Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Cell ; 151(4): 821-834, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23141539

RESUMEN

Neuronal activity influences genes involved in circuit development and information processing. However, the molecular basis of this process remains poorly understood. We found that HDAC4, a histone deacetylase that shuttles between the nucleus and cytoplasm, controls a transcriptional program essential for synaptic plasticity and memory. The nuclear import of HDAC4 and its association with chromatin is negatively regulated by NMDA receptors. In the nucleus, HDAC4 represses genes encoding constituents of central synapses, thereby affecting synaptic architecture and strength. Furthermore, we show that a truncated form of HDAC4 encoded by an allele associated with mental retardation is a gain-of-function nuclear repressor that abolishes transcription and synaptic transmission despite the loss of the deacetylase domain. Accordingly, mice carrying a mutant that mimics this allele exhibit deficits in neurotransmission, spatial learning, and memory. These studies elucidate a mechanism of experience-dependent plasticity and define the biological role of HDAC4 in the brain.


Asunto(s)
Transporte Activo de Núcleo Celular , Encéfalo/metabolismo , Histona Desacetilasas/metabolismo , Memoria , Plasticidad Neuronal , Neuronas/metabolismo , Sinapsis/metabolismo , Transcripción Genética , Animales , Ratones , Prosencéfalo/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Factores de Transcripción/metabolismo
2.
Mol Cell ; 71(5): 689-702.e9, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30193096

RESUMEN

Hsp90 is an essential chaperone that guards proteome integrity and amounts to 2% of cellular protein. We now find that Hsp90 also has the ability to directly interact with and deform membranes via an evolutionarily conserved amphipathic helix. Using a new cell-free system and in vivo measurements, we show this amphipathic helix allows exosome release by promoting the fusion of multivesicular bodies (MVBs) with the plasma membrane. We dissect the relationship between Hsp90 conformation and membrane-deforming function and show that mutations and drugs that stabilize the open Hsp90 dimer expose the helix and allow MVB fusion, while these effects are blocked by the closed state. Hence, we structurally separated the Hsp90 membrane-deforming function from its well-characterized chaperone activity, and we show that this previously unrecognized function is required for exosome release.


Asunto(s)
Membrana Celular/metabolismo , Exosomas/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Animales , Sistema Libre de Células/metabolismo , Drosophila/metabolismo , Femenino , Masculino , Chaperonas Moleculares/metabolismo , Cuerpos Multivesiculares/metabolismo , Unión Proteica/fisiología , Conformación Proteica
3.
EMBO J ; 40(17): e106914, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34313336

RESUMEN

The interphase nuclear envelope (NE) is extensively remodeled during nuclear pore complex (NPC) insertion. How this remodeling occurs and why it requires Torsin ATPases, which also regulate lipid metabolism, remains poorly understood. Here, we show that Drosophila Torsin (dTorsin) affects lipid metabolism via the NEP1R1-CTDNEP1 phosphatase and the Lipin phosphatidic acid (PA) phosphatase. This includes that Torsins remove NEP1R1-CTDNEP1 from the NE in fly and mouse cells, leading to subsequent Lipin exclusion from the nucleus. NEP1R1-CTDNEP1 downregulation also restores nuclear pore membrane fusion in post-mitotic dTorsinKO fat body cells. However, dTorsin-associated nuclear pore defects do not correlate with lipidomic abnormalities and are not resolved by silencing of Lipin. Further testing confirmed that membrane fusion continues in cells with hyperactivated Lipin. It also led to the surprising finding that excessive PA metabolism inhibits recruitment of the inner ring complex Nup35 subunit, resulting in elongated channel-like structures in place of mature nuclear pores. We conclude that the NEP1R1-CTDNEP1 phosphatase affects interphase NPC biogenesis by lipid-dependent and lipid-independent mechanisms, explaining some of the pleiotropic effects of Torsins.


Asunto(s)
Proteínas de Drosophila/metabolismo , Poro Nuclear/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Cuerpo Adiposo/citología , Cuerpo Adiposo/metabolismo , Metabolismo de los Lípidos , Fusión de Membrana , Fosfoproteínas Fosfatasas/genética
4.
Proc Natl Acad Sci U S A ; 116(1): 277-286, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30578322

RESUMEN

The mitochondrial intramembrane rhomboid protease PARL has been implicated in diverse functions in vitro, but its physiological role in vivo remains unclear. Here we show that Parl ablation in mouse causes a necrotizing encephalomyelopathy similar to Leigh syndrome, a mitochondrial disease characterized by disrupted energy production. Mice with conditional PARL deficiency in the nervous system, but not in muscle, develop a similar phenotype as germline Parl KOs, demonstrating the vital role of PARL in neurological homeostasis. Genetic modification of two major PARL substrates, PINK1 and PGAM5, do not modify this severe neurological phenotype. Parl-/- brain mitochondria are affected by progressive ultrastructural changes and by defects in Complex III (CIII) activity, coenzyme Q (CoQ) biosynthesis, and mitochondrial calcium metabolism. PARL is necessary for the stable expression of TTC19, which is required for CIII activity, and of COQ4, which is essential in CoQ biosynthesis. Thus, PARL plays a previously overlooked constitutive role in the maintenance of the respiratory chain in the nervous system, and its deficiency causes progressive mitochondrial dysfunction and structural abnormalities leading to neuronal necrosis and Leigh-like syndrome.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Enfermedad de Leigh/etiología , Metaloproteasas/deficiencia , Proteínas Mitocondriales/deficiencia , Ubiquinona/metabolismo , Animales , Encéfalo/metabolismo , Calcio/metabolismo , Enfermedad de Leigh/metabolismo , Enfermedad de Leigh/fisiopatología , Hígado/metabolismo , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Encefalomiopatías Mitocondriales/metabolismo , Encefalomiopatías Mitocondriales/fisiopatología , Músculo Esquelético/metabolismo , Especies Reactivas de Oxígeno/metabolismo
5.
BMC Biol ; 19(1): 152, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34330271

RESUMEN

BACKGROUND: Array tomography (AT) is a high-resolution imaging method to resolve fine details at the organelle level and has the advantage that it can provide 3D volumes to show the tissue context. AT can be carried out in a correlative way, combing light and electron microscopy (LM, EM) techniques. However, the correlation between modalities can be a challenge and delineating specific regions of interest in consecutive sections can be time-consuming. Integrated light and electron microscopes (iLEMs) offer the possibility to provide well-correlated images and may pose an ideal solution for correlative AT. Here, we report a workflow to automate navigation between regions of interest. RESULTS: We use a targeted approach that allows imaging specific tissue features, like organelles, cell processes, and nuclei at different scales to enable fast, directly correlated in situ AT using an integrated light and electron microscope (iLEM-AT). Our workflow is based on the detection of section boundaries on an initial transmitted light acquisition that serves as a reference space to compensate for changes in shape between sections, and we apply a stepwise refinement of localizations as the magnification increases from LM to EM. With minimal user interaction, this enables autonomous and speedy acquisition of regions containing cells and cellular organelles of interest correlated across different magnifications for LM and EM modalities, providing a more efficient way to obtain 3D images. We provide a proof of concept of our approach and the developed software tools using both Golgi neuronal impregnation staining and fluorescently labeled protein condensates in cells. CONCLUSIONS: Our method facilitates tracing and reconstructing cellular structures over multiple sections, is targeted at high resolution ILEMs, and can be integrated into existing devices, both commercial and custom-built systems.


Asunto(s)
Imagenología Tridimensional , Tomografía , Coloración y Etiquetado , Tomografía Computarizada por Rayos X , Flujo de Trabajo
6.
EMBO J ; 36(10): 1392-1411, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28331029

RESUMEN

Presynaptic terminals are metabolically active and accrue damage through continuous vesicle cycling. How synapses locally regulate protein homeostasis is poorly understood. We show that the presynaptic lipid phosphatase synaptojanin is required for macroautophagy, and this role is inhibited by the Parkinson's disease mutation R258Q. Synaptojanin drives synaptic endocytosis by dephosphorylating PI(4,5)P2, but this function appears normal in SynaptojaninRQ knock-in flies. Instead, R258Q affects the synaptojanin SAC1 domain that dephosphorylates PI(3)P and PI(3,5)P2, two lipids found in autophagosomal membranes. Using advanced imaging, we show that SynaptojaninRQ mutants accumulate the PI(3)P/PI(3,5)P2-binding protein Atg18a on nascent synaptic autophagosomes, blocking autophagosome maturation at fly synapses and in neurites of human patient induced pluripotent stem cell-derived neurons. Additionally, we observe neurodegeneration, including dopaminergic neuron loss, in SynaptojaninRQ flies. Thus, synaptojanin is essential for macroautophagy within presynaptic terminals, coupling protein turnover with synaptic vesicle cycling and linking presynaptic-specific autophagy defects to Parkinson's disease.


Asunto(s)
Autofagosomas/metabolismo , Autofagia , Proteínas del Tejido Nervioso/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Terminales Presinápticos/enzimología , Terminales Presinápticos/metabolismo , Sustitución de Aminoácidos , Animales , Proteínas Relacionadas con la Autofagia/análisis , Células Cultivadas , Drosophila , Humanos , Proteínas de la Membrana/análisis , Mutación Missense , Proteínas del Tejido Nervioso/genética , Enfermedad de Parkinson/patología , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolasas/genética
7.
J Cell Sci ; 127(Pt 13): 2825-39, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24806965

RESUMEN

Searching and evaluating the Human Protein Atlas for transmembrane proteins enabled us to identify an integral membrane protein, TMEM115, that is enriched in the Golgi complex. Biochemical and cell biological analysis suggested that TMEM115 has four candidate transmembrane domains located in the N-terminal region. Both the N- and C-terminal domains are oriented towards the cytoplasm. Immunofluorescence analysis supports that TMEM115 is enriched in the Golgi cisternae. Functionally, TMEM115 knockdown or overexpression delays Brefeldin-A-induced Golgi-to-ER retrograde transport, phenocopying cells with mutations or silencing of the conserved oligomeric Golgi (COG) complex. Co-immunoprecipitation and in vitro binding experiments reveals that TMEM115 interacts with the COG complex, and might self-interact to form dimers or oligomers. A short region (residues 206-229) immediately to the C-terminal side of the fourth transmembrane domain is both necessary and sufficient for Golgi targeting. Knockdown of TMEM115 also reduces the binding of the lectins peanut agglutinin (PNA) and Helix pomatia agglutinin (HPA), suggesting an altered O-linked glycosylation profile. These results establish that TMEM115 is an integral membrane protein of the Golgi stack regulating Golgi-to-ER retrograde transport and is likely to be part of the machinery of the COG complex.


Asunto(s)
Aparato de Golgi/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Células HeLa , Humanos , Ratones , Unión Proteica , Transporte de Proteínas , Proteínas de Transporte Vesicular/metabolismo
8.
Development ; 140(18): 3819-25, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23924633

RESUMEN

Mobilization of endogenous retrotransposons can destabilize the genome, an imminent danger during epigenetic reprogramming of cells in the germline. The P-element-induced wimpy testis (PIWI)-interacting RNA (piRNA) pathway is known to silence retrotransposons in the mouse testes. Several piRNA pathway components localize to the unique, germline structure known as the nuage. In this study, we surveyed mouse ovaries and found, for the first time, transient appearance of nuage-like structures in oocytes of primordial follicles. Mouse vasa homolog (MVH), Piwi-like 2 (PIWIL2/MILI) and tudor domain-containing 9 (TDRD9) are present in these structures, whereas aggregates of germ cell protein with ankyrin repeats, sterile alpha motif and leucine zipper (GASZ) localize separately in the cytoplasm. Retrotransposons are silenced in primordial ovarian follicles, and de-repressed upon reduction of piRNA expression in Mvh, Mili or Gasz mutants. However, these null-mutant females, unlike their male counterparts, are fertile, uncoupling retrotransposon activation from sterility.


Asunto(s)
Estructuras Celulares/metabolismo , Silenciador del Gen , Folículo Ovárico/metabolismo , Retroelementos/genética , Animales , Estructuras Celulares/ultraestructura , Femenino , Regulación de la Expresión Génica , Células Germinativas/metabolismo , Infertilidad Femenina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Oogénesis , Folículo Ovárico/ultraestructura , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo
9.
EMBO Rep ; 15(6): 714-22, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24711543

RESUMEN

While molecular regulation of insulin granule exocytosis is relatively well understood, insulin granule biogenesis and maturation and its influence on glucose homeostasis are relatively unclear. Here, we identify a novel protein highly expressed in insulin-secreting cells and name it BIG3 due to its similarity to BIG/GBF of the Arf-GTP exchange factor (GEF) family. BIG3 is predominantly localized to insulin- and clathrin-positive trans-Golgi network (TGN) compartments. BIG3-deficient insulin-secreting cells display increased insulin content and granule number and elevated insulin secretion upon stimulation. Moreover, BIG3 deficiency results in faster processing of proinsulin to insulin and chromogranin A to ß-granin in ß-cells. BIG3-knockout mice exhibit postprandial hyperinsulinemia, hyperglycemia, impaired glucose tolerance, and insulin resistance. Collectively, these results demonstrate that BIG3 negatively modulates insulin granule biogenesis and insulin secretion and participates in the regulation of systemic glucose homeostasis.


Asunto(s)
Homeostasis/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas/metabolismo , Vesículas Secretoras/química , Animales , Calorimetría Indirecta , Glucosa/fisiología , Homeostasis/fisiología , Hiperglucemia/genética , Insulina/análisis , Resistencia a la Insulina/genética , Secreción de Insulina , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados , Proteínas/genética , Red trans-Golgi/metabolismo
10.
Mol Ther ; 22(9): 1593-604, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24903577

RESUMEN

Impaired wound healing is a major source of morbidity in diabetic patients. Poor outcome has, in part, been related to increased inflammation, poor angiogenesis, and deficiencies in extracellular matrix components. Despite the enormous impact of these chronic wounds, effective therapies are lacking. Here, we showed that the topical application of recombinant matricellular protein angiopoietin-like 4 (ANGPTL4) accelerated wound reepithelialization in diabetic mice, in part, by improving angiogenesis. ANGPTL4 expression is markedly elevated upon normal wound injury. In contrast, ANGPTL4 expression remains low throughout the healing period in diabetic wounds. Exogenous ANGPTL4 modulated several regulatory networks involved in cell migration, angiogenesis, and inflammation, as evidenced by an altered gene expression signature. ANGPTL4 influenced the expression profile of endothelial-specific CD31 in diabetic wounds, returning its profile to that observed in wild-type wounds. We showed ANGPTL4-induced nitric oxide production through an integrin/JAK/STAT3-mediated upregulation of inducible nitric oxide synthase (iNOS) expression in wound epithelia, thus revealing a hitherto unknown mechanism by which ANGPTL4 regulated angiogenesis via keratinocyte-to-endothelial-cell communication. These data show that the replacement of ANGPTL4 may be an effective adjunctive or new therapeutic avenue for treating poor healing wounds. The present finding also confirms that therapeutic angiogenesis remains an attractive treatment modality for diabetic wound healing.


Asunto(s)
Angiopoyetinas/administración & dosificación , Diabetes Mellitus Experimental/complicaciones , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Transcripción STAT3/metabolismo , Angiopoyetinas/farmacología , Animales , Comunicación Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Queratinocitos/metabolismo , Ratones , Repitelización , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Transducción de Señal
11.
Am J Physiol Endocrinol Metab ; 307(7): E611-8, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25139048

RESUMEN

We recently identified BIG3 as a negative regulator of insulin granule biogenesis and reported increased insulin secretion in BIG3-knockout (BKO) mice. To pinpoint the site of action for BIG3, we investigated whether BIG3 regulates quantal insulin granule exocytosis. We established an assay to detect insulin granule exocytosis by recording ATP-elicited currents at high temporal resolution by patch clamp. Similarly to insulin, ATP release was increased in BKO ß-cells. Although the frequency of insulin granule exocytosis was increased in BKO ß-cells, quantal size or release kinetics remained unchanged. Electron microscopy studies showed that the number of insulin granules was increased by >60% in BKO ß-cells. However, the number of morphologically docked granules was unaltered. The number of insulin granules having significant distances away from plasma membrane was greatly increased in BKO ß-cells. Thus, BIG3 negatively regulates insulin granule exocytosis by restricting insulin granule biogenesis without the release kinetics of individual granules at the final exocytotic steps being affected. Depletion of BIG3 leads to an enlarged releasable pool of insulin granules, which accounts for increased release frequency and consequently increased insulin secretion.


Asunto(s)
Membrana Celular/metabolismo , Gránulos Citoplasmáticos/metabolismo , Exocitosis/fisiología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Potenciales de la Membrana/fisiología , Proteínas/fisiología , Animales , Secreción de Insulina , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Proteínas/genética
12.
Transl Psychiatry ; 14(1): 29, 2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38233378

RESUMEN

The neuropeptide corticotropin-releasing factor (CRF) exerts a pivotal role in modulating neuronal activity in the mammalian brain. The effects of CRF exhibit notable variations, depending on factors such as duration of exposure, concentration, and anatomical location. In the CA1 region of the hippocampus, the impact of CRF is dichotomous: chronic exposure to CRF impairs synapse formation and dendritic integrity, whereas brief exposure enhances synapse formation and plasticity. In the current study, we demonstrate long-term effects of acute CRF on the density and stability of mature mushroom spines ex vivo. We establish that both CRF receptors are present in this hippocampal region, and we pinpoint their precise subcellular localization within synapses by electron microscopy. Furthermore, both in vivo and ex vivo data collectively demonstrate that a transient surge of CRF in the CA1 activates the cyclin-dependent kinase 5 (Cdk5)-pathway. This activation leads to a notable augmentation in CRF-dependent spine formation. Overall, these data suggest that upon acute release of CRF in the CA1-SR synapse, both CRF-Rs can be activated and promote synaptic plasticity via activating different downstream signaling pathways, such as the Cdk5-pathway.


Asunto(s)
Hormona Liberadora de Corticotropina , Espinas Dendríticas , Animales , Hormona Liberadora de Corticotropina/metabolismo , Espinas Dendríticas/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Quinasa 5 Dependiente de la Ciclina/farmacología , Hipocampo/metabolismo , Receptores de Hormona Liberadora de Corticotropina , Sinapsis/metabolismo , Mamíferos/metabolismo
13.
J Neurosci ; 32(24): 8391-400, 2012 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-22699919

RESUMEN

Glutamatergic synapses are located mostly on dendritic spines in the adult nervous system. The spines serve as postsynaptic compartments, containing components that mediate and control the synaptic signal. Early in development, when glutamatergic synapses are initially forming, waves of excitatory activity pass through many parts of the nervous system and are driven in part by a class of heteropentameric ß2-containing nicotinic acetylcholine receptors (ß2*-nAChRs). These ß2*-nAChRs are widely distributed and, when activated, can depolarize the membrane and elevate intracellular calcium levels in neurons. We show here that ß2*-nAChRs are essential for acquisition of normal numbers of dendritic spines during development. Mice constitutively lacking the ß2-nAChR gene have fewer dendritic spines than do age-matched wild-type mice at all times examined. Activation of ß2*-nAChRs by nicotine either in vivo or in organotypic slice culture quickly elevates the number of spines. RNA interference studies both in vivo and in organotypic culture demonstrate that the ß2*-nAChRs act in a cell-autonomous manner to increase the number of spines. The increase depends on intracellular calcium and activation of calcium, calmodulin-dependent protein kinase II. Absence of ß2*-nAChRs in vivo causes a disproportionate number of glutamatergic synapses to be localized on dendritic shafts, rather than on spines as occurs in wild type. This shift in synapse location is found both in the hippocampus and cortex, indicating the breadth of the effect. Because spine synapses differ from shaft synapses in their signaling capabilities, the shift observed is likely to have significant consequences for network function.


Asunto(s)
Espinas Dendríticas/metabolismo , Receptores Nicotínicos/fisiología , Animales , Calcio/metabolismo , Señalización del Calcio/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Espinas Dendríticas/efectos de los fármacos , Ácido Glutámico/metabolismo , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Ratones , Ratones Noqueados , Neuronas/citología , Neuronas/efectos de los fármacos , Nicotina/farmacología , Subunidades de Proteína/fisiología , ARN Interferente Pequeño/genética , Receptores Nicotínicos/genética , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/fisiología , Sinapsis/ultraestructura
14.
J Neurosci ; 32(22): 7651-61, 2012 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-22649244

RESUMEN

Glutamate is the primary excitatory transmitter in adult brain, acting through synapses on dendritic spines and shafts. Early in development, however, when glutamatergic synapses are only beginning to form, nicotinic cholinergic excitation is already widespread; it is mediated by acetylcholine activating nicotinic acetylcholine receptors (nAChRs) that generate waves of activity across brain regions. A major class of nAChRs contributing at this time is a species containing α7 subunits (α7-nAChRs). These receptors are highly permeable to calcium, influence a variety of calcium-dependent events, and are diversely distributed throughout the developing CNS. Here we show that α7-nAChRs unexpectedly promote formation of glutamatergic synapses during development. The dependence on α7-nAChRs becomes clear when comparing wild-type (WT) mice with mice constitutively lacking the α7-nAChR gene. Ultrastructural analysis, immunostaining, and patch-clamp recording all reveal synaptic deficits when α7-nAChR input is absent. Similarly, nicotinic activation of α7-nAChRs in WT organotypic culture, as well as cell culture, increases the number of glutamatergic synapses. RNA interference demonstrates that the α7-nAChRs must be expressed in the neuron being innervated for normal innervation to occur. Moreover, the deficits persist throughout the developmental period of major de novo synapse formation and are still fully apparent in the adult. GABAergic synapses, in contrast, are undiminished in number under such conditions. As a result, mice lacking α7-nAChRs have an altered balance in the excitatory/inhibitory input they receive. This ratio represents a fundamental feature of neural networks and shows for the first time that endogenous nicotinic cholinergic signaling plays a key role in network construction.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Ácido Glutámico/metabolismo , Neuronas/fisiología , Receptores Nicotínicos/fisiología , Sinapsis/fisiología , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Células Cultivadas , Homólogo 4 de la Proteína Discs Large , Estimulación Eléctrica , Embrión de Mamíferos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Femenino , Antagonistas del GABA/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Guanilato-Quinasas/metabolismo , Hipocampo/citología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Neuritas/metabolismo , Neuritas/ultraestructura , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Piridazinas/farmacología , Compuestos de Piridinio , Compuestos de Amonio Cuaternario , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Receptores AMPA/metabolismo , Receptores Nicotínicos/deficiencia , Receptores Nicotínicos/genética , Bloqueadores de los Canales de Sodio/farmacología , Sinapsis/ultraestructura , Tetrodotoxina/farmacología , Factores de Tiempo , Transducción Genética/métodos , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Corteza Visual/citología , Corteza Visual/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7
15.
Neuron ; 111(9): 1402-1422.e13, 2023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-36827984

RESUMEN

Neuronal activity causes use-dependent decline in protein function. However, it is unclear how this is coupled to local quality control mechanisms. We show in Drosophila that the endocytic protein Endophilin-A (EndoA) connects activity-induced calcium influx to synaptic autophagy and neuronal survival in a Parkinson disease-relevant fashion. Mutations in the disordered loop, including a Parkinson disease-risk mutation, render EndoA insensitive to neuronal stimulation and affect protein dynamics: when EndoA is more flexible, its mobility in membrane nanodomains increases, making it available for autophagosome formation. Conversely, when EndoA is more rigid, its mobility reduces, blocking stimulation-induced autophagy. Balanced stimulation-induced autophagy is required for dopagminergic neuron survival, and a variant in the human ENDOA1 disordered loop conferring risk to Parkinson disease also blocks nanodomain protein mobility and autophagy both in vivo and in human-induced dopaminergic neurons. Thus, we reveal a mechanism that neurons use to connect neuronal activity to local autophagy and that is critical for neuronal survival.


Asunto(s)
Enfermedad de Parkinson , Animales , Humanos , Autofagia/genética , Calcio/metabolismo , Neuronas Dopaminérgicas/metabolismo , Drosophila/metabolismo , Mutación/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo
16.
Science ; 379(6632): eabn4705, 2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36705539

RESUMEN

Neuronal development in the human cerebral cortex is considerably prolonged compared with that of other mammals. We explored whether mitochondria influence the species-specific timing of cortical neuron maturation. By comparing human and mouse cortical neuronal maturation at high temporal and cell resolution, we found a slower mitochondria development in human cortical neurons compared with that in the mouse, together with lower mitochondria metabolic activity, particularly that of oxidative phosphorylation. Stimulation of mitochondria metabolism in human neurons resulted in accelerated development in vitro and in vivo, leading to maturation of cells weeks ahead of time, whereas its inhibition in mouse neurons led to decreased rates of maturation. Mitochondria are thus important regulators of the pace of neuronal development underlying human-specific brain neoteny.


Asunto(s)
Mitocondrias , Neurogénesis , Neuronas , Animales , Humanos , Ratones , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Metabolismo Energético , Mitocondrias/metabolismo , Neuronas/metabolismo
17.
Methods Cell Biol ; 162: 205-221, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33707013

RESUMEN

Many areas of biology have benefited from advances in light microscopy (LM). However, one limitation of the LM approach is that numerous critically important aspects of subcellular machineries are well beyond the resolution of conventional LM. For studying these, electron microscopy (EM) remains the technique of choice to visualize and identify macromolecules at the ultrastructural level. The most powerful approach is combining both techniques, LM and EM (i.e., to apply correlative light/electron microscopy, CLEM) to image exactly the same region of interest. This combination allows, for example, to immuno-localize proteins by LM and then to visualize the ultrastructural context of the same region of the sample. However, the identification and correlation of the regions of interest (ROIs) at the levels of LM and EM remains a major challenge, mostly due to the difficulties with correlation along the Z-axis for both modalities. In this chapter, we address this difficulty and describe an approach for performing CLEM in tissue samples using marks from near-infrared branding as indicators of a ROI, and then using serial block face-scanning electron microscopy (SBF-SEM) to identify and approach this ROI. Once a ROI has been approached, serial sections are collected on grids for high-resolution imaging by transmission EM, and subsequent correlation with LM images showing labeled proteins.


Asunto(s)
Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión
18.
Transl Psychiatry ; 11(1): 378, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34234103

RESUMEN

Biological responses to stress are complex and highly conserved. Corticotropin-releasing factor (CRF) plays a central role in regulating these lifesaving physiological responses to stress. We show that, in mice, CRF rapidly changes Schaffer Collateral (SC) input into hippocampal CA1 pyramidal cells (PC) by modulating both functional and structural aspects of these synapses. Host exposure to acute stress, in vivo CRF injection, and ex vivo CRF application all result in fast de novo formation and remodeling of existing dendritic spines. Functionally, CRF leads to a rapid increase in synaptic strength of SC input into CA1 neurons, e.g., increase in spontaneous neurotransmitter release, paired-pulse facilitation, and repetitive excitability and improves synaptic plasticity: long-term potentiation (LTP) and long-term depression (LTD). In line with the changes in synaptic function, CRF increases the number of presynaptic vesicles, induces redistribution of vesicles towards the active zone, increases active zone size, and improves the alignment of the pre- and postsynaptic compartments. Therefore, CRF rapidly enhances synaptic communication in the hippocampus, potentially playing a crucial role in the enhanced memory consolidation in acute stress.


Asunto(s)
Hormona Liberadora de Corticotropina , Células Piramidales , Animales , Hipocampo , Potenciación a Largo Plazo , Ratones , Sinapsis , Transmisión Sináptica
19.
Front Cell Dev Biol ; 9: 737621, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34977003

RESUMEN

Life science research often needs to define where molecules are located within the complex environment of a cell or tissue. Genetically encoded fluorescent proteins and or fluorescence affinity-labeling are the go-to methods. Although recent fluorescent microscopy methods can provide localization of fluorescent molecules with relatively high resolution, an ultrastructural context is missing. This is solved by imaging a region of interest with correlative light and electron microscopy (CLEM). We have adopted a protocol that preserves both genetically-encoded and antibody-derived fluorescent signals in resin-embedded cell and tissue samples and provides high-resolution electron microscopy imaging of the same thin section. This method is particularly suitable for dedicated CLEM instruments that combine fluorescence and electron microscopy optics. In addition, we optimized scanning EM imaging parameters for samples of varying thicknesses. These protocols will enable rapid acquisition of CLEM information from samples and can be adapted for three-dimensional EM.

20.
Diabetes ; 70(2): 492-503, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33277337

RESUMEN

FURIN is a proprotein convertase (PC) responsible for proteolytic activation of a wide array of precursor proteins within the secretory pathway. It maps to the PRC1 locus, a type 2 diabetes susceptibility locus, but its specific role in pancreatic ß-cells is largely unknown. The aim of this study was to determine the role of FURIN in glucose homeostasis. We show that FURIN is highly expressed in human islets, whereas PCs that potentially could provide redundancy are expressed at considerably lower levels. ß-cell-specific Furin knockout (ßFurKO) mice are glucose intolerant as a result of smaller islets with lower insulin content and abnormal dense-core secretory granule morphology. mRNA expression analysis and differential proteomics on ßFurKO islets revealed activation of activating transcription factor 4 (ATF4), which was mediated by mammalian target of rapamycin C1 (mTORC1). ßFurKO cells show impaired cleavage or shedding of vacuolar-type ATPase (V-ATPase) subunits Ac45 and prorenin receptor, respectively, and impaired lysosomal acidification. Blocking V-ATPase pharmacologically in ß-cells increased mTORC1 activity, suggesting involvement of the V-ATPase proton pump in the phenotype. Taken together, these results suggest a model of mTORC1-ATF4 hyperactivation and impaired lysosomal acidification in ß-cells lacking Furin, causing ß-cell dysfunction.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Furina/metabolismo , Células Secretoras de Insulina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Furina/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA