Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 13(2): e1006603, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28182653

RESUMO

Environmental insults such as oxidative stress can damage cell membranes. Lysosomes are particularly sensitive to membrane permeabilization since their function depends on intraluminal acidic pH and requires stable membrane-dependent proton gradients. Among the catalog of oxidative stress-responsive genes is the Lipocalin Apolipoprotein D (ApoD), an extracellular lipid binding protein endowed with antioxidant capacity. Within the nervous system, cell types in the defense frontline, such as astrocytes, secrete ApoD to help neurons cope with the challenge. The protecting role of ApoD is known from cellular to organism level, and many of its downstream effects, including optimization of autophagy upon neurodegeneration, have been described. However, we still cannot assign a cellular mechanism to ApoD gene that explains how this protection is accomplished. Here we perform a comprehensive analysis of ApoD intracellular traffic and demonstrate its role in lysosomal pH homeostasis upon paraquat-induced oxidative stress. By combining single-lysosome in vivo pH measurements with immunodetection, we demonstrate that ApoD is endocytosed and targeted to a subset of vulnerable lysosomes in a stress-dependent manner. ApoD is functionally stable in this acidic environment, and its presence is sufficient and necessary for lysosomes to recover from oxidation-induced alkalinization, both in astrocytes and neurons. This function is accomplished by preventing lysosomal membrane permeabilization. Two lysosomal-dependent biological processes, myelin phagocytosis by astrocytes and optimization of neurodegeneration-triggered autophagy in a Drosophila in vivo model, require ApoD-related Lipocalins. Our results uncover a previously unknown biological function of ApoD, member of the finely regulated and evolutionary conserved gene family of extracellular Lipocalins. They set a lipoprotein-mediated regulation of lysosomal membrane integrity as a new mechanism at the hub of many cellular functions, critical for the outcome of a wide variety of neurodegenerative diseases. These results open therapeutic opportunities by providing a route of entry and a repair mechanism for lysosomes in pathological situations.


Assuntos
Astrócitos/metabolismo , Lisossomos/metabolismo , Neurônios/metabolismo , Estresse Oxidativo , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Apolipoproteínas D/genética , Apolipoproteínas D/metabolismo , Apolipoproteínas D/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/ultraestrutura , Autofagia/efeitos dos fármacos , Autofagia/genética , Linhagem Celular Tumoral , Células Cultivadas , Drosophila , Células HEK293 , Herbicidas/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Immunoblotting , Lipocalinas/farmacologia , Lisossomos/química , Camundongos Knockout , Microscopia Confocal , Microscopia Eletrônica , Modelos Biológicos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/prevenção & controle , Neurônios/efeitos dos fármacos , Paraquat/farmacologia , Fagossomos/metabolismo
2.
Glia ; 66(3): 670-687, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29222871

RESUMO

To compact the extracellular sides of myelin, an important transition must take place: from membrane sliding, while building the wraps, to membrane adhesion and water exclusion. Removal of the negatively charged glycocalyx becomes the limiting factor in such transition. What is required to initiate this membrane-zipping process? Knocking-out the Lipocalin Apolipoprotein D (ApoD), essential for lysosomal functional integrity in glial cells, results in a specific defect in myelin extracellular leaflet compaction in peripheral and central nervous system, which results in reduced conduction velocity and suboptimal behavioral outputs: motor learning is compromised. Myelination initiation, growth, intracellular leaflet compaction, myelin thickness or internodal length remain unaltered. Lack of ApoD specifically modifies Plp and P0 protein expression, but not Mbp or Mag. Late in myelin maturation period, ApoD affects lipogenic and growth-related, but not stress-responsive, signaling pathways. Without ApoD, the sialylated glycocalyx is maintained and ganglioside content remains high. In peripheral nervous system, Neu3 membrane sialidase and lysosomal Neu1 are coordinately expressed with ApoD in subsets of Schwann cells. ApoD-KO myelin becomes depleted of Neu3 and enriched in Fyn, a kinase with pivotal roles in transducing axon-derived signals into myelin properties. In the absence of ApoD, partial permeabilization of lysosomes alters Neu1 location as well. Exogenous ApoD rescues ApoD-KO hypersialylated glycocalyx in astrocytes, demonstrating that ApoD is necessary and sufficient to control glycocalyx composition in glial cells. By ensuring lysosomal functional integrity and adequate subcellular location of effector and regulatory proteins, ApoD guarantees the glycolipid recycling and glycocalyx removal required to complete myelin compaction.


Assuntos
Apolipoproteínas D/metabolismo , Glicocálix/metabolismo , Lisossomos/metabolismo , Bainha de Mielina/metabolismo , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Apolipoproteínas D/administração & dosagem , Apolipoproteínas D/genética , Astrócitos/citologia , Astrócitos/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Escherichia coli , Espaço Extracelular/metabolismo , Deficiências da Aprendizagem/metabolismo , Deficiências da Aprendizagem/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Mucolipidoses/metabolismo , Neuraminidase/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Nervo Isquiático/citologia , Nervo Isquiático/crescimento & desenvolvimento , Nervo Isquiático/metabolismo
3.
J Cell Sci ; 129(9): 1866-77, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26966186

RESUMO

Proper development of the nervous system requires a temporally and spatially orchestrated set of events including differentiation, synapse formation and neurotransmission. Nerve growth factor (NGF) acting through the TrkA neurotrophin receptor (also known as NTRK1) regulates many of these events. However, the molecular mechanisms responsible for NGF-regulated secretion are not completely understood. Here, we describe a new signaling pathway involving TrkA, ARMS (also known as Kidins220), synembryn-B and Rac1 in NGF-mediated secretion in PC12 cells. Whereas overexpression of ARMS blocked NGF-mediated secretion, without affecting basal secretion, a decrease in ARMS resulted in potentiation. Similar effects were observed with synembryn-B, a protein that interacts directly with ARMS. Downstream of ARMS and synembryn-B are Gαq and Trio proteins, which modulate the activity of Rac1 in response to NGF. Expression of dominant-negative Rac1 rescued the secretion defects of cells overexpressing ARMS or synembryn-B. Thus, this neurotrophin pathway represents a new mechanism responsible for NGF-regulated secretion.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas de Membrana/metabolismo , Fator de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Animais , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Proteínas de Membrana/genética , Camundongos , Fator de Crescimento Neural/genética , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Células PC12 , Fosfoproteínas/genética , Ratos , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
4.
J Cell Sci ; 129(4): 729-42, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26743081

RESUMO

Detailed immunocytochemical analyses comparing wild-type (WT), GRF1-knockout (KO), GRF2-KO and GRF1/2 double-knockout (DKO) mouse retinas uncovered the specific accumulation of misplaced, 'ectopic' cone photoreceptor nuclei in the photoreceptor segment (PS) area of retinas from GRF2-KO and GRF1/2-DKO, but not of WT or GRF1-KO mice. Localization of ectopic nuclei in the PS area of GRF2-depleted retinas occurred postnatally and peaked between postnatal day (P)11 and P15. Mechanistically, the generation of this phenotype involved disruption of the outer limiting membrane and intrusion into the PS layer by cone nuclei displaying significant perinuclear accumulation of signaling molecules known to participate in nuclear migration and cytoskeletal reorganization, such as PAR3, PAR6 and activated, phosphorylated forms of PAK, MLC2 and VASP. Electroretinographic recordings showed specific impairment of cone-mediated retinal function in GRF2-KO and GRF1/2-DKO retinas compared with WT controls. These data identify defective cone nuclear migration as a novel phenotype in mouse retinas lacking GRF2 and support a crucial role of GRF2 in control of the nuclear migration processes required for proper postnatal development and function of retinal cone photoreceptors.


Assuntos
Núcleo Celular/fisiologia , Células Fotorreceptoras Retinianas Cones/ultraestrutura , Fatores ras de Troca de Nucleotídeo Guanina/fisiologia , Animais , Transporte Biológico , Eletrorretinografia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Retina/citologia , Retina/fisiologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Transdução de Sinais
5.
J Neurosci ; 34(9): 3320-39, 2014 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-24573290

RESUMO

Overexpression and/or abnormal cleavage of amyloid precursor protein (APP) are linked to Alzheimer's disease (AD) development and progression. However, the molecular mechanisms regulating cellular levels of APP or its processing, and the physiological and pathological consequences of altered processing are not well understood. Here, using mouse and human cells, we found that neuronal damage induced by UV irradiation leads to specific APP, APLP1, and APLP2 decline by accelerating their secretase-dependent processing. Pharmacological inhibition of endosomal/lysosomal activity partially protects UV-induced APP processing implying contribution of the endosomal and/or lysosomal compartments in this process. We found that a biological consequence of UV-induced γ-secretase processing of APP is impairment of APP axonal transport. To probe the functional consequences of impaired APP axonal transport, we isolated and analyzed presumptive APP-containing axonal transport vesicles from mouse cortical synaptosomes using electron microscopy, biochemical, and mass spectrometry analyses. We identified a population of morphologically heterogeneous organelles that contains APP, the secretase machinery, molecular motors, and previously proposed and new residents of APP vesicles. These possible cargoes are enriched in proteins whose dysfunction could contribute to neuronal malfunction and diseases of the nervous system including AD. Together, these results suggest that damage-induced APP processing might impair APP axonal transport, which could result in failure of synaptic maintenance and neuronal dysfunction.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Transporte Axonal/efeitos da radiação , Axônios/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Neurônios/citologia , Raios Ultravioleta , Precursor de Proteína beta-Amiloide/deficiência , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Axônios/ultraestrutura , Células Cultivadas , Embrião de Mamíferos , Hipocampo/citologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuroblastoma/patologia , Neurônios/efeitos da radiação , Presenilina-1/deficiência , Presenilina-2/deficiência , Transfecção
6.
Nat Genet ; 38(6): 674-81, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16682973

RESUMO

The molecular basis of nephronophthisis, the most frequent genetic cause of renal failure in children and young adults, and its association with retinal degeneration and cerebellar vermis aplasia in Joubert syndrome are poorly understood. Using positional cloning, we here identify mutations in the gene CEP290 as causing nephronophthisis. It encodes a protein with several domains also present in CENPF, a protein involved in chromosome segregation. CEP290 (also known as NPHP6) interacts with and modulates the activity of ATF4, a transcription factor implicated in cAMP-dependent renal cyst formation. NPHP6 is found at centrosomes and in the nucleus of renal epithelial cells in a cell cycle-dependent manner and in connecting cilia of photoreceptors. Abrogation of its function in zebrafish recapitulates the renal, retinal and cerebellar phenotypes of Joubert syndrome. Our findings help establish the link between centrosome function, tissue architecture and transcriptional control in the pathogenesis of cystic kidney disease, retinal degeneration, and central nervous system development.


Assuntos
Fator 4 Ativador da Transcrição/genética , Antígenos de Neoplasias/genética , Mutação , Proteínas de Neoplasias/genética , Animais , Proteínas de Ciclo Celular , Proteínas do Citoesqueleto , Feminino , Ligação Genética , Humanos , Hibridização In Situ , Masculino , Linhagem , Síndrome , Peixe-Zebra
7.
Nat Genet ; 37(3): 282-8, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15723066

RESUMO

Nephronophthisis (NPHP) is the most frequent genetic cause of chronic renal failure in children. Identification of four genes mutated in NPHP subtypes 1-4 (refs. 4-9) has linked the pathogenesis of NPHP to ciliary functions. Ten percent of affected individuals have retinitis pigmentosa, constituting the renal-retinal Senior-Loken syndrome (SLSN). Here we identify, by positional cloning, mutations in an evolutionarily conserved gene, IQCB1 (also called NPHP5), as the most frequent cause of SLSN. IQCB1 encodes an IQ-domain protein, nephrocystin-5. All individuals with IQCB1 mutations have retinitis pigmentosa. Hence, we examined the interaction of nephrocystin-5 with RPGR (retinitis pigmentosa GTPase regulator), which is expressed in photoreceptor cilia and associated with 10-20% of retinitis pigmentosa. We show that nephrocystin-5, RPGR and calmodulin can be coimmunoprecipitated from retinal extracts, and that these proteins localize to connecting cilia of photoreceptors and to primary cilia of renal epithelial cells. Our studies emphasize the central role of ciliary dysfunction in the pathogenesis of SLSN.


Assuntos
Proteínas de Ligação a Calmodulina/genética , Proteínas de Ligação a Calmodulina/metabolismo , Calmodulina/metabolismo , Proteínas do Olho/metabolismo , Mutação , Sequência de Aminoácidos , Northern Blotting , Proteínas de Ligação a Calmodulina/química , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Linhagem , Síndrome , Técnicas do Sistema de Duplo-Híbrido
8.
Hum Mol Genet ; 20(13): 2560-70, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21493626

RESUMO

Mutations in the MYO7A gene cause a deaf-blindness disorder, known as Usher syndrome 1B.  In the retina, the majority of MYO7A is in the retinal pigmented epithelium (RPE), where many of the reactions of the visual retinoid cycle take place.  We have observed that the retinas of Myo7a-mutant mice are resistant to acute light damage. In exploring the basis of this resistance, we found that Myo7a-mutant mice have lower levels of RPE65, the RPE isomerase that has a key role in the retinoid cycle.  We show for the first time that RPE65 normally undergoes a light-dependent translocation to become more concentrated in the central region of the RPE cells.  This translocation requires MYO7A, so that, in Myo7a-mutant mice, RPE65 is partly mislocalized in the light.  RPE65 is degraded more quickly in Myo7a-mutant mice, perhaps due to its mislocalization, providing a plausible explanation for its lower levels.  Following a 50-60% photobleach, Myo7a-mutant retinas exhibited increased all-trans-retinyl ester levels during the initial stages of dark recovery, consistent with a deficiency in RPE65 activity.  Lastly, MYO7A and RPE65 were co-immunoprecipitated from RPE cell lysate by antibodies against either of the proteins, and the two proteins were partly colocalized, suggesting a direct or indirect interaction.  Together, the results support a role for MYO7A in the translocation of RPE65, illustrating the involvement of a molecular motor in the spatiotemporal organization of the retinoid cycle in vision.


Assuntos
Proteínas do Olho/metabolismo , Miosinas/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Proteínas do Olho/genética , Humanos , Espaço Intracelular/metabolismo , Luz/efeitos adversos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miosina VIIa , Miosinas/genética , Ligação Proteica/fisiologia , Transporte Proteico/genética , Transporte Proteico/efeitos da radiação , Retina/metabolismo , Retina/patologia , Retina/efeitos da radiação , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Síndromes de Usher/genética , Síndromes de Usher/metabolismo , Síndromes de Usher/patologia
9.
Cells ; 12(21)2023 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-37947653

RESUMO

Using constitutive GRF1/2 knockout mice, we showed previously that GRF2 is a key regulator of nuclear migration in retinal cone photoreceptors. To evaluate the functional relevance of that cellular process for two putative targets of the GEF activity of GRF2 (RAC1 and CDC42), here we compared the structural and functional retinal phenotypes resulting from conditional targeting of RAC1 or CDC42 in the cone photoreceptors of constitutive GRF2KO and GRF2WT mice. We observed that single RAC1 disruption did not cause any obvious morphological or physiological changes in the retinas of GRF2WT mice, and did not modify either the phenotypic alterations previously described in the retinal photoreceptor layer of GRF2KO mice. In contrast, the single ablation of CDC42 in the cone photoreceptors of GRF2WT mice resulted in clear alterations of nuclear movement that, unlike those of the GRF2KO retinas, were not accompanied by electrophysiological defects or slow, progressive cone cell degeneration. On the other hand, the concomitant disruption of GRF2 and CDC42 in the cone photoreceptors resulted, somewhat surprisingly, in a normalized pattern of nuclear positioning/movement, similar to that physiologically observed in GRF2WT mice, along with worsened patterns of electrophysiological responses and faster rates of cell death/disappearance than those previously recorded in single GRF2KO cone cells. Interestingly, the increased rates of cone cell apoptosis/death observed in single GRF2KO and double-knockout GRF2KO/CDC42KO retinas correlated with the electron microscopic detection of significant ultrastructural alterations (flattening) of their retinal ribbon synapses that were not otherwise observed at all in single-knockout CDC42KO retinas. Our observations identify GRF2 and CDC42 (but not RAC1) as key regulators of retinal processes controlling cone photoreceptor nuclear positioning and survival, and support the notion of GRF2 loss-of-function mutations as potential drivers of cone retinal dystrophies.


Assuntos
Fator 2 de Liberação do Nucleotídeo Guanina , Células Fotorreceptoras Retinianas Cones , Animais , Camundongos , Camundongos Knockout , Retina , Células Fotorreceptoras Retinianas Cones/ultraestrutura , Sinapses/ultraestrutura
10.
Cells ; 11(2)2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35053342

RESUMO

The insulin-degrading enzyme (IDE) is a zinc-dependent metalloendopeptidase that belongs to the M16A metalloprotease family. IDE is markedly expressed in the brain, where it is particularly relevant due to its in vitro amyloid beta (Aß)-degrading activity. The subcellular localization of IDE, a paramount aspect to understand how this enzyme can perform its proteolytic functions in vivo, remains highly controversial. In this work, we addressed IDE subcellular localization from an evolutionary perspective. Phylogenetic analyses based on protein sequence and gene and protein structure were performed. An in silico analysis of IDE signal peptide suggests an evolutionary shift in IDE exportation at the prokaryote/eukaryote divide. Subcellular localization experiments in microglia revealed that IDE is mostly cytosolic. Furthermore, IDE associates to membranes by their cytoplasmatic side and further partitions between raft and non-raft domains. When stimulated, microglia change into a secretory active state, produces numerous multivesicular bodies and IDE associates with their membranes. The subsequent inward budding of such membranes internalizes IDE in intraluminal vesicles, which later allows IDE to be exported outside the cells in small extracellular vesicles. We further demonstrate that such an IDE exportation mechanism is regulated by stimuli relevant for microglia in physiological conditions and upon aging and neurodegeneration.


Assuntos
Evolução Molecular , Insulisina/metabolismo , Microglia/enzimologia , Animais , Linhagem Celular , Células Cultivadas , Sequência Conservada , Citosol/metabolismo , Vesículas Extracelulares/metabolismo , Insulisina/ultraestrutura , Microdomínios da Membrana/metabolismo , Metaloendopeptidases/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/ultraestrutura , Corpos Multivesiculares/metabolismo , Filogenia , Frações Subcelulares/metabolismo
11.
Front Neuroanat ; 16: 983151, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36213611

RESUMO

Apicobasal polarity is a hallmark of retinal pigment epithelium cells and is required to perform their functions; however, the precise roles of the different proteins that execute polarity are still poorly understood. Here, we have studied the expression and location of Scribble, the core member of the polarity basal protein complex in epithelial-derived cells, in human and mouse RPE cells in both control and pathological conditions. We found that Scribble specifically localizes at the basolateral membrane of mouse and human RPE cells. In addition, we observed an increase in the expression of Scribble during human RPE development in culture, while it acquires a well-defined basolateral pattern as this process is completed. Finally, the expression and location of Scribble were analyzed in human RPE cells in experimental conditions that mimic the toxic environment suffered by these cells during AMD development and found an increase in Scribble expression in cells that develop a pathological phenotype, suggesting that the protein could be altered in cells under stress conditions, as occurs in AMD. Together, our results demonstrate, for the first time, that Scribble is expressed in both human and mouse RPE and is localized at the basolateral membrane in mature cells. Furthermore, Scribble shows impaired expression and location in RPE cells in pathological conditions, suggesting a possible role for this protein in the development of pathologies, such as AMD.

12.
Cell Death Dis ; 13(4): 383, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35444190

RESUMO

Synaptic loss, neuronal death, and circuit remodeling are common features of central nervous system neurodegenerative disorders. Retinitis pigmentosa (RP), the leading cause of inherited blindness, is a group of retinal dystrophies characterized by photoreceptor dysfunction and death. The insulin receptor, a key controller of metabolism, also regulates neuronal survival and synaptic formation, maintenance, and activity. Indeed, deficient insulin receptor signaling has been implicated in several brain neurodegenerative pathologies. We present evidence linking impaired insulin receptor signaling with RP. We describe a selective decrease in the levels of the insulin receptor and its downstream effector phospho-S6 in retinal horizontal cell terminals in the rd10 mouse model of RP, as well as aberrant synapses between rod photoreceptors and the postsynaptic terminals of horizontal and bipolar cells. A gene therapy strategy to induce sustained proinsulin, the insulin precursor, production restored retinal insulin receptor signaling, by increasing S6 phosphorylation, without peripheral metabolic consequences. Moreover, proinsulin preserved photoreceptor synaptic connectivity and prolonged visual function in electroretinogram and optomotor tests. These findings point to a disease-modifying role of insulin receptor and support the therapeutic potential of proinsulin in retinitis pigmentosa.


Assuntos
Proinsulina , Retinose Pigmentar , Animais , Modelos Animais de Doenças , Insulina , Camundongos , Camundongos Endogâmicos C57BL , Proinsulina/farmacologia , Receptor de Insulina , Retinose Pigmentar/patologia , Sinapses/metabolismo
13.
J Clin Invest ; 118(8): 2908-16, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18654668

RESUMO

Familial macular degeneration is a clinically and genetically heterogeneous group of disorders characterized by progressive central vision loss. Here we show that an R373C missense mutation in the prominin 1 gene (PROM1) causes 3 forms of autosomal-dominant macular degeneration. In transgenic mice expressing R373C mutant human PROM1, both mutant and endogenous PROM1 were found throughout the layers of the photoreceptors, rather than at the base of the photoreceptor outer segments, where PROM1 is normally localized. Moreover, the outer segment disk membranes were greatly overgrown and misoriented, indicating defective disk morphogenesis. Immunoprecipitation studies showed that PROM1 interacted with protocadherin 21 (PCDH21), a photoreceptor-specific cadherin, and with actin filaments, both of which play critical roles in disk membrane morphogenesis. Collectively, our results identify what we believe to be a novel complex involved in photoreceptor disk morphogenesis and indicate a possible role for PROM1 and PCDH21 in macular degeneration.


Assuntos
Antígenos CD/genética , Glicoproteínas/genética , Degeneração Macular/genética , Mutação de Sentido Incorreto , Peptídeos/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Antígeno AC133 , Citoesqueleto de Actina/metabolismo , Animais , Antígenos CD/metabolismo , Proteínas Relacionadas a Caderinas , Caderinas/metabolismo , Eletrorretinografia , Glicoproteínas/metabolismo , Humanos , Degeneração Macular/fisiopatologia , Camundongos , Camundongos Transgênicos , Morfogênese , Proteínas do Tecido Nervoso/metabolismo , Peptídeos/metabolismo , Células Fotorreceptoras de Vertebrados/ultraestrutura
14.
Cancers (Basel) ; 13(17)2021 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-34503072

RESUMO

Autophagy is a physiological process by which various damaged or non-essential cytosolic components are recycled, contributing to cell survival under stress conditions. In cancer, autophagy can have antitumor or protumor effects depending on the developmental stage. Here, we use Western blotting, immunochemistry, and transmission electron microscopy to demonstrate that the antitumor peptide TAT-Cx43266-283, a c-Src inhibitor, blocks autophagic flux in glioblastoma stem cells (GSCs) under basal and nutrient-deprived conditions. Upon nutrient deprivation, GSCs acquired a dormant-like phenotype that was disrupted by inhibition of autophagy with TAT-Cx43266-283 or chloroquine (a classic autophagy inhibitor), leading to GSC death. Remarkably, dasatinib, a clinically available c-Src inhibitor, could not replicate TAT-Cx43266-283 effect on dormant GSCs, revealing for the first time the possible involvement of pathways other than c-Src in TAT-Cx43266-283 effect. TAT-Cx43266-283 exerts an antitumor effect both in nutrient-complete and nutrient-deprived environments, which constitutes an advantage over chloroquine and dasatinib, whose effects depend on nutrient environment. Finally, our analysis of the levels of autophagy-related proteins in healthy and glioma donors suggests that autophagy is upregulated in glioblastoma, further supporting the interest in inhibiting this process in the most aggressive brain tumor and the potential use of TAT-Cx43266-283 as a therapy for this type of cancer.

15.
Front Cell Dev Biol ; 9: 701853, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34262913

RESUMO

Apicobasal polarity is essential for epithelial cell function, yet the roles of different proteins in its completion is not fully understood. Here, we have studied the role of the polarity protein, CRB2, in human retinal pigment epithelial (RPE) cells during polarization in vitro, and in mature murine RPE cells in vivo. After establishing a simplified protocol for the culture of human fetal RPE cells, we studied the temporal sequence of the expression and localization of polarity and cell junction proteins during polarization in these epithelial cells. We found that CRB2 plays a key role in tight junction maintenance as well as in cell cycle arrest. In addition, our studies in vivo show that the knockdown of CRB2 in the RPE affects to the distribution of different apical polarity proteins and results in perturbed retinal homeostasis, manifested by the invasion of activated microglial cells into the subretinal space. Together our results demonstrate that CRB2 is a key protein for the development and maintenance of a polarized epithelium.

16.
J Neurosci ; 29(18): 5758-67, 2009 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-19420244

RESUMO

Many neurodegenerative diseases exhibit axonal pathology, transport defects, and aberrant phosphorylation and aggregation of the microtubule binding protein tau. While mutant tau protein in frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP17) causes aberrant microtubule binding and assembly of tau into filaments, the pathways leading to tau-mediated neurotoxicity in Alzheimer's disease and other neurodegenerative disorders in which tau protein is not genetically modified remain unknown. To test the hypothesis that axonal transport defects alone can cause pathological abnormalities in tau protein and neurodegeneration in the absence of mutant tau or amyloid beta deposits, we induced transport defects by deletion of the kinesin light chain 1 (KLC1) subunit of the anterograde motor kinesin-1. We found that upon aging, early selective axonal transport defects in mice lacking the KLC1 protein (KLC1-/-) led to axonopathies with cytoskeletal disorganization and abnormal cargo accumulation. In addition, increased c-jun N-terminal stress kinase activation colocalized with aberrant tau in dystrophic axons. Surprisingly, swollen dystrophic axons exhibited abnormal tau hyperphosphorylation and accumulation. Thus, directly interfering with axonal transport is sufficient to activate stress kinase pathways initiating a biochemical cascade that drives normal tau protein into a pathological state found in a variety of neurodegenerative disorders including Alzheimer's disease.


Assuntos
Axônios/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/citologia , Estresse Fisiológico/fisiologia , Proteínas tau/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fatores Etários , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Animais Recém-Nascidos , Axônios/ultraestrutura , Células Cultivadas , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Proteínas de Fluorescência Verde/genética , Hipocampo/citologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Cinesinas , Quimografia/métodos , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Varredura/métodos , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Neurofilamentos/metabolismo , Neurônios/ultraestrutura , Organelas/metabolismo , Organelas/ultraestrutura , Transporte Proteico/genética , Estatísticas não Paramétricas , Transfecção/métodos , Proteínas tau/genética
17.
J Neurosci ; 29(50): 15810-8, 2009 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-20016096

RESUMO

Mutations in the head and tail domains of the motor protein myosin VIIA (MYO7A) cause deaf-blindness (Usher syndrome type 1B, USH1B) and nonsyndromic deafness (DFNB2, DFNA11). The head domain binds to F-actin and serves as the MYO7A motor domain, but little is known about the function of the tail domain. In a genetic screen, we have identified polka mice, which carry a mutation (c.5742 + 5G > A) that affects splicing of the MYO7A transcript and truncates the MYO7A tail domain at the C-terminal FERM domain. In the inner ear, expression of the truncated MYO7A protein is severely reduced, leading to defects in hair cell development. In retinal pigment epithelial (RPE) cells, the truncated MYO7A protein is expressed at comparative levels to wild-type protein but fails to associate with and transport melanosomes. We conclude that the C-terminal FERM domain of MYO7A is critical for melanosome transport in RPE cells. Our findings also suggest that MYO7A mutations can lead to tissue-specific effects on protein levels, which may explain why some mutations in MYO7A lead to deafness without retinal impairment.


Assuntos
Alelos , Proteínas do Citoesqueleto/genética , Melanossomas/metabolismo , Miosinas/genética , Epitélio Pigmentado da Retina/metabolismo , Sequência de Aminoácidos , Animais , Percepção Auditiva/genética , Transporte Biológico/genética , Melanossomas/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes Neurológicos , Dados de Sequência Molecular , Miosina VIIa , Estrutura Terciária de Proteína/genética , Epitélio Pigmentado da Retina/citologia , Síndromes de Usher/genética , Síndromes de Usher/metabolismo
18.
J Biol Chem ; 284(50): 34628-39, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-19801628

RESUMO

The extreme polarized morphology of neurons poses a challenging problem for intracellular trafficking pathways. The distant synaptic terminals must communicate via axonal transport with the cell soma for neuronal survival, function, and repair. Multiple classes of organelles transported along axons may establish and maintain the polarized morphology of neurons, as well as control signaling and neuronal responses to extracellular cues such as neurotrophic or stress factors. We reported previously that the motor-binding protein Sunday Driver (syd), also known as JIP3 or JSAP1, links vesicular axonal transport to injury signaling. To better understand syd function in axonal transport and in the response of neurons to injury, we developed a purification strategy based on anti-syd antibodies conjugated to magnetic beads to identify syd-associated axonal vesicles. Electron microscopy analyses revealed two classes of syd-associated vesicles of distinct morphology. To identify the molecular anatomy of syd vesicles, we determined their protein composition by mass spectrometry. Gene Ontology analyses of each vesicle protein content revealed their unique identity and indicated that one class of syd vesicles belongs to the endocytic pathway, whereas another may belong to an anterogradely transported vesicle pool. To validate these findings, we examined the transport and localization of components of syd vesicles within axons of mouse sciatic nerve. Together, our results lead us to propose that endocytic syd vesicles function in part to carry injury signals back to the cell body, whereas anterograde syd vesicles may play a role in axonal outgrowth and guidance.


Assuntos
Transporte Axonal/fisiologia , Axônios/metabolismo , Axônios/ultraestrutura , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Organelas/metabolismo , Sinaptossomos , Vesículas Transportadoras , Proteínas Adaptadoras de Transdução de Sinal , Animais , Axônios/patologia , Endocitose/fisiologia , Endossomos/metabolismo , Endossomos/ultraestrutura , Feminino , Separação Imunomagnética , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Motores Moleculares/metabolismo , Proteínas do Tecido Nervoso , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Nervo Isquiático/citologia , Nervo Isquiático/patologia , Transdução de Sinais/fisiologia , Sinaptossomos/metabolismo , Sinaptossomos/ultraestrutura , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/ultraestrutura
19.
Hum Mol Genet ; 17(22): 3474-86, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18694898

RESUMO

Overexpression of amyloid precursor protein (APP), as well as mutations in the APP and presenilin genes, causes rare forms of Alzheimer's disease (AD). These genetic changes have been proposed to cause AD by elevating levels of amyloid-beta peptides (Abeta), which are thought to be neurotoxic. Since overexpression of APP also causes defects in axonal transport, we tested whether defects in axonal transport were the result of Abeta poisoning of the axonal transport machinery. Because directly varying APP levels also alters APP domains in addition to Abeta, we perturbed Abeta generation selectively by combining APP transgenes in Drosophila and mice with presenilin-1 (PS1) transgenes harboring mutations that cause familial AD (FAD). We found that combining FAD mutant PS1 with FAD mutant APP increased Abeta42/Abeta40 ratios and enhanced amyloid deposition as previously reported. Surprisingly, however, this combination suppressed rather than increased APP-induced axonal transport defects in both Drosophila and mice. In addition, neuronal apoptosis induced by expression of FAD mutant human APP in Drosophila was suppressed by co-expressing FAD mutant PS1. We also observed that directly elevating Abeta with fusions to the Familial British and Danish Dementia-related BRI protein did not enhance axonal transport phenotypes in APP transgenic mice. Finally, we observed that perturbing Abeta ratios in the mouse by combining FAD mutant PS1 with FAD mutant APP did not enhance APP-induced behavioral defects. A potential mechanism to explain these findings was suggested by direct analysis of axonal transport in the mouse, which revealed that axonal transport or entry of APP into axons is reduced by FAD mutant PS1. Thus, we suggest that APP-induced axonal defects are not caused by Abeta.


Assuntos
Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Transporte Axonal , Axônios/metabolismo , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Axônios/patologia , Comportamento Animal , Cérebro/metabolismo , Drosophila , Medo , Feminino , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Presenilinas/genética , Presenilinas/metabolismo , Transgenes
20.
Exp Eye Res ; 90(2): 267-76, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19895810

RESUMO

The transcription factor Prox1 acts in rodent retinogenesis, at least in promoting cell cycle withdrawal and horizontal cell production. In the mature retina, this protein is detected at the inner nuclear layer of all vertebrate groups. We have made a neurochemical characterisation of Prox1(+) cell types in two different vertebrate groups: mammals and fish. As well as Prox1(+) horizontal cells, we have observed Prox1(+)/PKC-alpha(+) rod bipolar cells in mouse and cone ON and mixed b bipolar cells in goldfish. In mouse, only some CB(+) and CR(+) amacrine cells are Prox1(+) and the TH(+) and CR(+) amacrine cells are Prox1(-). However, in goldfish all CR(+) amacrine cells and TH(+) interplexiform cells are Prox1(+) and in the GCL displaced amacrine cells are also Prox1(+). Besides its expression in different interneuron subpopulations, we demonstrate, for the first time, the presence of Prox1 in the GS(+) and CRALBP(+) Müller cells in the retina of adult mammals and in developing and mature retina of fish. The presence of Prox1 in these cells appears to be related to survival or maintenance of their phenotype. We also demonstrate that in fish, where retinal formation persists into adulthood, Prox1 is expressed in dividing PCNA(+) cells at the peripheral growing zone, in rod progenitors at the inner and outer nuclear layers as well as in early progenitors during a retinal regeneration process after cryo-lesion of the peripheral growing zone. Therefore, Prox1 functions in vertebrate retinogenesis may be more complex than previously expected.


Assuntos
Células Amácrinas/metabolismo , Proteínas de Homeodomínio/metabolismo , Neuroglia/metabolismo , Células Bipolares da Retina/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células-Tronco/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Contagem de Células , Técnica Indireta de Fluorescência para Anticorpo , Carpa Dourada , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos C57BL , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA