response to mitochondrial energy dysfunction by studying mice with cardiomyocyte-specific loss of the mitochondrial phosphate carrier (SLC25A3), an established model that develops cardiomyopathy as a result of defective mitochondrial ATP synthesis. Mitochondrial energy dysfunction induced a striking pattern of acylome remodeling, with significantly increased posttranslational acetylation and malonylation. Mass spectrometry-based proteomics further revealed that energy dysfunction-induced remodeling of the acetylome and malonylome preferentially impacts mitochondrial proteins. Acetylation and malonylation modified a highly interconnected interactome of mitochondrial proteins, and both modifications were present on the enzyme isocitrate dehydrogenase 2 (IDH2). Intriguingly, IDH2 activity was enhanced in SLC25A3-deleted mitochondria, and further study of IDH2 sites targeted by both acetylation and malonylation revealed that these modifications can have site-specific and distinct functional effects. Finally, we uncovered a novel cross talk between the two modifications, whereby mitochondrial energy dysfunction-induced acetylation of sirtuin 5 (SIRT5), inhibited its function. Because SIRT5 is a mitochondrial deacylase with demalonylase activity, this finding suggests that acetylation can modulate the malonylome. Together, our results position acylations as an arm of the mitochondrial response to energy dysfunction and suggest a mechanism by which focal disruption to the energy production machinery can have an expanded impact on global mitochondrial function.


Assuntos
Cardiomiopatias/genética , Proteínas de Transporte de Cátions/genética , Isocitrato Desidrogenase/genética , Mitocôndrias Cardíacas/metabolismo , Proteínas Mitocondriais/genética , Miócitos Cardíacos/metabolismo , Proteínas de Transporte de Fosfato/genética , Processamento de Proteína Pós-Traducional , Proteínas Carreadoras de Solutos/genética , Acetilação , Animais , Transporte Biológico , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Proteínas de Transporte de Cátions/deficiência , Metabolismo Energético , Feminino , Redes Reguladoras de Genes , Isocitrato Desidrogenase/metabolismo , Masculino , Malonatos/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias Cardíacas/genética , Mitocôndrias Cardíacas/patologia , Proteínas Mitocondriais/deficiência , Modelos Moleculares , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/patologia , Proteínas de Transporte de Fosfato/deficiência , Fosfatos , Conformação Proteica , Mapeamento de Interação de Proteínas , Transdução de Sinais , Sirtuínas/genética , Sirtuínas/metabolismo , Proteínas Carreadoras de Solutos/deficiência
7.
J Neurosci Res ; 99(1): 67-89, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32436302

RESUMO

The function(s) of the Biogenesis of Lysosome-related Organelles Complex-1 (BLOC-1) during brain development is to date largely unknown. Here, we investigated how its absence alters the trajectory of postnatal brain development using as model the pallid mouse. Most of the defects observed early postnatally in the mutant mice were more prominent in males than in females and in the hippocampus. Male mutant mice, but not females, had smaller brains as compared to sex-matching wild types at postnatal day 1 (P1), this deficit was largely recovered by P14 and P45. An abnormal cytoarchitecture of the pyramidal cell layer of the hippocampus was observed in P1 pallid male, but not female, or juvenile mice (P45), along with severely decreased expression levels of the radial glial marker Glutamate-Aspartate Transporter. Transcriptomic analyses showed that the overall response to the lack of functional BLOC-1 was more pronounced in hippocampi at P1 than at P45 or in the cerebral cortex. These observations suggest that absence of BLOC-1 renders males more susceptible to perinatal brain maldevelopment and although most abnormalities appear to have been resolved in juvenile animals, still permanent defects may be present, resulting in faulty neuronal circuits, and contribute to previously reported cognitive and behavioral phenotypes in adult BLOC-1-deficient mice.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurogênese/fisiologia , Caracteres Sexuais , Animais , Animais Recém-Nascidos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes
8.
J Neurosci ; 39(18): 3561-3581, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30833507

RESUMO

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.


Assuntos
Síndrome da Deleção 22q11/metabolismo , Encéfalo/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Translocador 1 do Nucleotídeo Adenina/metabolismo , Animais , Comportamento Animal , Linhagem Celular , Deleção Cromossômica , Cromossomos Humanos Par 22/metabolismo , Drosophila , Feminino , Fibroblastos/metabolismo , Humanos , Masculino , Proteínas Mitocondriais/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Proteoma , Esquizofrenia/metabolismo
9.
Biochem Soc Trans ; 48(5): 2101-2115, 2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-32897294

RESUMO

Microcephaly is a rare, yet devastating, neurodevelopmental condition caused by genetic or environmental insults, such as the Zika virus infection. Microcephaly manifests with a severely reduced head circumference. Among the known heritable microcephaly genes, a significant proportion are annotated with centrosome-related ontologies. Centrosomes are microtubule-organizing centers, and they play fundamental roles in the proliferation of the neuronal progenitors, the neural stem cells (NSCs), which undergo repeated rounds of asymmetric cell division to drive neurogenesis and brain development. Many of the genes, pathways, and developmental paradigms that dictate NSC development in humans are conserved in Drosophila melanogaster. As such, studies of Drosophila NSCs lend invaluable insights into centrosome function within NSCs and help inform the pathophysiology of human microcephaly. This mini-review will briefly survey causative links between deregulated centrosome functions and microcephaly with particular emphasis on insights learned from Drosophila NSCs.


Assuntos
Centrossomo/ultraestrutura , Drosophila melanogaster/metabolismo , Células-Tronco Neurais/metabolismo , Animais , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Divisão Celular , Proliferação de Células , Modelos Animais de Doenças , Humanos , Microcefalia/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Mitose , Neurogênese , Neurônios , Células-Tronco/citologia , Zika virus , Infecção por Zika virus/metabolismo
10.
J Neurosci ; 38(44): 9364-9374, 2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-30381428

RESUMO

How do neurons adapt their endolysosomal system to address the particular challenge of membrane transport across their elaborate cellular landscape and to maintain proteostasis for the lifetime of the organism? Here we review recent findings that address this central question. We discuss the cellular and molecular mechanisms of endolysosomal trafficking and the autophagy pathway in neurons, as well as their role in neuronal development and degeneration. These studies highlight the importance of understanding the basic cell biology of endolysosomal trafficking and autophagy and their roles in the maintenance of proteostasis within the context of neurons, which will be critical for developing effective therapies for various neurodevelopmental and neurodegenerative disorders.


Assuntos
Autofagia/fisiologia , Endossomos/metabolismo , Lisossomos/metabolismo , Proteostase/fisiologia , Animais , Humanos , Transporte Proteico/fisiologia
11.
J Neurosci ; 36(49): 12393-12411, 2016 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-27927957

RESUMO

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.


Assuntos
Proteína 3 Relacionada a Actina/genética , Angiopoietinas/genética , Proteínas de Transporte/genética , Proteínas Associadas à Distrofina/genética , Lectinas/genética , Esquizofrenia/genética , Sinapses , Actinas/genética , Proteína 2 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Animais , Células Cultivadas , Citoesqueleto/genética , Drosophila melanogaster , Disbindina , Feminino , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Endogâmicos C57BL , Polimerização , Proteoma
12.
Hum Mol Genet ; 24(19): 5512-23, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26199316

RESUMO

Environmental factors and susceptible genomes interact to determine the risk of neurodevelopmental disorders. Although few genes and environmental factors have been linked, the intervening cellular and molecular mechanisms connecting a disorder susceptibility gene with environmental factors remain mostly unexplored. Here we focus on the schizophrenia susceptibility gene DTNBP1 and its product dysbindin, a subunit of the BLOC-1 complex, and describe a neuronal pathway modulating copper metabolism via ATP7A. Mutations in ATP7A result in Menkes disease, a disorder of copper metabolism. Dysbindin/BLOC-1 and ATP7A genetically and biochemically interact. Furthermore, disruption of this pathway causes alteration in the transcriptional profile of copper-regulatory and dependent factors in the hippocampus of dysbindin/BLOC-1-null mice. Dysbindin/BLOC-1 loss-of-function alleles do not affect cell and tissue copper content, yet they alter the susceptibility to toxic copper challenges in both mammalian cells and Drosophila. Our results demonstrate that perturbations downstream of the schizophrenia susceptibility gene DTNBP1 confer susceptibility to copper, a metal that in excess is a neurotoxin and whose depletion constitutes a micronutrient deficiency.


Assuntos
Cobre/metabolismo , Proteínas de Drosophila/genética , Proteínas Associadas à Distrofina/genética , Esquizofrenia/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Células Cultivadas , ATPases Transportadoras de Cobre , Modelos Animais de Doenças , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Disbindina , Proteínas Associadas à Distrofina/metabolismo , Predisposição Genética para Doença , Hipocampo/metabolismo , Camundongos , Neurônios/metabolismo
13.
J Neurosci ; 35(1): 325-38, 2015 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-25568125

RESUMO

Neurodevelopmental disorders arise from single or multiple gene defects. However, the way multiple loci interact to modify phenotypic outcomes remains poorly understood. Here, we studied phenotypes associated with mutations in the schizophrenia susceptibility gene dysbindin (dysb), in isolation or in combination with null alleles in the dysb network component Blos1. In humans, the Blos1 ortholog Bloc1s1 encodes a polypeptide that assembles, with dysbindin, into the octameric BLOC-1 complex. We biochemically confirmed BLOC-1 presence in Drosophila neurons, and measured synaptic output and complex adaptive behavior in response to BLOC-1 perturbation. Homozygous loss-of-function alleles of dysb, Blos1, or compound heterozygotes of these alleles impaired neurotransmitter release, synapse morphology, and homeostatic plasticity at the larval neuromuscular junction, and impaired olfactory habituation. This multiparameter assessment indicated that phenotypes were differentially sensitive to genetic dosages of loss-of-function BLOC-1 alleles. Our findings suggest that modification of a second genetic locus in a defined neurodevelopmental regulatory network does not follow a strict additive genetic inheritance, but rather, precise stoichiometry within the network determines phenotypic outcomes.


Assuntos
Proteínas de Transporte/genética , Proteínas de Drosophila/genética , Dosagem de Genes/fisiologia , Rede Nervosa/fisiologia , Plasticidade Neuronal/fisiologia , Esquizofrenia/genética , Sinapses/genética , Animais , Animais Geneticamente Modificados , Drosophila , Disbindina , Proteínas Associadas à Distrofina , Feminino , Rede Nervosa/ultraestrutura , Esquizofrenia/fisiopatologia , Sinapses/ultraestrutura , Transmissão Sináptica/fisiologia
14.
J Neurosci ; 35(19): 7643-53, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25972187

RESUMO

Dysbindin is a schizophrenia susceptibility factor and subunit of the biogenesis of lysosome-related organelles complex 1 (BLOC-1) required for lysosome-related organelle biogenesis, and in neurons, synaptic vesicle assembly, neurotransmission, and plasticity. Protein networks, or interactomes, downstream of dysbindin/BLOC-1 remain partially explored despite their potential to illuminate neurodevelopmental disorder mechanisms. Here, we conducted a proteome-wide search for polypeptides whose cellular content is sensitive to dysbindin/BLOC-1 loss of function. We identified components of the vesicle fusion machinery as factors downregulated in dysbindin/BLOC-1 deficiency in neuroectodermal cells and iPSC-derived human neurons, among them the N-ethylmaleimide-sensitive factor (NSF). Human dysbindin/BLOC-1 coprecipitates with NSF and vice versa, and both proteins colocalized in a Drosophila model synapse. To test the hypothesis that NSF and dysbindin/BLOC-1 participate in a pathway-regulating synaptic function, we examined the role for NSF in dysbindin/BLOC-1-dependent synaptic homeostatic plasticity in Drosophila. As previously described, we found that mutations in dysbindin precluded homeostatic synaptic plasticity elicited by acute blockage of postsynaptic receptors. This dysbindin mutant phenotype is fully rescued by presynaptic expression of either dysbindin or Drosophila NSF. However, neither reduction of NSF alone or in combination with dysbindin haploinsufficiency impaired homeostatic synaptic plasticity. Our results demonstrate that dysbindin/BLOC-1 expression defects result in altered cellular content of proteins of the vesicle fusion apparatus and therefore influence synaptic plasticity.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas Associadas à Distrofina/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas Sensíveis a N-Etilmaleimida/metabolismo , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Animais , Animais Geneticamente Modificados , Linhagem Celular Tumoral , Drosophila , Proteínas de Drosophila/genética , Disbindina , Proteínas Associadas à Distrofina/genética , Regulação da Expressão Gênica/genética , Humanos , Melanoma/patologia , Proteínas Sensíveis a N-Etilmaleimida/genética , Rede Nervosa/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroblastoma/patologia , Junção Neuromuscular/genética , Junção Neuromuscular/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/genética , Proteínas SNARE/metabolismo , Sinapses/genética , Vesículas Sinápticas/genética , Vesículas Sinápticas/metabolismo
15.
J Neurophysiol ; 115(1): 470-85, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26538609

RESUMO

Conventional anti-Parkinsonian dopamine replacement therapy is often complicated by side effects that limit the use of these medications. There is a continuing need to develop nondopaminergic approaches to treat Parkinsonism. One such approach is to use medications that normalize dopamine depletion-related firing abnormalities in the basal ganglia-thalamocortical circuitry. In this study, we assessed the potential of a specific T-type calcium channel blocker (ML218) to eliminate pathologic burst patterns of firing in the basal ganglia-receiving territory of the motor thalamus in Parkinsonian monkeys. We also carried out an anatomical study, demonstrating that the immunoreactivity for T-type calcium channels is strongly expressed in the motor thalamus in normal and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys. At the electron microscopic level, dendrites accounted for >90% of all tissue elements that were immunoreactive for voltage-gated calcium channel, type 3.2-containing T-type calcium channels in normal and Parkinsonian monkeys. Subsequent in vivo electrophysiologic studies in awake MPTP-treated Parkinsonian monkeys demonstrated that intrathalamic microinjections of ML218 (0.5 µl of a 2.5-mM solution, injected at 0.1-0.2 µl/min) partially normalized the thalamic activity by reducing the proportion of rebound bursts and increasing the proportion of spikes in non-rebound bursts. The drug also attenuated oscillatory activity in the 3-13-Hz frequency range and increased gamma frequency oscillations. However, ML218 did not normalize Parkinsonism-related changes in firing rates and oscillatory activity in the beta frequency range. Whereas the described changes are promising, a more complete assessment of the cellular and behavioral effects of ML218 (or similar drugs) is needed for a full appraisal of their anti-Parkinsonian potential.


Assuntos
Compostos Azabicíclicos/administração & dosagem , Compostos Azabicíclicos/farmacologia , Benzamidas/administração & dosagem , Benzamidas/farmacologia , Bloqueadores dos Canais de Cálcio/administração & dosagem , Canais de Cálcio Tipo T/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Transtornos Parkinsonianos/fisiopatologia , Tálamo/efeitos dos fármacos , Tálamo/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Gânglios da Base/metabolismo , Gânglios da Base/ultraestrutura , Canais de Cálcio Tipo T/metabolismo , Dendritos/metabolismo , Dendritos/ultraestrutura , Macaca mulatta , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Vias Neurais/ultraestrutura , Transtornos Parkinsonianos/metabolismo , Tálamo/metabolismo , Tálamo/ultraestrutura
16.
Biol Cell ; 107(7): 245-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26133153

RESUMO

The EMBO worskhop at the "end of the world'" (al fin del mundo), a meeting on membrane trafficking and its implication for polarity and diseases, took place in the Chilean Patagonia surrounded by the landscapes once witnessed by Charles Darwin. The meeting showcased some of the best membrane trafficking science with an emphasis in neuroscience and disease models. Speakers from Europe, USA, South America and the graduate students behind it; embarked on an enthusiastic and eclectic dialog where a wide range of cell types, model genetic systems, and diseases where discussed. This meeting demonstrated the power of trafficking concepts to integrate diverse biology and to formulate mechanisms of normal and disease cells.


Assuntos
Membrana Celular , Doenças do Sistema Nervoso , Neurociências , Animais , Transporte Biológico Ativo , Chile , Congressos como Assunto , Educação , Humanos
17.
J Biol Chem ; 289(20): 14291-300, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24713699

RESUMO

Post-mortem analysis has revealed reduced levels of the protein dysbindin in the brains of those suffering from the neurodevelopmental disorder schizophrenia. Consequently, mechanisms controlling the cellular levels of dysbindin and its interacting partners may participate in neurodevelopmental processes impaired in that disorder. To address this question, we studied loss of function mutations in the genes encoding dysbindin and its interacting BLOC-1 subunits. We focused on BLOC-1 mutants affecting synapse composition and function in addition to their established systemic pigmentation, hematological, and lung phenotypes. We tested phenotypic homogeneity and gene dosage effects in the mouse null alleles muted (Bloc1s5(mu/mu)) and dysbindin (Bloc1s8(sdy/sdy)). Transcripts of NMDA receptor subunits and GABAergic interneuron markers, as well as expression of BLOC-1 subunit gene products, were affected differently in the brains of Bloc1s5(mu/mu) and Bloc1s8(sdy/sdy) mice. Unlike Bloc1s8(sdy/sdy), elimination of one or two copies of Bloc1s5 generated indistinguishable pallidin transcript phenotypes. We conclude that monogenic mutations abrogating the expression of a protein complex subunit differentially affect the expression of other complex transcripts and polypeptides as well as their downstream effectors. We propose that the genetic disruption of different subunits of protein complexes and combinations thereof diversifies phenotypic presentation of pathway deficiencies, contributing to the wide phenotypic spectrum and complexity of neurodevelopmental disorders.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas Mutantes/metabolismo , Mutação , Fenótipo , Subunidades Proteicas/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Animais , Disbindina , Proteínas Associadas à Distrofina , Hipocampo/metabolismo , Humanos , Camundongos , Proteínas Mutantes/genética , Neurotransmissores/metabolismo , Pigmentação/genética , Subunidades Proteicas/genética , Esquizofrenia/etiologia , Esquizofrenia/genética , Esquizofrenia/metabolismo , Transcrição Gênica/genética , Ácido gama-Aminobutírico/metabolismo
18.
Neurobiol Dis ; 81: 154-61, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25583185

RESUMO

ATP7A mutations impair copper metabolism resulting in three distinct genetic disorders in humans. These diseases are characterized by neurological phenotypes ranging from intellectual disability to neurodegeneration. Severe ATP7A loss-of-function alleles trigger Menkes disease, a copper deficiency condition where systemic and neurodegenerative phenotypes dominate clinical outcomes. The pathogenesis of these manifestations has been attributed to the hypoactivity of a limited number of copper-dependent enzymes, a hypothesis that we refer as the oligoenzymatic pathogenic hypothesis. This hypothesis, which has dominated the field for 25 years, only explains some systemic Menkes phenotypes. However, we argue that this hypothesis does not fully account for the Menkes neurodegeneration or neurodevelopmental phenotypes. Here, we propose revisions of the oligoenzymatic hypothesis that could illuminate the pathogenesis of Menkes neurodegeneration and neurodevelopmental defects through unsuspected overlap with other neurological conditions including Parkinson's, intellectual disability, and schizophrenia.


Assuntos
Adenosina Trifosfatases/genética , Proteínas de Transporte de Cátions/genética , Síndrome dos Cabelos Torcidos/genética , Mutação/genética , Doenças Neurodegenerativas/genética , Transtornos do Neurodesenvolvimento/genética , Animais , Biologia Computacional , ATPases Transportadoras de Cobre , Humanos , Síndrome dos Cabelos Torcidos/complicações , Camundongos , Doenças Neurodegenerativas/etiologia , Transtornos do Neurodesenvolvimento/etiologia
19.
Hum Mol Genet ; 22(25): 5215-28, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-23918659

RESUMO

Mutations in Vps33 isoforms cause pigment dilution in mice (Vps33a, buff) and Drosophila (car) and the neurogenic arthrogryposis, renal dysfunction and cholestasis syndrome in humans (ARC1, VPS33B). The later disease is also caused by mutations in VIPAS39, (Vps33b interacting protein, apical-basolateral polarity regulator, SPE-39 homolog; ARC2), a protein that interacts with the HOmotypic fusion and Protein Sorting (HOPS) complex, a tether necessary for endosome-lysosome traffic. These syndromes offer insight into fundamental endosome traffic processes unique to metazoans. However, the molecular and cellular mechanisms underlying these mutant phenotypes remain poorly understood. Here we investigate interactions of wild-type and disease-causing mutations in VIPAS39/SPE-39 and Vps33b by yeast two hybrid, immunoprecipitation and quantitative fluorescent microscopy. We find that although few mutations prevent interaction between VIPAS39/SPE-39 and Vps33b, some mutants fragment VIPAS39/SPE-39-positive endosomes, but all mutants alter the subcellular localization of Vps33b to VIPAS39/SPE-39-positive endosomes. Our data suggest that the ARC syndrome may result through impaired VIPAS39/SPE-39 and Vps33b-dependent endosomal maturation or fusion.


Assuntos
Artrogripose/genética , Proteínas de Transporte/genética , Colestase/genética , Endossomos/genética , Insuficiência Renal/genética , Proteínas de Transporte Vesicular/genética , Animais , Artrogripose/patologia , Proteínas de Transporte/metabolismo , Colestase/patologia , Endossomos/patologia , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lisossomos/genética , Lisossomos/metabolismo , Proteínas de Membrana , Camundongos , Mutação , Proteínas Nucleares/genética , Transporte Proteico/genética , Insuficiência Renal/patologia , Técnicas do Sistema de Duplo-Híbrido , Proteínas de Transporte Vesicular/metabolismo
20.
Proc Natl Acad Sci U S A ; 109(26): 10376-81, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22685202

RESUMO

The newly discovered Ca(2+)-activated Cl(-) channel (CaCC), Anoctamin 1 (Ano1 or TMEM16A), has been implicated in vital physiological functions including epithelial fluid secretion, gut motility, and smooth muscle tone. Overexpression of Ano1 in HEK cells or Xenopus oocytes is sufficient to generate Ca(2+)-activated Cl(-) currents, but the details of channel composition and the regulatory factors that control channel biology are incompletely understood. We used a highly sensitive quantitative SILAC proteomics approach to obtain insights into stoichiometric protein networks associated with the Ano1 channel. These studies provide a comprehensive footprint of putative Ano1 regulatory networks. We find that Ano1 associates with the signaling/scaffolding proteins ezrin, radixin, moesin, and RhoA, which link the plasma membrane to the cytoskeleton with very high stoichiometry. Ano1, ezrin, and moesin/radixin colocalize apically in salivary gland epithelial cells, and overexpression of moesin and Ano1 in HEK cells alters the subcellular localization of both proteins. Moreover, interfering RNA for moesin modifies Ano1 current without affecting its surface expression level. Another network associated with Ano1 includes the SNARE and SM proteins VAMP3, syntaxins 2 and -4, and syntaxin-binding proteins munc18b and munc18c, which are integral to translocation of vesicles to the plasma membrane. A number of other regulatory proteins, including GTPases, Ca(2+)-binding proteins, kinases, and lipid-interacting proteins are enriched in the Ano1 complex. These data provide stoichiometrically prioritized information about mechanisms regulating Ano1 function and trafficking to polarized domains of the plasma membrane.


Assuntos
Canais de Cloreto/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Anoctamina-1 , Linhagem Celular , Cromatografia de Afinidade , Eletroforese em Gel de Poliacrilamida , Humanos , Xenopus
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa