Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 18(12): 1342-1352, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29058703

RESUMEN

T cells reorganize their metabolic profiles after being activated, but the systemic metabolic effect of sustained activation of the immune system has remained unexplored. Here we report that augmented T cell responses in Pdcd1-/- mice, which lack the inhibitory receptor PD-1, induced a metabolic serum signature characterized by depletion of amino acids. We found that the depletion of amino acids in serum was due to the accumulation of amino acids in activated Pdcd1-/- T cells in the lymph nodes. A systemic decrease in tryptophan and tyrosine led to substantial deficiency in the neurotransmitters serotonin and dopamine in the brain, which resulted in behavioral changes dominated by anxiety-like behavior and exacerbated fear responses. Together these data indicate that excessive activation of T cells causes a systemic metabolomic shift with consequences that extend beyond the immune system.


Asunto(s)
Ansiedad/fisiopatología , Conducta Animal/fisiología , Miedo/fisiología , Activación de Linfocitos/inmunología , Receptor de Muerte Celular Programada 1/genética , Linfocitos T/inmunología , Aminoácidos/sangre , Animales , Encéfalo/metabolismo , Dopamina/deficiencia , Interferón gamma/sangre , Quinurenina/sangre , Ganglios Linfáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Muerte Celular Programada 1/deficiencia , Serotonina/deficiencia , Linfocitos T/metabolismo , Triptófano/metabolismo , Tirosina/metabolismo
2.
J Biol Chem ; 299(12): 105461, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37977220

RESUMEN

Müller glial cells, which are the most predominant glial subtype in the retina, play multiple important roles, including the maintenance of structural integrity, homeostasis, and physiological functions of the retina. We have previously found that the Rax homeoprotein is expressed in postnatal and mature Müller glial cells in the mouse retina. However, the function of Rax in postnatal and mature Müller glial cells remains to be elucidated. In the current study, we first investigated Rax function in retinal development using retroviral lineage analysis and found that Rax controls the specification of late-born retinal cell types, including Müller glial cells in the postnatal retina. We next generated Rax tamoxifen-induced conditional KO (Rax iCKO) mice, where Rax can be depleted in mTFP-labeled Müller glial cells upon tamoxifen treatment, by crossing Raxflox/flox mice with Rlbp1-CreERT2 mice, which we have produced. Immunohistochemical analysis showed a characteristic of reactive gliosis and enhanced gliosis of Müller glial cells in Rax iCKO retinas under normal and stress conditions, respectively. We performed RNA-seq analysis on mTFP-positive cells purified from the Rax iCKO retina and found significantly reduced expression of suppressor of cytokinesignaling-3 (Socs3). Reporter gene assays showed that Rax directly transactivates the Socs3 promoter. We observed decreased expression of Socs3 in Müller glial cells of Rax iCKO retinas by immunostaining. Taken together, the present results suggest that Rax suppresses inflammation in Müller glial cells by transactivating Socs3. This study sheds light on the transcriptional regulatory mechanisms underlying retinal Müller glial cell homeostasis.


Asunto(s)
Células Ependimogliales , Proteínas del Ojo , Proteínas de Homeodominio , Homeostasis , Retina , Factores de Transcripción , Animales , Ratones , Células Ependimogliales/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Gliosis/genética , Gliosis/metabolismo , Gliosis/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Homeostasis/genética , Retina/citología , Retina/crecimiento & desarrollo , Retina/metabolismo , Retina/patología , RNA-Seq , Tamoxifeno/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional
3.
Hum Mol Genet ; 31(4): 535-547, 2022 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-34508581

RESUMEN

Intellectual disability (ID) is a neurodevelopmental disorder affecting approximately 0.5-3% of the population in the developed world. Individuals with ID exhibit deficits in intelligence, impaired adaptive behavior and often visual impairments. Cytoplasmic fragile X mental retardation 1 (FMR1)-interacting protein 2 (CYFIP2) is an interacting partner of the FMR protein, whose loss results in fragile X syndrome, the most common inherited cause of ID. Recently, CYFIP2 variants have been found in patients with early-onset epileptic encephalopathy, developmental delay and ID. Such individuals often exhibit visual impairments; however, the underlying mechanism is poorly understood. In the present study, we investigated the role of Cyfip2 in retinal and visual functions by generating and analyzing Cyfip2 conditional knockout (CKO) mice. While we found no major differences in the layer structures and cell compositions between the control and Cyfip2 CKO retinas, a subset of genes associated with the transporter and channel activities was differentially expressed in Cyfip2 CKO retinas than in the controls. Multi-electrode array recordings showed more sustained and stronger responses to positive flashes of the ON ganglion cells in the Cyfip2 CKO retina than in the controls, although electroretinogram analysis revealed that Cyfip2 deficiency unaffected the photoreceptor and ON bipolar cell functions. Furthermore, analysis of initial and late phase optokinetic responses demonstrated that Cyfip2 deficiency impaired the visual function at the organismal level. Together, our results shed light on the molecular mechanism underlying the visual impairments observed in individuals with CYFIP2 variants and, more generally, in patients with neurodevelopmental disorders, including ID.


Asunto(s)
Síndrome del Cromosoma X Frágil , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Humanos , Discapacidad Intelectual/genética , Ratones , Trastornos del Neurodesarrollo/genética , Células Ganglionares de la Retina/metabolismo , Agudeza Visual
4.
J Biol Chem ; 298(3): 101686, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35131266

RESUMEN

In humans, ciliary dysfunction causes ciliopathies, which present as multiple organ defects, including developmental and sensory abnormalities. Sdccag8 is a centrosomal/basal body protein essential for proper cilia formation. Gene mutations in SDCCAG8 have been found in patients with ciliopathies manifesting a broad spectrum of symptoms, including hypogonadism. Among these mutations, several that are predicted to truncate the SDCCAG8 carboxyl (C) terminus are also associated with such symptoms; however, the underlying mechanisms are poorly understood. In the present study, we identified the Sdccag8 C-terminal region (Sdccag8-C) as a module that interacts with the ciliopathy proteins, Ick/Cilk1 and Mak, which were shown to be essential for the regulation of ciliary protein trafficking and cilia length in mammals in our previous studies. We found that Sdccag8-C is essential for Sdccag8 localization to centrosomes and cilia formation in cultured cells. We then generated a mouse mutant in which Sdccag8-C was truncated (Sdccag8ΔC/ΔC mice) using a CRISPR-mediated stop codon knock-in strategy. In Sdccag8ΔC/ΔC mice, we observed abnormalities in cilia formation and ciliopathy-like organ phenotypes, including cleft palate, polydactyly, retinal degeneration, and cystic kidney, which partially overlapped with those previously observed in Ick- and Mak-deficient mice. Furthermore, Sdccag8ΔC/ΔC mice exhibited a defect in spermatogenesis, which was a previously uncharacterized phenotype of Sdccag8 dysfunction. Together, these results shed light on the molecular and pathological mechanisms underlying ciliopathies observed in patients with SDCCAG8 mutations and may advance our understanding of protein-protein interaction networks involved in cilia development.


Asunto(s)
Autoantígenos , Ciliopatías , Enfermedades Renales Quísticas , Proteínas de Neoplasias , Animales , Autoantígenos/metabolismo , Cuerpos Basales , Cilios/metabolismo , Ciliopatías/genética , Ciliopatías/metabolismo , Femenino , Homeostasis , Humanos , Enfermedades Renales Quísticas/metabolismo , Masculino , Mamíferos , Ratones , Mutación , Proteínas de Neoplasias/metabolismo , Proteínas/metabolismo
5.
J Biol Chem ; 298(9): 102293, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35868558

RESUMEN

MicroRNA-124a (miR-124a) is one of the most abundantly expressed microRNAs in the central nervous system and is encoded in mammals by the three genomic loci miR-124a-1/2/3; however, its in vivo roles in neuronal development and function remain ambiguous. In the present study, we investigated the effect of miR-124a loss on neuronal differentiation in mice and in embryonic stem (ES) cells. Since miR-124a-3 exhibits only background expression levels in the brain and we were unable to obtain miR-124a-1/2/3 triple knockout (TKO) mice by mating, we generated and analyzed miR-124a-1/2 double knockout (DKO) mice. We found that these DKO mice exhibit perinatal lethality. RNA-seq analysis demonstrated that the expression levels of proneural and neuronal marker genes were almost unchanged between the control and miR-124a-1/2 DKO brains; however, genes related to neuronal synaptic formation and function were enriched among downregulated genes in the miR-124a-1/2 DKO brain. In addition, we found the transcription regulator Tardbp/TDP-43, loss of which leads to defects in neuronal maturation and function, was inactivated in the miR-124a-1/2 DKO brain. Furthermore, Tardbp knockdown suppressed neurite extension in cultured neuronal cells. We also generated miR-124a-1/2/3 TKO ES cells using CRISPR-Cas9 as an alternative to TKO mice. Phase-contrast microscopic, immunocytochemical, and gene expression analyses showed that miR-124a-1/2/3 TKO ES cell lines were able to differentiate into neurons. Collectively, these results suggest that miR-124a plays a role in neuronal maturation rather than neurogenesis in vivo and advance our understanding of the functional roles of microRNAs in central nervous system development.


Asunto(s)
Proteínas de Unión al ADN , MicroARNs , Neurogénesis , Neuronas , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ratones , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Embrionarias de Ratones , Neurogénesis/genética , Neuronas/citología , Neuronas/metabolismo
6.
EMBO J ; 38(23): e101409, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31696965

RESUMEN

Adaptation is a general feature of sensory systems. In rod photoreceptors, light-dependent transducin translocation and Ca2+ homeostasis are involved in light/dark adaptation and prevention of cell damage by light. However, the underlying regulatory mechanisms remain unclear. Here, we identify mammalian Cul3-Klhl18 ubiquitin ligase as a transducin translocation modulator during light/dark adaptation. Under dark conditions, Klhl18-/- mice exhibited decreased rod light responses and subcellular localization of the transducin α-subunit (Tα), similar to that observed in light-adapted Klhl18+/+ mice. Cul3-Klhl18 promoted ubiquitination and degradation of Unc119, a rod Tα-interacting protein. Unc119 overexpression phenocopied Tα mislocalization observed in Klhl18-/- mice. Klhl18 weakly recognized casein kinase-2-phosphorylated Unc119 protein, which is dephosphorylated by Ca2+ -dependent phosphatase calcineurin. Calcineurin inhibition increased Unc119 expression and Tα mislocalization in rods. These results suggest that Cul3-Klhl18 modulates rod Tα translocation during light/dark adaptation through Unc119 ubiquitination, which is affected by phosphorylation. Notably, inactivation of the Cul3-Klhl18 ligase and calcineurin inhibitors FK506 and cyclosporine A that are known immunosuppressant drugs repressed light-induced photoreceptor damage, suggesting potential therapeutic targets.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cullin/fisiología , Adaptación a la Oscuridad , Luz , Células Fotorreceptoras Retinianas Bastones/metabolismo , Transducina/metabolismo , Ubiquitina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular/genética , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Células Fotorreceptoras de Vertebrados/metabolismo , Transporte de Proteínas , Retina/lesiones , Retina/metabolismo , Retina/patología , Transducina/genética
7.
J Biol Chem ; 295(38): 13363-13376, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32732286

RESUMEN

ICK (also known as CILK1) is a mitogen-activated protein kinase-like kinase localized at the ciliary tip. Its deficiency is known to result in the elongation of cilia and causes ciliopathies in humans. However, little is known about how ICK is transported to the ciliary tip. We here show that the C-terminal noncatalytic region of ICK interacts with the intraflagellar transport (IFT)-B complex of the IFT machinery and participates in its transport to the ciliary tip. Furthermore, total internal reflection fluorescence microscopy demonstrated that ICK undergoes bidirectional movement within cilia, similarly to IFT particles. Analysis of ICK knockout cells demonstrated that ICK deficiency severely impairs the retrograde trafficking of IFT particles and ciliary G protein-coupled receptors. In addition, we found that in ICK knockout cells, ciliary proteins are accumulated at the bulged ciliary tip, which appeared to be torn off and released into the environment as an extracellular vesicle. The exogenous expression of various ICK constructs in ICK knockout cells indicated that the IFT-dependent transport of ICK, as well as its kinase activity and phosphorylation at the canonical TDY motif, is essential for ICK function. Thus, we unequivocally show that ICK transported to the ciliary tip is required for retrograde ciliary protein trafficking and consequently for normal ciliary function.


Asunto(s)
Cilios/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencias de Aminoácidos , Cilios/genética , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Dominios Proteicos , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas
8.
Gastroenterology ; 158(6): 1650-1666.e15, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32032583

RESUMEN

BACKGROUND & AIMS: Gastric chief cells, a mature cell type that secretes digestive enzymes, have been proposed to be the origin of metaplasia and cancer through dedifferentiation or transdifferentiation. However, studies supporting this claim have had technical limitations, including issues with the specificity of chief cell markers and the toxicity of drugs used. We therefore sought to identify genes expressed specifically in chief cells and establish a model to trace these cells. METHODS: We performed transcriptome analysis of Mist1-CreERT-traced cells, with or without chief cell depletion. Gpr30-rtTA mice were generated and crossed to TetO-Cre mice, and lineage tracing was performed after crosses to R26-TdTomato mice. Additional lineage tracing experiments were performed using Mist1-CreERT, Kitl-CreERT, Tff1-Cre, and Tff2-Cre mice crossed to reporter mice. Mice were given high-dose tamoxifen or DMP-777 or were infected with Helicobacter pylori to induce gastric metaplasia. We studied mice that expressed mutant forms of Ras in gastric cells, using TetO-KrasG12D, LSL-KrasG12D, and LSL-HrasG12V mice. We analyzed stomach tissues from GPR30-knockout mice. Mice were given dichloroacetate to inhibit pyruvate dehydrogenase kinase (PDK)-dependent cell competition. RESULTS: We identified GPR30, the G-protein-coupled form of the estrogen receptor, as a cell-specific marker of chief cells in gastric epithelium of mice. Gpr30-rtTA mice crossed to TetO-Cre;R26-TdTomato mice had specific expression of GPR30 in chief cells, with no expression noted in isthmus stem cells or lineage tracing of glands. Expression of mutant Kras in GPR30+ chief cells did not lead to the development of metaplasia or dysplasia but, instead, led to a reduction in labeled numbers of chief cells and a compensatory expansion of neck lineage, which was derived from upper Kitl+ clones. Administration of high-dose tamoxifen, DMP-777, or H pylori decreased the number of labeled chief cells. Chief cells were eliminated from epithelia via GPR30- and PDK-dependent cell competition after metaplastic stimuli, whereas loss of GRP30 or inhibition of PDK activity preserved chief cell numbers and attenuated neck lineage cell expansion. CONCLUSIONS: In tracing studies of mice, we found that most chief cells are lost during metaplasia and therefore are unlikely to contribute to gastric carcinogenesis. Expansion of cells that coexpress neck and chief lineage markers, known as spasmolytic polypeptide-expressing metaplasia, does not occur via dedifferentiation from chief cells but, rather, through a compensatory response from neck progenitors to replace the eliminated chief cells.


Asunto(s)
Células Principales Gástricas/fisiología , Mucosa Gástrica/patología , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Azetidinas/toxicidad , Comunicación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Ácido Dicloroacético/administración & dosificación , Modelos Animales de Enfermedad , Mucosa Gástrica/citología , Mucosa Gástrica/efectos de los fármacos , Infecciones por Helicobacter/microbiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Metaplasia/inducido químicamente , Metaplasia/microbiología , Metaplasia/patología , Ratones , Ratones Noqueados , Piperazinas/toxicidad , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/antagonistas & inhibidores , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética , Células Madre/fisiología , Tamoxifeno/toxicidad
9.
Exp Eye Res ; 212: 108770, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34562437

RESUMEN

PURPOSE: Cancer-associated retinal ON bipolar cell dysfunction (CARBD), which includes melanoma-associated retinopathy (MAR), has been reported to be caused by autoantibodies against the molecules expressed in ON bipolar cells, including TRPM1. The purpose of this study was to determine the antigenic regions of the autoantibodies against TRPM1 in the sera of CARBD patients, in whom we previously detected anti-TRPM1 autoantibodies. METHODS: The antigenic regions against TRPM1 in the sera of eight CARBD patients were examined by Western blots using HEK293T cells transfected with the plasmids expressing FLAG-tagged TRPM1 fragments. The clinical course of these patients was also documented. RESULTS: The clinical course differed among the patients. The electroretinograms (ERGs) and symptoms were improved in three patients, deteriorated in one patient, remained unchanged for a long time in one patient, and were not followable in three patients. Seven of the eight sera possessed multiple antigenic regions: two sera contained at least four antigen recognition regions, and three sera had at least three regions. The antigen regions were spread over the entire TRPM1 protein: five sera in the N-terminal intracellular domain, six sera in the transmembrane-containing region, and six sera in the C-terminal intracellular domain. No significant relationship was observed between the location of the antigen epitope and the patients' clinical course. CONCLUSIONS: The antigenic regions of anti-TRPM1 autoantibodies in CARBD patients were present not only in the N-terminal intracellular domain, which was reported in an earlier report, but also in the transmembrane-containing region and in the C-terminal intracellular domain. In addition, the antigenic regions for TRPM1 were found to vary among the CARBD patients examined, and most of the sera had multiple antigenic regions.


Asunto(s)
Autoanticuerpos/sangre , Síndromes Paraneoplásicos Oculares/inmunología , Células Bipolares de la Retina/metabolismo , Canales Catiónicos TRPM/inmunología , Anciano , Western Blotting , Electrorretinografía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Síndromes Paraneoplásicos Oculares/metabolismo , Síndromes Paraneoplásicos Oculares/patología , Células Bipolares de la Retina/patología , Estudios Retrospectivos , Células Tumorales Cultivadas
10.
Cell Mol Life Sci ; 77(7): 1251-1266, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31586239

RESUMEN

In vertebrate central nervous systems (CNSs), highly diverse neurons are selectively connected via synapses, which are essential for building an intricate neural network. The vertebrate retina is part of the CNS and is comprised of a distinct laminar organization, which serves as a good model system to study developmental synapse formation mechanisms. In the retina outer plexiform layer, rods and cones, two types of photoreceptor cells, elaborate selective synaptic contacts with ON- and/or OFF-bipolar cell terminals as well as with horizontal cell terminals. In the mouse retina, three photoreceptor subtypes and at least 15 bipolar subtypes exist. Previous and recent studies have significantly progressed our understanding of how selective synapse formation, between specific subtypes of photoreceptor and bipolar cells, is designed at the molecular level. In the ON pathway, photoreceptor-derived secreted and transmembrane proteins directly interact in trans with the GRM6 (mGluR6) complex, which is localized to ON-bipolar cell dendritic terminals, leading to selective synapse formation. Here, we review our current understanding of the key factors and mechanisms underlying selective synapse formation of photoreceptor cells with bipolar and horizontal cells in the retina. In addition, we describe how defects/mutations of the molecules involved in photoreceptor synapse formation are associated with human retinal diseases and visual disorders.


Asunto(s)
Neurogénesis , Células Fotorreceptoras de Vertebrados/fisiología , Sinapsis/fisiología , Animales , Dendritas/fisiología , Humanos , Modelos Biológicos , Enfermedades de la Retina/patología
11.
Eur J Neurosci ; 52(4): 3160-3165, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32027443

RESUMEN

Visual information in the retina is processed via two pathways: ON and OFF pathways that originate from ON and OFF bipolar cells. The differences in the receptors that mediate signal transmission from photoreceptors imply that the response speed to light signals differs between ON and OFF pathways. We studied the initial optokinetic responses (OKRs) of mice using two-frame motion stimuli presented with interstimulus intervals (ISIs) to understand functional difference of these pathways. When two successive image frames were presented with an ISI, observers often perceived motion in the opposite direction of the actual shift. This directional reversal results from the biphasic nature of the temporal filters in visual systems whose characteristics can be estimated from the dependence on ISIs. We examined the dependence on ISIs in the OKRs of TRPM1-/- mice, whose ON bipolar cells are dysfunctional, as well as in those of wild-type control mice. Wild type and TRPM1-/- mice showed comparable OKRs in the veridical direction when no ISI was present. Both types of mice showed OKRs that decreased and eventually reversed as the ISI increased, but with a directional reversal at a shorter ISI in TRPM1-/- than wild-type mice. In addition, the temporal filters of TRPM1-/- mice estimated from dependence on ISIs were tuned for higher frequencies, suggesting that compared with wild-type mice, the visual system of TRPM1-/- mice responds to light signals with faster dynamics. We conclude that the ON and OFF pathways contribute to initial OKRs by providing visual signals processed with different temporal resolutions.


Asunto(s)
Nistagmo Optoquinético , Retina , Animales , Ratones , Estimulación Luminosa , Tiempo de Reacción , Células Bipolares de la Retina
12.
Doc Ophthalmol ; 141(2): 195-204, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32239315

RESUMEN

PURPOSE: To report a patient with thymoma-associated retinopathy presenting as having a good visual prognosis. METHODS: Case report and literature review. CASE REPORT: A 42-year-old female patient was referred to our hospital for complaints of sudden visual-field defects bilaterally. Decimal corrected visual acuity (VA) was 1.5 and 1.2 in the right (RE) and left eyes (LE), respectively. Fundus autofluorescence revealed hyper-autofluorescence from the posterior pole to mid-peripheral retina in both eyes. Full-field electroretinography (ERG) amplitudes were reduced to 20-50% and 30-50% of our controls for the scotopic and photopic conditions, respectively. A systemic examination revealed the presence of thymoma, and the patient underwent thymectomy and immunosuppression therapies. Immunohistochemical analysis using the patient's serum showed immunolabeling on the photoreceptor inner segment and outer plexiform layer in the monkey retina. Two years later, VA remained at 1.5 and 1.2 in RE and LE. ERG amplitudes improved to 30-60% of the controls for the scotopic conditions. However, photopic ERG showed no remarkable change. CONCLUSIONS: To our knowledge, improvement of reduced rod-mediated ERG responses has not been described in seven previously reported patients with thymoma-associated retinopathy. The good visual prognosis of our patient may be associated with well-timed intervention.


Asunto(s)
Síndromes Paraneoplásicos Oculares/fisiopatología , Células Fotorreceptoras Retinianas Bastones/fisiología , Timoma/fisiopatología , Neoplasias del Timo/fisiopatología , Adulto , Terapia Combinada , Electrorretinografía , Femenino , Humanos , Inmunosupresores/uso terapéutico , Síndromes Paraneoplásicos Oculares/diagnóstico , Síndromes Paraneoplásicos Oculares/terapia , Timectomía , Timoma/diagnóstico , Timoma/terapia , Neoplasias del Timo/diagnóstico , Neoplasias del Timo/terapia , Tomografía de Coherencia Óptica , Trastornos de la Visión/fisiopatología , Agudeza Visual/fisiología , Campos Visuales/fisiología
13.
Proc Natl Acad Sci U S A ; 114(39): E8264-E8273, 2017 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-28900001

RESUMEN

Precise transcriptional regulation controlled by a transcription factor network is known to be crucial for establishing correct neuronal cell identities and functions in the CNS. In the retina, the expression of various cone and rod photoreceptor cell genes is regulated by multiple transcription factors; however, the role of epigenetic regulation in photoreceptor cell gene expression has been poorly understood. Here, we found that Samd7, a rod-enriched sterile alpha domain (SAM) domain protein, is essential for silencing nonrod gene expression through H3K27me3 regulation in rod photoreceptor cells. Samd7-null mutant mice showed ectopic expression of nonrod genes including S-opsin in rod photoreceptor cells and rod photoreceptor cell dysfunction. Samd7 physically interacts with Polyhomeotic homologs (Phc proteins), components of the Polycomb repressive complex 1 (PRC1), and colocalizes with Phc2 and Ring1B in Polycomb bodies. ChIP assays showed a significant decrease of H3K27me3 in the genes up-regulated in the Samd7-deficient retina, showing that Samd7 deficiency causes the derepression of nonrod gene expression in rod photoreceptor cells. The current study suggests that Samd7 is a cell type-specific PRC1 component epigenetically defining rod photoreceptor cell identity.


Asunto(s)
Proteínas del Ojo/metabolismo , Regulación de la Expresión Génica , Complejo Represivo Polycomb 1/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Animales , Proteínas del Ojo/genética , Ratones , Ratones Mutantes , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
15.
J Neurosci ; 37(8): 2073-2085, 2017 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-28115485

RESUMEN

Cellular asymmetries play crucial roles in development and organ function. The planar cell polarity (PCP) signaling pathway is involved in the establishment of cellular asymmetry within the plane of a cell sheet. Inner ear sensory hair cells (HCs), which have several rows of staircase-like stereocilia and one kinocilium located at the vertex of the stereocilia protruding from the apical surface of each HC, exhibit a typical form of PCP. Although connections between cilia and PCP signaling in vertebrate development have been reported, their precise nature is not well understood. During inner ear development, several ciliary proteins are known to play a role in PCP formation. In the current study, we investigated a functional role for intestinal cell kinase (Ick), which regulates intraflagellar transport (IFT) at the tip of cilia, in the mouse inner ear. A lack of Ick in the developing inner ear resulted in PCP defects in the cochlea, including misorientation or misshaping of stereocilia and aberrant localization of the kinocilium and basal body in the apical and middle turns, leading to auditory dysfunction. We also observed abnormal ciliary localization of Ift88 in both HCs and supporting cells. Together, our results show that Ick ciliary kinase is essential for PCP formation in inner ear HCs, suggesting that ciliary transport regulation is important for PCP signaling.SIGNIFICANCE STATEMENT The cochlea in the inner ear is the hearing organ. Planar cell polarity (PCP) in hair cells (HCs) in the cochlea is essential for mechanotransduction and refers to the asymmetric structure consisting of stereociliary bundles and the kinocilium on the apical surface of the cell body. We reported previously that a ciliary kinase, Ick, regulates intraflagellar transport (IFT). Here, we found that loss of Ick leads to abnormal localization of the IFT component in kinocilia, PCP defects in HCs, and hearing dysfunction. Our study defines the association of ciliary transport regulation with PCP formation in HCs and hearing function.


Asunto(s)
Polaridad Celular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Células Ciliadas Auditivas Internas/fisiología , Audición/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Animales Recién Nacidos , Antígenos/metabolismo , Dineínas Citoplasmáticas/genética , Dineínas Citoplasmáticas/metabolismo , Embrión de Mamíferos , Potenciales Evocados Auditivos del Tronco Encefálico/genética , Células Ciliadas Auditivas Internas/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Rastreo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Emisiones Otoacústicas Espontáneas/genética , Factor de Transcripción PAX2/genética , Factor de Transcripción PAX2/metabolismo , Proteínas Serina-Treonina Quinasas/genética
16.
J Neurosci ; 37(41): 9889-9900, 2017 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-28899920

RESUMEN

Neurotransmission plays an essential role in neural circuit formation in the central nervous system (CNS). Although neurotransmission has been recently clarified as a key modulator of retinal circuit development, the roles of individual synaptic transmissions are not yet fully understood. In the current study, we investigated the role of neurotransmission from photoreceptor cells to ON bipolar cells in development using mutant mouse lines of both sexes in which this transmission is abrogated. We found that deletion of the ON bipolar cation channel TRPM1 results in the abnormal contraction of rod bipolar terminals and a decreased number of their synaptic connections with amacrine cells. In contrast, these histological alterations were not caused by a disruption of total glutamate transmission due to loss of the ON bipolar glutamate receptor mGluR6 or the photoreceptor glutamate transporter VGluT1. In addition, TRPM1 deficiency led to the reduction of total dendritic length, branch numbers, and cell body size in AII amacrine cells. Activated Goα, known to close the TRPM1 channel, interacted with TRPM1 and induced the contraction of rod bipolar terminals. Furthermore, overexpression of Channelrhodopsin-2 partially rescued rod bipolar cell development in the TRPM1-/- retina, whereas the rescue effect by a constitutively closed form of TRPM1 was lower than that by the native form. Our results suggest that TRPM1 channel opening is essential for rod bipolar pathway establishment in development.SIGNIFICANCE STATEMENT Neurotransmission has been recognized recently as a key modulator of retinal circuit development in the CNS. However, the roles of individual synaptic transmissions are not yet fully understood. In the current study, we focused on neurotransmission between rod photoreceptor cells and rod bipolar cells in the retina. We used genetically modified mouse models which abrogate each step of neurotransmission: presynaptic glutamate release, postsynaptic glutamate reception, or transduction channel function. We found that the TRPM1 transduction channel is required for the development of rod bipolar cells and their synaptic formation with subsequent neurons, independently of glutamate transmission. This study advances our understanding of neurotransmission-mediated retinal circuit refinement.


Asunto(s)
Células Amacrinas/fisiología , Retina/crecimiento & desarrollo , Células Bipolares de la Retina/fisiología , Células Fotorreceptoras Retinianas Bastones/fisiología , Canales Catiónicos TRPM/fisiología , Vías Visuales/crecimiento & desarrollo , Vías Visuales/fisiología , Animales , Channelrhodopsins , Dendritas/fisiología , Dendritas/ultraestructura , Femenino , Ácido Glutámico/fisiología , Técnicas In Vitro , Masculino , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Retina/citología , Transmisión Sináptica/fisiología , Canales Catiónicos TRPM/genética , Proteína 1 de Transporte Vesicular de Glutamato/biosíntesis , Proteína 1 de Transporte Vesicular de Glutamato/genética
17.
EMBO J ; 33(11): 1227-42, 2014 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-24797473

RESUMEN

Cilia and flagella are formed and maintained by intraflagellar transport (IFT) and play important roles in sensing and moving across species. At the distal tip of the cilia/flagella, IFT complexes turn around to switch from anterograde to retrograde transport; however, the underlying regulatory mechanism is unclear. Here, we identified ICK localization at the tip of cilia as a regulator of ciliary transport. In ICK-deficient mice, we found ciliary defects in neuronal progenitor cells with Hedgehog signal defects. ICK-deficient cells formed cilia with mislocalized Hedgehog signaling components. Loss of ICK caused the accumulation of IFT-A, IFT-B, and BBSome components at the ciliary tips. In contrast, overexpression of ICK induced the strong accumulation of IFT-B, but not IFT-A or BBSome components at ciliary tips. In addition, ICK directly phosphorylated Kif3a, while inhibition of this Kif3a phosphorylation affected ciliary formation. Our results suggest that ICK is a Kif3a kinase and essential for proper ciliogenesis in development by regulating ciliary transport at the tip of cilia.


Asunto(s)
Cilios/metabolismo , Flagelos/metabolismo , Cinesinas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Transporte Biológico , Encéfalo/anomalías , Cilios/genética , Femenino , Flagelos/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Cinesinas/genética , Pulmón/anomalías , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Mutación , Neuronas/citología , Especificidad de Órganos , Fenotipo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Retina/citología , Transducción de Señal , Células Madre/ultraestructura
18.
J Biol Chem ; 291(47): 24465-24474, 2016 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-27681595

RESUMEN

In the retina, aberrant opsin transport from cell bodies to outer segments leads to retinal degenerative diseases such as retinitis pigmentosa. Opsin transport is facilitated by the intraflagellar transport (IFT) system that mediates the bidirectional movement of proteins within cilia. In contrast to functions of the anterograde transport executed by IFT complex B (IFT-B), the precise functions of the retrograde transport mediated by IFT complex A (IFT-A) have not been well studied in photoreceptor cilia. Here, we analyzed developing zebrafish larvae carrying a null mutation in ift122 encoding a component of IFT-A. ift122 mutant larvae show unexpectedly mild phenotypes, compared with those of mutants defective in IFT-B. ift122 mutants exhibit a slow onset of progressive photoreceptor degeneration mainly after 7 days post-fertilization. ift122 mutant larvae also develop cystic kidney but not curly body, both of which are typically observed in various ciliary mutants. ift122 mutants display a loss of cilia in the inner ear hair cells and nasal pit epithelia. Loss of ift122 causes disorganization of outer segment discs. Ectopic accumulation of an IFT-B component, ift88, is observed in the ift122 mutant photoreceptor cilia. In addition, pulse-chase experiments using GFP-opsin fusion proteins revealed that ift122 is required for the efficient transport of opsin and the distal elongation of outer segments. These results show that IFT-A is essential for the efficient transport of outer segment proteins, including opsin, and for the survival of retinal photoreceptor cells, rendering the ift122 mutant a unique model for human retinal degenerative diseases.


Asunto(s)
Opsinas/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Degeneración Retiniana/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Cilios/genética , Cilios/metabolismo , Células Ciliadas Auditivas Internas/metabolismo , Humanos , Mutación , Opsinas/genética , Transporte de Proteínas/genética , Degeneración Retiniana/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
19.
J Neurochem ; 143(1): 100-111, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28675567

RESUMEN

The retinal and anterior neural fold homeobox gene (Rax) controls development of the eye and the forebrain. Postnatal expression of Rax in the brain is restricted to the pineal gland, a forebrain structure devoted to melatonin synthesis. The role of Rax in pineal function is unknown. In order to investigate the role of Rax in pineal function while circumventing forebrain abnormalities of the global Rax knockout, we generated an eye and pineal-specific Rax conditional knockout mouse. Deletion of Rax in the pineal gland did not affect morphology of the gland, suggesting that Rax is not essential for pineal gland development. In contrast, deletion of Rax in the eye generated an anophthalmic phenotype. In addition to the loss of central visual pathways, the suprachiasmatic nucleus of the hypothalamus housing the circadian clock was absent, indicating that the retinohypothalamic tract is required for the nucleus to develop. Telemetric analyses confirmed the lack of a functional circadian clock. Arylalkylamine N-acetyltransferase (Aanat) transcripts, encoding the melatonin rhythm-generating enzyme, were undetectable in the pineal gland of the Rax conditional knockout under normal conditions, whereas the paired box 6 homeobox gene, known to regulate pineal development, was up-regulated. By injecting isoproterenol, which mimics a nocturnal situation in the pineal gland, we were able to induce pineal expression of Aanat in the Rax conditional knockout mouse, but Aanat transcript levels were significantly lower than those of Rax-proficient mice. Our data suggest that Rax controls pineal gene expression and via Aanat may modulate melatonin synthesis.


Asunto(s)
Ritmo Circadiano/fisiología , Proteínas del Ojo/fisiología , Genes Homeobox/fisiología , Proteínas de Homeodominio/fisiología , Glándula Pineal/metabolismo , Núcleo Supraquiasmático/metabolismo , Factores de Transcripción/fisiología , Vías Visuales/metabolismo , Animales , N-Acetiltransferasa de Arilalquilamina/biosíntesis , N-Acetiltransferasa de Arilalquilamina/genética , Proteínas del Ojo/genética , Femenino , Perfilación de la Expresión Génica/métodos , Proteínas de Homeodominio/genética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Células Neuroendocrinas/metabolismo , Retina/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
20.
J Pineal Res ; 63(2)2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28512916

RESUMEN

Tryptophan, an essential amino acid through a series of enzymatic reactions gives rise to various metabolites, viz. serotonin and melatonin, that regulate distinct biological functions. We show here that tryptophan metabolism in the pineal gland favors bone mass accrual through production of melatonin, a pineal-derived neurohormone. Pineal gland-specific deletion of Tph1, the enzyme that catalyzes the first step in the melatonin biosynthesis lead to a decrease in melatonin levels and a low bone mass due to an isolated decrease in bone formation while bone resorption parameters remained unaffected. Skeletal analysis of the mice deficient in MT1 or MT2 melatonin receptors showed a low bone mass in MT2-/- mice while MT1-/- mice had a normal bone mass compared to the WT mice. This low bone mass in the MT2-/- mice was due to an isolated decrease in osteoblast numbers and bone formation. In vitro assays of the osteoblast cultures derived from the MT1-/- and MT2-/- mice showed a cell intrinsic defect in the proliferation, differentiation and mineralization abilities of MT2-/- osteoblasts compared to WT counterparts, and the mutant cells did not respond to melatonin addition. Finally, we demonstrate that daily oral administration of melatonin can increase bone accrual during growth and can cure ovariectomy-induced structural and functional degeneration of bone by specifically increasing bone formation. By identifying pineal-derived melatonin as a regulator of bone mass through MT2 receptors, this study expands the role played by tryptophan derivatives in the regulation of bone mass and underscores its therapeutic relevance in postmenopausal osteoporosis.


Asunto(s)
Huesos/metabolismo , Melatonina/farmacología , Osteoblastos/metabolismo , Glándula Pineal/metabolismo , Receptor de Melatonina MT2/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Huesos/patología , Calcificación Fisiológica/efectos de los fármacos , Femenino , Humanos , Melatonina/metabolismo , Ratones , Ratones Noqueados , Tamaño de los Órganos/efectos de los fármacos , Osteoblastos/patología , Osteoporosis Posmenopáusica/tratamiento farmacológico , Osteoporosis Posmenopáusica/genética , Osteoporosis Posmenopáusica/metabolismo , Osteoporosis Posmenopáusica/patología , Glándula Pineal/patología , Receptor de Melatonina MT1/genética , Receptor de Melatonina MT1/metabolismo , Receptor de Melatonina MT2/genética , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA