Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(23): e2316858121, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38805270

RESUMEN

In mammals, CLOCK and BMAL1 proteins form a heterodimer that binds to E-box sequences and activates transcription of target genes, including Period (Per). Translated PER proteins then bind to the CLOCK-BMAL1 complex to inhibit its transcriptional activity. However, the molecular mechanism and the impact of this PER-dependent inhibition on the circadian clock oscillation remain elusive. We previously identified Ser38 and Ser42 in a DNA-binding domain of CLOCK as phosphorylation sites at the PER-dependent inhibition phase. In this study, knockout rescue experiments showed that nonphosphorylatable (Ala) mutations at these sites shortened circadian period, whereas their constitutive-phospho-mimetic (Asp) mutations completely abolished the circadian rhythms. Similarly, we found that nonphosphorylatable (Ala) and constitutive-phospho-mimetic (Glu) mutations at Ser78 in a DNA-binding domain of BMAL1 also shortened the circadian period and abolished the rhythms, respectively. The mathematical modeling predicted that these constitutive-phospho-mimetic mutations weaken the DNA binding of the CLOCK-BMAL1 complex and that the nonphosphorylatable mutations inhibit the PER-dependent displacement (reduction of DNA-binding ability) of the CLOCK-BMAL1 complex from DNA. Biochemical experiments supported the importance of these phosphorylation sites for displacement of the complex in the PER2-dependent inhibition. Our results provide direct evidence that phosphorylation of CLOCK-Ser38/Ser42 and BMAL1-Ser78 plays a crucial role in the PER-dependent inhibition and the determination of the circadian period.


Asunto(s)
Factores de Transcripción ARNTL , Proteínas CLOCK , Relojes Circadianos , Proteínas Circadianas Period , Animales , Humanos , Ratones , Factores de Transcripción ARNTL/metabolismo , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/química , Relojes Circadianos/genética , Ritmo Circadiano/fisiología , Ritmo Circadiano/genética , Proteínas CLOCK/metabolismo , Proteínas CLOCK/genética , ADN/metabolismo , Células HEK293 , Mutación , Células 3T3 NIH , Proteínas Circadianas Period/metabolismo , Proteínas Circadianas Period/genética , Fosforilación , Unión Proteica , Dominios Proteicos
2.
Development ; 150(4)2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36458527

RESUMEN

Ramified, polarized protoplasmic astrocytes interact with synapses via perisynaptic astrocyte processes (PAPs) to form tripartite synapses. These astrocyte-synapse interactions mutually regulate their structures and functions. However, molecular mechanisms for tripartite synapse formation remain elusive. We developed an in vitro co-culture system for mouse astrocytes and neurons that induced astrocyte ramifications and PAP formation. Co-cultured neurons were required for astrocyte ramifications in a neuronal activity-dependent manner, and synaptically-released glutamate and activation of astrocytic mGluR5 metabotropic glutamate receptor were likely involved in astrocyte ramifications. Astrocytic Necl2 trans-interacted with axonal Necl3, inducing astrocyte-synapse interactions and astrocyte functional polarization by recruiting EAAT1/2 glutamate transporters and Kir4.1 K+ channel to the PAPs, without affecting astrocyte ramifications. This Necl2/3 trans-interaction increased functional synapse number. Thus, astrocytic Necl2, synaptically-released glutamate and axonal Necl3 cooperatively formed tripartite glutamatergic synapses in vitro. Studies on hippocampal mossy fiber synapses in Necl3 knockout and Necl2/3 double knockout mice confirmed these previously unreported mechanisms for astrocyte-synapse interactions and astrocyte functional polarization in vivo.


Asunto(s)
Ácido Glutámico , Sinapsis , Ratones , Animales , Sinapsis/fisiología , Ratones Noqueados , Ácido Glutámico/farmacología , Astrocitos/fisiología , Fibras Musgosas del Hipocampo
3.
Cell ; 141(6): 1056-67, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20550939

RESUMEN

In the mouse olfactory system, the anatomical locations of olfactory sensory neurons (OSNs) roughly correlate with their axonal projection sites along the dorsal-ventral (D-V) axis of the olfactory bulb (OB). Here we report that an axon guidance receptor, Neuropilin-2 (Nrp2), and its repulsive ligand, Semaphorin-3F (Sema3F), are expressed by OSNs in a complementary manner that is important for establishing olfactory map topography. Sema3F is secreted by early-arriving axons of OSNs and is deposited at the anterodorsal OB to repel Nrp2-positive axons that arrive later. Sequential arrival of OSN axons as well as the graded and complementary expression of Nrp2 and Sema3F by OSNs help to form the topographic order along the D-V axis.


Asunto(s)
Axones/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Bulbo Olfatorio/metabolismo , Animales , Expresión Génica , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Neuropilina-2/metabolismo , Receptores de Superficie Celular/metabolismo , Inactivación del Cromosoma X
4.
Pflugers Arch ; 475(4): 489-504, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36749388

RESUMEN

Uric acid, the end product of purine metabolism in humans, is crucial because of its anti-oxidant activity and a causal relationship with hyperuricemia and gout. Several physiologically important urate transporters regulate this water-soluble metabolite in the human body; however, the existence of latent transporters has been suggested in the literature. We focused on the Escherichia coli urate transporter YgfU, a nucleobase-ascorbate transporter (NAT) family member, to address this issue. Only SLC23A proteins are members of the NAT family in humans. Based on the amino acid sequence similarity to YgfU, we hypothesized that SLC23A1, also known as sodium-dependent vitamin C transporter 1 (SVCT1), might be a urate transporter. First, we identified human SVCT1 and mouse Svct1 as sodium-dependent low-affinity/high-capacity urate transporters using mammalian cell-based transport assays. Next, using the CRISPR-Cas9 system followed by the crossing of mice, we generated Svct1 knockout mice lacking both urate transporter 1 and uricase. In the hyperuricemic mice model, serum urate levels were lower than controls, suggesting that Svct1 disruption could reduce serum urate. Given that Svct1 physiologically functions as a renal vitamin C re-absorber, it could also be involved in urate re-uptake from urine, though additional studies are required to obtain deeper insights into the underlying mechanisms. Our findings regarding the dual-substrate specificity of SVCT1 expand the understanding of urate handling systems and functional evolutionary changes in NAT family proteins.


Asunto(s)
Transportadores de Anión Orgánico , Ácido Úrico , Animales , Humanos , Ratones , Secuencia de Aminoácidos , Ácido Ascórbico/metabolismo , Transporte Biológico , Mamíferos/metabolismo , Transportadores de Anión Orgánico/metabolismo , Transportadores de Sodio Acoplados a la Vitamina C/genética , Transportadores de Sodio Acoplados a la Vitamina C/metabolismo , Ácido Úrico/metabolismo
5.
Biochem Biophys Res Commun ; 676: 190-197, 2023 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-37523817

RESUMEN

Brk/Ptk6, Srms, and Frk constitute a Src-related but distinct family of tyrosine kinases called Brk family kinases (BFKs) in higher vertebrates. To date, however, their biological roles have remained largely unknown. In this study, we generated BFK triple-knockout (BFK/TKO) mice lacking all BFK members using CRISPR/Cas9-mediated genome editing. BFK/TKO mice exhibited impaired intestinal homeostasis, represented by a reduced stem/progenitor cell population and defective recovery from radiation-induced severe mucosal damage, specifically in the ileum, which is the most distal segment of the small intestine. RNA-seq analysis revealed that BFK/TKO ileal epithelium showed markedly elevated IL-22/STAT3 signaling, resulting in the aberrant activation of mucosal immune response and altered composition of the ileal microbiota. Since single- or double-knockout of BFK genes did not elicit such abnormalities, BFKs may redundantly confer robust homeostasis to the ileum, the most recently added intestinal segment that plays crucial roles in nutrient absorption and mucosal immunity. Given that BFK diversification preceded the appearance of the ileum in vertebrate phylogeny, the present study highlights the coevolution of genes and organs, the former of which shapes up the latter in higher vertebrates.


Asunto(s)
Íleon , Transducción de Señal , Ratones , Animales , Intestino Delgado , Homeostasis
6.
Biochem Biophys Res Commun ; 657: 119-127, 2023 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-37002985

RESUMEN

Studying the non-human primate (NHP) brain is required for the translation of rodent research to humans, but remains a challenge for molecular, cellular, and circuit-level analyses in the NHP brain due to the lack of in vitro NHP brain system. Here, we report an in vitro NHP cerebral model using marmoset (Callithrix jacchus) embryonic stem cell-derived cerebral assembloids (CAs) that recapitulate inhibitory neuron migration and cortical network activity. Cortical organoids (COs) and ganglionic eminence organoids (GEOs) were induced from cjESCs and fused to generate CAs. GEO cells expressing the inhibitory neuron marker LHX6 migrated toward the cortical side of CAs. COs developed their spontaneous neural activity from a synchronized pattern to an unsynchronized pattern as COs matured. CAs containing excitatory and inhibitory neurons showed mature neural activity with an unsynchronized pattern. The CAs represent a powerful in vitro model for studying excitatory and inhibitory neuron interactions, cortical dynamics, and their dysfunction. The marmoset assembloid system will provide an in vitro platform for the NHP neurobiology and facilitate translation into humans in neuroscience research, regenerative medicine, and drug discovery.


Asunto(s)
Encéfalo , Callithrix , Animales , Encéfalo/fisiología , Neuronas , Neurogénesis , Células Madre Embrionarias
7.
Proc Natl Acad Sci U S A ; 117(31): 18175-18177, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32690690

RESUMEN

Recent genome-wide association studies have revealed some genetic loci associated with serum uric acid levels and susceptibility to gout/hyperuricemia which contain potential candidates of physiologically important urate transporters. One of these novel loci is located upstream of SGK1 and SLC2A12, suggesting that variations in these genes increase the risks of hyperuricemia and gout. We herein focused on SLC2A12 encoding a transporter, GLUT12, the physiological function of which remains unclear. As GLUT12 belongs to the same protein family as a well-recognized urate transporter GLUT9, we hypothesized that GLUT12 mediates membrane transport of urate. Therefore, we conducted functional assays and analyzed Glut12 knockout hyperuricemia model mice, generated using the CRISPR-Cas9 system. Our results revealed that GLUT12 acts as a physiological urate transporter and its dysfunction elevates the blood urate concentration. This study provides insights into the deeper understanding of the urate regulatory system in the body, which is also important for pathophysiology of gout/hyperuricemia.


Asunto(s)
Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Hiperuricemia/sangre , Ácido Úrico/sangre , Animales , Regulación de la Expresión Génica , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Ratones , Ratones Noqueados , Ácido Úrico/metabolismo
8.
Int J Mol Sci ; 24(5)2023 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-36902044

RESUMEN

White adipocytes act as lipid storage, and play an important role in energy homeostasis. The small GTPase Rac1 has been implicated in the regulation of insulin-stimulated glucose uptake in white adipocytes. Adipocyte-specific rac1-knockout (adipo-rac1-KO) mice exhibit atrophy of subcutaneous and epididymal white adipose tissue (WAT); white adipocytes in these mice are significantly smaller than controls. Here, we aimed to investigate the mechanisms underlying the aberrations in the development of Rac1-deficient white adipocytes by employing in vitro differentiation systems. Cell fractions containing adipose progenitor cells were obtained from WAT and subjected to treatments that induced differentiation into adipocytes. In concordance with observations in vivo, the generation of lipid droplets was significantly attenuated in Rac1-deficient adipocytes. Notably, the induction of various enzymes responsible for de novo synthesis of fatty acids and triacylglycerol in the late stage of adipogenic differentiation was almost completely suppressed in Rac1-deficient adipocytes. Furthermore, the expression and activation of transcription factors, such as the CCAAT/enhancer-binding protein (C/EBP) ß, which is required for the induction of lipogenic enzymes, were largely inhibited in Rac1-deficient cells in both early and late stages of differentiation. Altogether, Rac1 is responsible for adipogenic differentiation, including lipogenesis, through the regulation of differentiation-related transcription.


Asunto(s)
Lipogénesis , Proteínas de Unión al GTP Monoméricas , Ratones , Animales , Proteínas de Unión al GTP Monoméricas/metabolismo , Adipogénesis , Diferenciación Celular , Triglicéridos/metabolismo , Tejido Adiposo Blanco/metabolismo , Células Madre/metabolismo , Células 3T3-L1 , Tejido Adiposo/metabolismo
9.
Int J Mol Sci ; 22(19)2021 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-34639094

RESUMEN

Insulin stimulates glucose uptake in adipose tissue and skeletal muscle by inducing plasma membrane translocation of the glucose transporter GLUT4. Although the small GTPase Rac1 is a key regulator downstream of phosphoinositide 3-kinase (PI3K) and the protein kinase Akt2 in skeletal muscle, it remains unclear whether Rac1 also regulates glucose uptake in white adipocytes. Herein, we investigated the physiological role of Rac1 in white adipocytes by employing adipocyte-specific rac1 knockout (adipo-rac1-KO) mice. Subcutaneous and epididymal white adipose tissues (WATs) in adipo-rac1-KO mice showed significant reductions in size and weight. Actually, white adipocytes lacking Rac1 were smaller than controls. Insulin-stimulated glucose uptake and GLUT4 translocation were abrogated in rac1-KO white adipocytes. On the other hand, GLUT4 translocation was augmented by constitutively activated PI3K or Akt2 in control, but not in rac1-KO, white adipocytes. Similarly, to skeletal muscle, the involvement of another small GTPase RalA downstream of Rac1 was demonstrated. In addition, mRNA levels of various lipogenic enzymes were down-regulated in rac1-KO white adipocytes. Collectively, these results suggest that Rac1 is implicated in insulin-dependent glucose uptake and lipogenesis in white adipocytes, and reduced insulin responsiveness due to the deficiency of Rac1 may be a likely explanation for atrophy of WATs.


Asunto(s)
Tejido Adiposo Blanco/patología , Transportador de Glucosa de Tipo 4/metabolismo , Glucosa/metabolismo , Insulina/farmacología , Neuropéptidos/fisiología , Proteína de Unión al GTP rac1/fisiología , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Atrofia , Femenino , Transportador de Glucosa de Tipo 4/genética , Hipoglucemiantes/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Edulcorantes/farmacología
10.
Hum Mol Genet ; 27(7): 1174-1185, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29360985

RESUMEN

Congenital muscular dystrophies (CMDs) are characterized by progressive weakness and degeneration of skeletal muscle. In several forms of CMD, abnormal glycosylation of α-dystroglycan (α-DG) results in conditions collectively known as dystroglycanopathies, which are associated with central nervous system involvement. We recently demonstrated that fukutin, the gene responsible for Fukuyama congenital muscular dystrophy, encodes the ribitol-phosphate transferase essential for dystroglycan function. Brain pathology in patients with dystroglycanopathy typically includes cobblestone lissencephaly, mental retardation, and refractory epilepsy; however, some patients exhibit average intelligence, with few or almost no structural defects. Currently, there is no effective treatment for dystroglycanopathy, and the mechanisms underlying the generation of this broad clinical spectrum remain unknown. Here, we analysed four distinct mouse models of dystroglycanopathy: two brain-selective fukutin conditional knockout strains (neuronal stem cell-selective Nestin-fukutin-cKO and forebrain-selective Emx1-fukutin-cKO), a FukutinHp strain with the founder retrotransposal insertion in the fukutin gene, and a spontaneous Large-mutant Largemyd strain. These models exhibit variations in the severity of brain pathology, replicating the clinical heterogeneity of dystroglycanopathy. Immunofluorescence analysis of the developing cortex suggested that residual glycosylation of α-DG at embryonic day 13.5 (E13.5), when cortical dysplasia is not yet apparent, may contribute to subsequent phenotypic heterogeneity. Surprisingly, delivery of fukutin or Large into the brains of mice at E12.5 prevented severe brain malformation in Emx1-fukutin-cKO and Largemyd/myd mice, respectively. These findings indicate that spatiotemporal persistence of functionally glycosylated α-DG may be crucial for brain development and modulation of glycosylation during the fetal stage could be a potential therapeutic strategy for dystroglycanopathy.


Asunto(s)
Encéfalo/embriología , Distroglicanos/metabolismo , Feto/embriología , Técnicas de Transferencia de Gen , Terapia Genética , Malformaciones del Desarrollo Cortical/terapia , Animales , Encéfalo/patología , Distroglicanos/genética , Femenino , Feto/patología , Glicosilación , Masculino , Malformaciones del Desarrollo Cortical/embriología , Malformaciones del Desarrollo Cortical/genética , Malformaciones del Desarrollo Cortical/patología , Ratones , Ratones Transgénicos
11.
Development ; 144(10): 1863-1875, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28512198

RESUMEN

Rac signaling impacts a relatively large number of downstream targets; however, few studies have established an association between Rac pathways and pathological conditions. In the present study, we generated mice with double knockout of Rac1 and Rac3 (Atoh1-Cre;Rac1flox/flox;Rac3-/- ) in cerebellar granule neurons (CGNs). We observed impaired tangential migration at E16.5, as well as numerous apoptotic CGNs at the deepest layer of the external granule layer (EGL) in the medial cerebellum of Atoh1-Cre;Rac1flox/flox;Rac3-/- mice at P8. Atoh1-Cre;Rac1flox/flox;Rac3-/- CGNs differentiated normally until expression of p27kip1 and NeuN in the deep EGL at P5. Primary CGNs and cerebellar microexplants from Atoh1-Cre;Rac1flox/flox;Rac3-/- mice exhibited impaired neuritogenesis, which was more apparent in Map2-positive dendrites. Such findings suggest that impaired tangential migration and final differentiation of CGNs have resulted in decreased cerebellum size and agenesis of the medial internal granule layer, respectively. Furthermore, Rac depleted/deleted cells exhibited decreased levels of Mid1 and impaired mTORC1 signaling. Mid1 depletion in CGNs produced mild impairments in neuritogenesis and reductions in mTORC1 signaling. Thus, a novel Rac-signaling pathway (Rac1-Mid1-mTORC1) may be involved in medial cerebellar development.


Asunto(s)
Cerebelo/embriología , Proteínas/fisiología , Proteínas de Unión al GTP rac/fisiología , Animales , Diferenciación Celular/genética , Células Cultivadas , Cerebelo/metabolismo , Células HEK293 , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Complejos Multiproteicos/fisiología , Neurogénesis/genética , Organogénesis/genética , Proteínas/genética , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/fisiología , Ubiquitina-Proteína Ligasas , Proteínas de Unión al GTP rac/genética
12.
Mol Reprod Dev ; 86(8): 928-930, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31215717

RESUMEN

The common marmoset is a small nonhuman primate in which the application of transgenesis and genetic knockout techniques allows the generation of gene-modified models of human diseases. However, its longer generation time than that of rodents is a major obstacle to the widespread use of gene-modified marmosets for biomedical research. In this study, we examined the feasibility of shortening the generation time by using prepubertal marmoset males as gamete donors. We collected late round stage spermatids (Steps 5-7), elongated spermatids, and testicular spermatozoa from the testis of a prepubertal 11-month-old male marmoset and injected them into in vitro-matured oocytes. After 7 days in culture, two embryos from elongated spermatid injection and two embryos from sperm injection were transferred into two separate recipient females. The recipient female that received elongated spermatid injection-derived embryos became pregnant and gave birth to one female infant. This is the first demonstration that a spermatid from a prepubertal male primate can support full-term development. Using this method, we can expect to obtain offspring of gene-modified males 6 months to a year earlier than with natural mating.


Asunto(s)
Nacimiento Vivo , Inyecciones de Esperma Intracitoplasmáticas , Espermátides , Animales , Callithrix , Femenino , Masculino , Embarazo
13.
Proc Natl Acad Sci U S A ; 113(8): 2282-7, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26858447

RESUMEN

In Purkinje cells (PCs) of the cerebellum, a single "winner" climbing fiber (CF) monopolizes proximal dendrites, whereas hundreds of thousands of parallel fibers (PFs) innervate distal dendrites, and both CF and PF inputs innervate a narrow intermediate domain. It is unclear how this segregated CF and PF innervation is established on PC dendrites. Through reconstruction of dendritic innervation by serial electron microscopy, we show that from postnatal day 9-15 in mice, both CF and PF innervation territories vigorously expand because of an enlargement of the region of overlapping innervation. From postnatal day 15 onwards, segregation of these territories occurs with robust shortening of the overlapping proximal region. Thus, innervation territories by the heterologous inputs are refined during the early postnatal period. Intriguingly, this transition is arrested in mutant mice lacking the type 1 metabotropic glutamate receptor (mGluR1) or protein kinase Cγ (PKCγ), resulting in the persistence of an abnormally expanded overlapping region. This arrested territory refinement is rescued by lentivirus-mediated expression of mGluR1α into mGluR1-deficient PCs. At the proximal dendrite of rescued PCs, PF synapses are eliminated and free spines emerge instead, whereas the number and density of CF synapses are unchanged. Because the mGluR1-PKCγ signaling pathway is also essential for the late-phase of CF synapse elimination, this signaling pathway promotes the two key features of excitatory synaptic wiring in PCs, namely CF monoinnervation by eliminating redundant CF synapses from the soma, and segregated territories of CF and PF innervation by eliminating competing PF synapses from proximal dendrites.


Asunto(s)
Células de Purkinje/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Animales , Dendritas/fisiología , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Modelos Neurológicos , Proteína Quinasa C/deficiencia , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Células de Purkinje/ultraestructura , Receptores de Glutamato Metabotrópico/deficiencia , Receptores de Glutamato Metabotrópico/genética , Transducción de Señal , Sinapsis/fisiología
14.
J Physiol ; 596(16): 3775-3791, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29874406

RESUMEN

KEY POINTS: Perioral tactile signals are transmitted via the infraorbital nerve (ION) to trigeminal nuclei. Each cerebellar Purkinje cell (PC) receives this signal as complex spikes (CSs) via a climbing fibre (CF) emerging from the inferior olive (IO). The anatomical pathway from trigeminal nuclei to the IO is not clearly identified. In the present study, we examined candidate anatomical pathways for perioral sensory signalling by analysing CSs recorded from PCs in male mice by single unit recording. CS generation by ION stimulation was inhibited by injection of a GABAA receptor agonist, muscimol, into the contralateral mesodiencephalic junction, which is referred to as the area parafascicularis prerubralis (PfPr). The number of CSs evoked by mechanical whisker stimulation was also decreased by contralateral PfPr inhibition. These results suggest the existence of a sensory signalling pathway to the IO via the PfPr in mice. ABSTRACT: Perioral tactile signals are transmitted via the infraorbital nerve (ION) to trigeminal nuclei. Each cerebellar Purkinje cell receives this signal as complex spikes (CSs) via a climbing fibre emerging from the inferior olive (IO). However, the anatomical pathway from the trigeminal nuclei to the IO is not clearly identified. In the present study, we recorded CSs from Purkinje cells in male mice by single unit recording, and examined the signal transduction pathway. CSs were evoked by electrical stimulation of the ipsilateral or contralateral ION with a latency of 20-70 ms. CS generation by ipsilateral ION stimulation was inhibited by injection of a GABAA receptor agonist, muscimol, into the contralateral mesodiencephalic junction, ranging from around the fasciculus retroflexus to the interstitial nucleus of Cajal, which is referred to as the area parafascicularis prerubralis (PfPr). CSs evoked by contralateral ION stimulation were also suppressed by muscimol injection into the PfPr, although the effective area was more restricted. Furthermore, CSs evoked by mechanical stimulation around the whisker region were suppressed by PfPr inhibition. We also found that the primary motor cortex plays a role to suppress this signalling pathway. These results indicate the existence of an anatomical pathway for conducting perioral sensory signals to the IO via the PfPr.


Asunto(s)
Cerebelo/fisiología , Diencéfalo/fisiología , Mesencéfalo/fisiología , Boca/fisiología , Núcleo Olivar/fisiología , Células de Purkinje/fisiología , Células Receptoras Sensoriales/fisiología , Animales , Células Cultivadas , Cerebelo/citología , Cerebelo/efectos de los fármacos , Diencéfalo/citología , Diencéfalo/efectos de los fármacos , Agonistas de Receptores de GABA-A/farmacología , Masculino , Mesencéfalo/citología , Mesencéfalo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Boca/citología , Boca/efectos de los fármacos , Muscimol/farmacología , Núcleo Olivar/citología , Núcleo Olivar/efectos de los fármacos , Células de Purkinje/citología , Células de Purkinje/efectos de los fármacos , Receptores de GABA-A/química , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/efectos de los fármacos
15.
J Biol Chem ; 292(4): 1240-1250, 2017 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-27941025

RESUMEN

Astrogliosis (i.e. glial scar), which is comprised primarily of proliferated astrocytes at the lesion site and migrated astrocytes from neighboring regions, is one of the key reactions in determining outcomes after CNS injury. In an effort to identify potential molecules/pathways that regulate astrogliosis, we sought to determine whether Rac/Rac-mediated signaling in astrocytes represents a novel candidate for therapeutic intervention following CNS injury. For these studies, we generated mice with Rac1 deletion under the control of the GFAP (glial fibrillary acidic protein) promoter (GFAP-Cre;Rac1flox/flox). GFAP-Cre;Rac1flox/flox (Rac1-KO) mice exhibited better recovery after spinal cord injury and exhibited reduced astrogliosis at the lesion site relative to control. Reduced astrogliosis was also observed in Rac1-KO mice following microbeam irradiation-induced injury. Moreover, knockdown (KD) or KO of Rac1 in astrocytes (LN229 cells, primary astrocytes, or primary astrocytes from Rac1-KO mice) led to delayed cell cycle progression and reduced cell migration. Rac1-KD or Rac1-KO astrocytes additionally had decreased levels of GSPT1 (G1 to S phase transition 1) expression and reduced responses of IL-1ß and GSPT1 to LPS treatment, indicating that IL-1ß and GSPT1 are downstream molecules of Rac1 associated with inflammatory condition. Furthermore, GSPT1-KD astrocytes had cell cycle delay, with no effect on cell migration. The cell cycle delay induced by Rac1-KD was rescued by overexpression of GSPT1. Based on these results, we propose that Rac1-GSPT1 represents a novel signaling axis in astrocytes that accelerates proliferation in response to inflammation, which is one important factor in the development of astrogliosis/glial scar following CNS injury.


Asunto(s)
Astrocitos/metabolismo , Gliosis/metabolismo , Neuropéptidos/metabolismo , Factores de Terminación de Péptidos/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Astrocitos/patología , Gliosis/genética , Gliosis/patología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ratones , Ratones Noqueados , Neuropéptidos/genética , Factores de Terminación de Péptidos/genética , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología , Proteína de Unión al GTP rac1/genética
16.
Mol Reprod Dev ; 85(5): 376-386, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29457675

RESUMEN

The common marmoset (Callithrix jacchus) represents a promising nonhuman primate model for the study of human diseases because of its small size, ease of handling, and availability of gene-modified animals. Here, we aimed to devise reproductive technology for marmoset spermatid injection using immature males for a possible rapid generational turnover. Spermatids at each step could be identified easily by their morphology under differential interference microscopy: thus, early round spermatids had a round nucleus with a few nucleolus-like structures and abundant cytoplasm, as in other mammals. The spermatids acquired oocyte-activating capacity at the late round spermatid stage, as confirmed by the resumption of meiosis and Ca2+ oscillations upon injection into mouse oocytes. The spermatids could be cryopreserved efficiently with a simple medium containing glycerol and CELL BANKER®. Late round or elongated spermatids first appeared at 10-12 months of age, 6-8 months before sexual maturation. Marmoset oocytes microinjected with frozen-thawed late round or elongated spermatids retrieved from a 12-month-old male marmoset developed to the 8-cell stage without the need for artificial oocyte activation stimulation. Thus, it might be possible to shorten the intergeneration time by spermatid injection, from 2 years (by natural mating) to 13-15 months including gestation.


Asunto(s)
Señalización del Calcio , Núcleo Celular/metabolismo , Criopreservación , Técnicas de Transferencia Nuclear , Oocitos/metabolismo , Espermátides/metabolismo , Animales , Callithrix , Femenino , Masculino , Ratones , Microinyecciones , Oocitos/citología , Espermátides/citología
17.
Glia ; 65(1): 182-197, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27726178

RESUMEN

Ror2 receptor tyrosine kinase plays crucial roles in developmental morphogenesis and tissue-/organo-genesis. In the developing brain, Ror2 is expressed in neural stem/progenitor cells (NPCs) and involved in the regulation of their stemness. However, it remains largely unknown about its role in the adult brain. In this study, we show that Ror2 is up-regulated in reactive astrocytes in the neocortices within 3 days following stab-wound injury. Intriguingly, Ror2-expressing astrocytes were detected primarily at the area surrounding the injury site, where astrocytes express Nestin, a marker of NPCs, and proliferate in response to injury. Furthermore, we show by using astrocyte-specific Ror2 knockout (KO) mice that a loss of Ror2 in astrocytes attenuates injury-induced proliferation of reactive astrocytes. It was also found that basic fibroblast growth factor (bFGF) is strongly up-regulated at 1 day post injury in the neocortices, and that stimulation of cultured quiescent astrocytes with bFGF restarts their cell cycle and induces expression of Ror2 during the G1 phase predominantly in proliferating cells. By using this culture method, we further show that the proportions of Ror2-expressing astrocytes increase following treatment with the histone deacetylases inhibitors including valproic acid, and that bFGF stimulation increases the levels of Ror2 expression within the respective cells. Moreover, we show that bFGF-induced cell cycle progression into S phase is inhibited or promoted in astrocytes from Ror2 KO mice or NPCs stably expressing Ror2-GFP, respectively. Collectively, these findings indicate that Ror2 plays a critical role in regulating the cell cycle progression of reactive astrocytes following brain injury, GLIA 2016. GLIA 2017;65:182-197.


Asunto(s)
Astrocitos/metabolismo , Lesiones Encefálicas/enzimología , Ciclo Celular/fisiología , División Celular , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Animales , Lesiones Encefálicas/patología , División Celular/fisiología , Células Cultivadas , Ratones Noqueados , Nestina/metabolismo , Células-Madre Neurales/metabolismo
18.
Prostate ; 77(15): 1489-1498, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28905415

RESUMEN

BACKGROUND: Neuroendocrine-differentiated prostate cancer (NEPCa) is refractory to androgen deprivation therapy and shows a poor prognosis. The underlying mechanisms responsible for neuroendocrine differentiation (NED) are yet to be clarified. In this study, we investigated the role of mammalian target of rapamycin (mTOR) in NEPCa. METHODS: We utilized a gain-of-function analysis by establishing a human PCa LNCaP stable line that expresses hyperactive mTOR (LNCaP-mTOR). Then, we employed a comprehensive mass spectrometric analysis to identify a key transcription factor in LNCaP-mTOR, followed by a loss-of-function analysis using CRISPR/Cas system. RESULTS: The activation of mTOR induced NED. We observed significant cell growth arrest in NED of LNCaP-mTOR, which accompanied increased expression of p21WAF1/CIP1 . A comprehensive mass spectrometric analysis identified interferon regulatory factor 1 (IRF1) as a key transcription factor in growth arrest of LNCaP-mTOR. The disruption of IRF1 gene in LNCaP-mTOR reversed cell growth arrest along with the suppression of its target p21WAF1/CIP1 . These results indicate that the growth arrest in NED is at least in part dependent on IRF1 through the induction of p21WAF1/CIP1 . CONCLUSIONS: We identified active mTOR as a novel inducer of NED, and elucidated a mechanism underlying the malignant transformation of NEPCa by recapitulating NED in vitro.


Asunto(s)
Factor 1 Regulador del Interferón/metabolismo , Tumores Neuroendocrinos/metabolismo , Neoplasias de la Próstata/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Técnicas de Inactivación de Genes , Xenoinjertos , Humanos , Factor 1 Regulador del Interferón/genética , Masculino , Ratones , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Serina-Treonina Quinasas TOR/genética , Regulación hacia Arriba
19.
Genesis ; 54(2): 65-77, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26713866

RESUMEN

The CRISPR/Cas system has rapidly emerged recently as a new tool for genome engineering, and is expected to allow for controlled manipulation of specific genomic elements in a variety of species. A number of recent studies have reported the use of CRISPR/Cas for gene disruption (knockout) or targeted insertion of foreign DNA elements (knock-in). Despite the ease of simple gene knockout and small insertions or nucleotide substitutions in mouse zygotes by the CRISPR/Cas system, targeted insertion of large DNA elements remains an apparent challenge. Here the generation of knock-in mice with successful targeted insertion of large donor DNA elements ranged from 3.0 to 7.1 kb at the ROSA26 locus using the CRISPR/Cas system was achieved. Multiple independent knock-in founder mice were obtained by injection of hCas9 mRNA/sgRNA/donor vector mixtures into the cytoplasm of C57BL/6N zygotes when the injected zygotes were treated with an inhibitor of actin polymerization, cytochalasin. Successful germ line transmission of three of these knock-in alleles was also confirmed. The results suggested that treatment of zygotes with actin polymerization inhibitors following microinjection could be a viable method to facilitate targeted insertion of large DNA elements by the CRISPR/Cas system, enabling targeted knock-in readily attainable in zygotes.


Asunto(s)
Sistemas CRISPR-Cas , Citocalasina B/farmacología , Técnicas de Sustitución del Gen/métodos , Ratones Mutantes/genética , Actinas/química , Secuencia de Aminoácidos , Animales , Citocalasina D/farmacología , ADN , Ratones , Ratones Endogámicos C57BL , Microinyecciones , Datos de Secuencia Molecular , ARN no Traducido/genética , Cigoto
20.
J Cell Sci ; 127(Pt 9): 2040-52, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24610943

RESUMEN

Cdc42 is a key regulator of dynamic actin organization. However, little is known about how Cdc42-dependent actin regulation influences steady-state actin structures in differentiated epithelia. We employed inner ear hair-cell-specific conditional knockout to analyze the role of Cdc42 in hair cells possessing highly elaborate stable actin protrusions (stereocilia). Hair cells of Atoh1-Cre;Cdc42(flox/flox) mice developed normally but progressively degenerated after maturation, resulting in progressive hearing loss particularly at high frequencies. Cochlear hair cell degeneration was more robust in inner hair cells than in outer hair cells, and began as stereocilia fusion and depletion, accompanied by a thinning and waving circumferential actin belt at apical junctional complexes (AJCs). Adenovirus-encoded GFP-Cdc42 expression in hair cells and fluorescence resonance energy transfer (FRET) imaging of hair cells from transgenic mice expressing a Cdc42-FRET biosensor indicated Cdc42 presence and activation at stereociliary membranes and AJCs in cochlear hair cells. Cdc42-knockdown in MDCK cells produced phenotypes similar to those of Cdc42-deleted hair cells, including abnormal microvilli and disrupted AJCs, and downregulated actin turnover represented by enhanced levels of phosphorylated cofilin. Thus, Cdc42 influenced the maintenance of stable actin structures through elaborate tuning of actin turnover, and maintained function and viability of cochlear hair cells.


Asunto(s)
Células Ciliadas Auditivas/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Actinas/metabolismo , Animales , Técnicas Biosensibles , Cóclea/citología , Cóclea/metabolismo , Perros , Transferencia Resonante de Energía de Fluorescencia , Humanos , Inmunohistoquímica , Hibridación in Situ , Células de Riñón Canino Madin Darby , Ratones , Microscopía Electroquímica de Rastreo , Microscopía Electrónica de Transmisión , Técnicas de Cultivo de Órganos/métodos , Proteína de Unión al GTP cdc42/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA