Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Adv ; 10(30): eadj9335, 2024 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-39058787

RESUMO

Mutations in Dystonin (DST), which encodes cytoskeletal linker proteins, cause hereditary sensory and autonomic neuropathy 6 (HSAN-VI) in humans and the dystonia musculorum (dt) phenotype in mice; however, the neuronal circuit underlying the HSAN-VI and dt phenotype is unresolved. dt mice exhibit dystonic movements accompanied by the simultaneous contraction of agonist and antagonist muscles and postnatal lethality. Here, we identified the sensory-motor circuit as a major causative neural circuit using a gene trap system that enables neural circuit-selective inactivation and restoration of Dst by Cre-mediated recombination. Sensory neuron-selective Dst deletion led to motor impairment, degeneration of proprioceptive sensory neurons, and disruption of the sensory-motor circuit. Restoration of Dst expression in sensory neurons using Cre driver mice or a single postnatal injection of Cre-expressing adeno-associated virus ameliorated sensory degeneration and improved abnormal movements. These findings demonstrate that the sensory-motor circuit is involved in the movement disorders in dt mice and that the sensory circuit is a therapeutic target for HSAN-VI.


Assuntos
Modelos Animais de Doenças , Distonina , Neuropatias Hereditárias Sensoriais e Autônomas , Células Receptoras Sensoriais , Animais , Camundongos , Células Receptoras Sensoriais/metabolismo , Distonina/genética , Neuropatias Hereditárias Sensoriais e Autônomas/genética , Distonia/genética , Humanos , Dependovirus/genética , Fenótipo
2.
Acta Neuropathol ; 147(1): 76, 2024 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-38658413

RESUMO

Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune disease of the CNS characterized by the production of disease-specific autoantibodies against aquaporin-4 (AQP4) water channels. Animal model studies suggest that anti-AQP4 antibodies cause a loss of AQP4-expressing astrocytes, primarily via complement-dependent cytotoxicity. Nonetheless, several aspects of the disease remain unclear, including: how anti-AQP4 antibodies cross the blood-brain barrier from the periphery to the CNS; how NMOSD expands into longitudinally extensive transverse myelitis or optic neuritis; how multiphasic courses occur; and how to prevent attacks without depleting circulating anti-AQP4 antibodies, especially when employing B-cell-depleting therapies. To address these knowledge gaps, we conducted a comprehensive 'stage-dependent' investigation of immune cell elements in situ in human NMOSD lesions, based on neuropathological techniques for autopsied/biopsied CNS materials. The present study provided three major findings. First, activated or netting neutrophils and melanoma cell adhesion molecule-positive (MCAM+) helper T (TH) 17/cytotoxic T (TC) 17 cells are prominent, and the numbers of these correlate with the size of NMOSD lesions in the initial or early-active stages. Second, forkhead box P3-positive (FOXP3+) regulatory T (Treg) cells are recruited to NMOSD lesions during the initial, early-active or late-active stages, suggesting rapid suppression of proinflammatory autoimmune events in the active stages of NMOSD. Third, compartmentalized resident memory immune cells, including CD103+ tissue-resident memory T (TRM) cells with long-lasting inflammatory potential, are detected under "standby" conditions in all stages. Furthermore, CD103+ TRM cells express high levels of granzyme B/perforin-1 in the initial or early-active stages of NMOSD in situ. We infer that stage-dependent compartmentalized immune traits orchestrate the pathology of anti-AQP4 antibody-guided NMOSD in situ. Our work further suggests that targeting activated/netting neutrophils, MCAM+ TH17/TC17 cells, and CD103+ TRM cells, as well as promoting the expansion of FOXP3+ Treg cells, may be effective in treating and preventing relapses of NMOSD.


Assuntos
Aquaporina 4 , Autoanticorpos , Neuromielite Óptica , Neutrófilos , Neuromielite Óptica/imunologia , Neuromielite Óptica/patologia , Aquaporina 4/imunologia , Humanos , Neutrófilos/imunologia , Neutrófilos/patologia , Feminino , Autoanticorpos/imunologia , Masculino , Pessoa de Meia-Idade , Memória Imunológica , Adulto , Idoso , Células Th17/imunologia , Células Th17/patologia
3.
Dis Model Mech ; 16(7)2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37415561

RESUMO

Phosphoinositides (PIPs) act as intracellular signaling molecules that regulate various cellular processes. Abnormalities in PIP metabolism cause various pathological conditions, including neurodegenerative diseases, cancer and immune disorders. Several neurological diseases with diverse phenotypes, such as ataxia with cerebellar atrophy or intellectual disability without brain malformation, are caused by mutations in INPP4A, which encodes a phosphoinositide phosphatase. We examined two strains of Inpp4a mutant mice with distinct cerebellar phenotypes: the Inpp4aΔEx1,2 mutant exhibited striatal degeneration without cerebellar atrophy, and the Inpp4aΔEx23 mutant exhibited a severe striatal phenotype with cerebellar atrophy. Both strains exhibited reduced expression of Inpp4a mutant proteins in the cerebellum. N-terminal-truncated Inpp4a proteins were expressed from the Inpp4aΔEx1,2 allele by alternative translation initiation and had phosphatase activity for PI(3,4)P2, whereas the Inpp4a mutant protein encoded by Inpp4aΔEx23 completely lacked phosphatase activity. Our results indicate that the diverse phenotypes observed in Inpp4a-related neurological diseases could be due to the varying protein expression levels and retained phosphatase activity in different Inpp4a variants. These findings provide insights into the role of INPP4A mutations in disease pathogenesis and may help to develop personalized therapy.


Assuntos
Cerebelo , Monoéster Fosfórico Hidrolases , Transdução de Sinais , Animais , Camundongos , Atrofia/patologia , Cerebelo/patologia , Fenótipo , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo
4.
J Biomol Struct Dyn ; 41(14): 6502-6517, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35938618

RESUMO

The human receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is a critical necroptosis regulator implicated in cancer, psoriasis, ulcerative colitis, rheumatoid arthritis, Alzheimer's disease, and multiple sclerosis. Currently, there are no specific RIPK1 antagonists in clinical practice. In this study, we took a target-based computational approach to identify blood-brain-barrier-permeable potent RIPK1 ligands with novel chemotypes. Using molecular docking, we virtually screened the Marine Natural Products (MNP) library of 14,492 small molecules. Initial 18 hits were subjected to detailed ADMET profiling for bioavailability, brain penetration, druglikeness, and toxicities and eventually yielded 548773-66-6 as the best ligand. RIPK1 548773-66-6 binding was validated through duplicated molecular dynamics (MD) simulations where the co-crystallized ligand L8D served as a reference. Trajectory analysis indicated negligible Root-Mean-Square-Deviations (RMSDs) of the best ligand 548773-66-6 relative to the protein backbone: 0.156 ± 0.043 nm and 0.296 ± 0.044 nm (mean ± standard deviations) in two individual simulations. Visual inspection confirmed that 548773-66-6 occupied the RIPK1 ligand-binding pocket associated with the kinase activation loop. Further computations demonstrated consistent hydrogen bond interactions of the ligand with the residue ASP156. Binding free energy estimation also supported stable interactions of 548773-66-6 and RIPK1. Together, our in silico analysis predicted 548773-66-6 as a novel ligand for RIPK1. Therefore, 548773-66-6 could be a viable lead for inhibiting necroptosis in central nervous system inflammatory disorders.Communicated by Ramaswamy H. Sarma.

5.
Cell Death Differ ; 29(5): 1028-1041, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34974536

RESUMO

Olig2 is indispensable for motoneuron and oligodendrocyte fate-specification in the pMN domain of embryonic spinal cords, and also involved in the proliferation and differentiation of several cell types in the nervous system, including neural progenitor cells (NPCs) and oligodendrocytes. However, how Olig2 controls these diverse biological processes remains unclear. Here, we demonstrated that a novel Olig2-binding protein, DEAD-box helicase 20 (Ddx20), is indispensable for the survival of NPCs and oligodendrocyte progenitor cells (OPCs). A central nervous system (CNS)-specific Ddx20 conditional knockout (cKO) demonstrated apoptosis and cell cycle arrest in NPCs and OPCs, through the potentiation of the p53 pathway in DNA damage-dependent and independent manners, including SMN complex disruption and the abnormal splicing of Mdm2 mRNA. Analyzes of Olig2 null NPCs showed that Olig2 contributed to NPC proliferation through Ddx20 protein stabilization. Our findings provide novel mechanisms underlying the Olig2-mediated proliferation of NPCs, via the Ddx20-p53 axis, in the embryonic CNS.


Assuntos
Células-Tronco Neurais , Células Precursoras de Oligodendrócitos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/genética , Células-Tronco Neurais/metabolismo , Células Precursoras de Oligodendrócitos/metabolismo , Oligodendroglia/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
6.
J Neurosci Methods ; 345: 108887, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32739417

RESUMO

BACKGROUND: Viral vector systems delivering transgenes in the retrograde direction through axons to neural cell bodies are powerful experimental tools for the functional analysis of specific neural pathways. Generally, the efficiency of viral vector-mediated retrograde gene transfer depends on the expression of requisite viral receptors in neural pathways projecting to the viral vector-injected regions. This is known as viral tropism and can limit the utility of retrograde viral vectors. The adeno-associated virus (AAV) vector has become an increasingly popular platform for gene delivery to neural cells in vivo, and it is therefore meaningful to develop a new type of retrograde gene transfer approach based on a tropism-free AAV vector system. NEW METHOD: The wild-type or mutant receptor gene of AAV was expressed to mitigate AAV tropism. RESULTS: Efficient AAV vector-mediated retrograde gene transfer was observed in diverse neural pathways by expression of the AAV receptor (AAVR) gene. Moreover, the expression of a minimal mutant of AAVR (miniAAVR), which maintains binding potential to AAV, demonstrated efficient retrograde gene expression comparable to that of AAVR. COMPARISON WITH EXISTING METHODS: The utility of existing AAV vector-mediated retrograde gene delivery methods is sometimes limited by tropism. Our newly developed AAV-AAVR and AAV-miniAAVR interaction approaches enabled efficient retrograde gene transfer into various neural pathways by mitigating tropism. CONCLUSIONS: AAV-AAVR and AAV-miniAAVR interaction approaches enabled us to induce efficient retrograde gene expression in targeted neural pathways and provide a powerful tool for analyzing specific neural pathways.


Assuntos
Dependovirus , Vetores Genéticos , Dependovirus/genética , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/genética , Vias Neurais , Transdução Genética
7.
Ann Neurol ; 80(4): 554-65, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27490250

RESUMO

OBJECTIVE: To clarify the histopathological alterations of microglia in the brains of patients with hereditary diffuse leukoencephalopathy with spheroids (HDLS) caused by mutations of the gene encoding the colony stimulating factor-1 receptor (CSF-1R). METHODS: We examined 5 autopsied brains and 1 biopsy specimen from a total of 6 patients with CSF-1R mutations. Detailed immunohistochemical, biochemical, and ultrastructural features of microglia were examined, and quantitative analyses were performed. RESULTS: In layers 3 to 4 of the frontal cortex in HDLS brains, microglia showed relatively uniform and delicate morphology, with thin and winding processes accompanying knotlike structures, and significantly smaller areas of Iba1 immunoreactivity and lower numbers of Iba1-positive cells were evident in comparison with control brains. On the other hand, in layers 5 to 6 and the underlying white matter, microglia were distributed unevenly; that is, in some areas they had accumulated densely, whereas in others they were scattered. Immunoblot analyses of microglia-associated proteins, including CD11b and DAP12, revealed that HDLS brains had significantly lower amounts of these proteins than diseased controls, although Ki-67-positive proliferative microglia were not reduced. Ultrastructurally, the microglial cytoplasm and processes in HDLS showed vesiculation of the rough endoplasmic reticulum and disaggregated polyribosomes, indicating depression of protein synthesis. On the other hand, macrophages were immunonegative for GLUT-5 or P2ry12, indicating that they were derived from bone marrow. INTERPRETATION: The pathogenesis of HDLS seems to be associated with microglial vulnerability and morphological alterations. Ann Neurol 2016;80:554-565.


Assuntos
Córtex Cerebelar/patologia , Lobo Frontal/patologia , Leucoencefalopatias/patologia , Microglia/patologia , Substância Branca/patologia , Autopsia , Biópsia , Humanos , Leucoencefalopatias/metabolismo , Microglia/ultraestrutura , Receptor de Fator Estimulador de Colônias de Macrófagos/genética
8.
Nat Commun ; 6: 10232, 2015 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-26659141

RESUMO

The thalamocortical tract carries sensory information to the neocortex. It has long been recognized that the neocortical pioneer axons of subplate neurons are essential for thalamocortical development. Herein we report that an axon guidance cue, draxin, is expressed in early-born neocortical neurons, including subplate neurons, and is necessary for thalamocortical development. In draxin(-/-) mice, thalamocortical axons do not enter the neocortex. This phenotype is sufficiently rescued by the transgenic expression of draxin in neocortical neurons. Genetic interaction data suggest that draxin acts through Deleted in colorectal cancer (DCC) and Neogenin (Neo1), to regulate thalamocortical projections in vivo. Draxin promotes the outgrowth of thalamic axons in vitro and this effect is abolished in thalamic neurons from Dcc and Neo1 double mutants. These results suggest that draxin from neocortical neurons controls thalamocortical projections into the neocortex, and that this effect is mediated through the DCC and Neo1 receptors.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neocórtex/fisiologia , Animais , Axônios/fisiologia , Feminino , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neocórtex/citologia , Neurônios/fisiologia , Tálamo/citologia , Tálamo/fisiologia
9.
Ann Neurol ; 78(3): 375-86, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26018084

RESUMO

OBJECTIVE: Focal cortical dysplasia (FCD) type IIb is a cortical malformation characterized by cortical architectural abnormalities, dysmorphic neurons, and balloon cells. It has been suggested that FCDs are caused by somatic mutations in cells in the developing brain. Here, we explore the possible involvement of somatic mutations in FCD type IIb. METHODS: We collected a total of 24 blood-brain paired samples with FCD, including 13 individuals with FCD type IIb, 5 with type IIa, and 6 with type I. We performed whole-exome sequencing using paired samples from 9 of the FCD type IIb subjects. Somatic MTOR mutations were identified and further investigated using all 24 paired samples by deep sequencing of the entire gene's coding region. Somatic MTOR mutations were confirmed by droplet digital polymerase chain reaction. The effect of MTOR mutations on mammalian target of rapamycin (mTOR) kinase signaling was evaluated by immunohistochemistry and Western blotting analyses of brain samples and by in vitro transfection experiments. RESULTS: We identified four lesion-specific somatic MTOR mutations in 6 of 13 (46%) individuals with FCD type IIb showing mutant allele rates of 1.11% to 9.31%. Functional analyses showed that phosphorylation of ribosomal protein S6 in FCD type IIb brain tissues with MTOR mutations was clearly elevated, compared to control samples. Transfection of any of the four MTOR mutants into HEK293T cells led to elevated phosphorylation of 4EBP, the direct target of mTOR kinase. INTERPRETATION: We found low-prevalence somatic mutations in MTOR in FCD type IIb, indicating that activating somatic mutations in MTOR cause FCD type IIb.


Assuntos
Encéfalo/patologia , Malformações do Desenvolvimento Cortical do Grupo II/genética , Mutação/genética , Serina-Treonina Quinases TOR/genética , Adolescente , Adulto , Criança , Feminino , Células HEK293 , Humanos , Masculino , Malformações do Desenvolvimento Cortical/diagnóstico , Malformações do Desenvolvimento Cortical/genética , Malformações do Desenvolvimento Cortical do Grupo II/diagnóstico
10.
J Neurosci Res ; 91(2): 178-86, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23172790

RESUMO

In chronic demyelinating lesions of the central nervous system, insufficient generation of oligodendrocytes (OLs) is not due to a lack of oligodendrocyte precursor cells (OPCs), because the accumulation of OPCs and premyelinating OLs can be observed within these lesions. Here we sought to identify the basis for the failure of OLs to achieve terminal differentiation in chronic demyelinating lesions through the utilization of plp1-overexpressing (Plp(tg/-)) mice. These mice are characterized by progressive demyelination in young adults and chronic demyelinating lesions at more mature stages. We show that neural stem cells, which are the precursors of OL-lineage cells, are present in the Plp(tg/-) mouse brain and that their multipotentiality and ability to self-renew are comparable to those of wild-type adults in culture. Lineage-tracing experiments using a transgenic mouse line, in which an inducible Cre recombinase is knocked in at the Olig2 locus, revealed that Olig2-lineage cells preferentially differentiated into OPCs and premyelinating OLs, but not into astrocytes, in the Plp(tg/-) mouse brain. These Olig2-lineage cells matured to express myelin basic protein but after that their processes degenerated in the chronic demyelinating lesions of the Plp(tg/-) brain. These results indicate that in chronic demyelinated lesions more OL-lineage cells are produced as part of the repair process, but their processes degenerate after maturation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem da Célula/fisiologia , Doenças Desmielinizantes/fisiopatologia , Proteína Básica da Mielina/metabolismo , Proteína Proteolipídica de Mielina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oligodendroglia/fisiologia , Fatores Etários , Animais , Antineoplásicos Hormonais/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Encéfalo/patologia , Diferenciação Celular , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/genética , Proteínas do Tecido Nervoso/genética , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/fisiologia , Fator de Transcrição 2 de Oligodendrócitos , Tamoxifeno/farmacologia
11.
Nat Neurosci ; 15(5): 700-2, 2012 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-22446882

RESUMO

Adult hypothalamic neurogenesis has recently been reported, but the cell of origin and the function of these newborn neurons are unknown. Using genetic fate mapping, we found that median eminence tanycytes generate newborn neurons. Blocking this neurogenesis altered the weight and metabolic activity of adult mice. These findings reveal a previously unreported neurogenic niche in the mammalian hypothalamus with important implications for metabolism.


Assuntos
Dieta Hiperlipídica , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Eminência Mediana/citologia , Neurogênese/fisiologia , Nicho de Células-Tronco/fisiologia , Fatores Etários , Animais , Animais Recém-Nascidos , Proteínas de Bactérias/genética , Peso Corporal/fisiologia , Bromodesoxiuridina/metabolismo , Contagem de Células , Proliferação de Células , Proteínas ELAV/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Histonas/metabolismo , Proteínas de Filamentos Intermediários/metabolismo , Proteínas Luminescentes/genética , Espectroscopia de Ressonância Magnética , Eminência Mediana/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Gravidez , Proteínas/genética , Proteínas/metabolismo , RNA não Traduzido , Radiação , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/genética , Fatores de Transcrição SOXB1/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia
12.
Nat Neurosci ; 15(3): 373-80, S1-2, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22246438

RESUMO

In brain development, distinct types of migration, radial migration and tangential migration, are shown by excitatory and inhibitory neurons, respectively. Whether these two types of migration operate by similar cellular mechanisms remains unclear. We examined neuronal migration in mice deficient in mDia1 (also known as Diap1) and mDia3 (also known as Diap2), which encode the Rho-regulated actin nucleators mammalian diaphanous homolog 1 (mDia1) and mDia3. mDia deficiency impaired tangential migration of cortical and olfactory inhibitory interneurons, whereas radial migration and consequent layer formation of cortical excitatory neurons were unaffected. mDia-deficient neuroblasts exhibited reduced separation of the centrosome from the nucleus and retarded nuclear translocation. Concomitantly, anterograde F-actin movement and F-actin condensation at the rear, which occur during centrosomal and nuclear movement of wild-type cells, respectively, were impaired in mDia-deficient neuroblasts. Blockade of Rho-associated protein kinase (ROCK), which regulates myosin II, also impaired nuclear translocation. These results suggest that Rho signaling via mDia and ROCK critically regulates nuclear translocation through F-actin dynamics in tangential migration, whereas this mechanism is dispensable in radial migration.


Assuntos
Proteínas de Transporte/metabolismo , Movimento Celular/fisiologia , Interneurônios/fisiologia , Ventrículos Laterais/citologia , Células-Tronco Neurais/fisiologia , Actinas/metabolismo , Amidas/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Proteínas de Transporte/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Movimento Celular/genética , Desoxiuridina/análogos & derivados , Proteínas do Domínio Duplacortina , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Proteínas da Matriz Extracelular/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Forminas , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Corpos Geniculados/citologia , Corpos Geniculados/embriologia , Corpos Geniculados/crescimento & desenvolvimento , Glutamato Descarboxilase/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Homeodomínio/metabolismo , Ventrículos Laterais/embriologia , Ventrículos Laterais/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/metabolismo , Proteínas Nucleares/metabolismo , Técnicas de Cultura de Órgãos , Parvalbuminas/metabolismo , Transporte Proteico/genética , Piridinas/farmacologia , Proteína Reelina , Proteínas Repressoras/metabolismo , Serina Endopeptidases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Tempo , Proteínas Supressoras de Tumor/metabolismo , Ácido gama-Aminobutírico/metabolismo , Quinases Associadas a rho/metabolismo
13.
PLoS One ; 7(12): e51581, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23284718

RESUMO

Heterogeneous classes of neurons are present in the spinal cord and are essential for its function. Expression patterns of transcription factors in neural progenitor cells determine neuron subtypes during development. Nkx2.2 is expressed in the progenitor cell pool located just ventrally to the Olig2-positive pool and is indispensable for V3 interneuron generation in the spinal cord and also for visceral motoneuron generation in the hindbrain. However, whether Nkx2.2-positive progenitor cells generate diverse classes of neuron is not fully understood. Using a chick lineage tracing method in a genetically-defined manner, we found that Nkx2.2-expressing progenitor cells differentiate into general visceral motoneurons as well as sim1-positive V3 interneurons. Surprisingly, we further observed that Nkx2.2-expressing progenitors differentiate into somatic motoneuron. Our findings suggest that the different classes of motoneurons are derived from more complex sources than were previously expected in the chick spinal cord.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Interneurônios/citologia , Neurônios Motores/citologia , Medula Espinal/citologia , Células-Tronco/citologia , Fatores de Transcrição/metabolismo , Animais , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Diferenciação Celular , Linhagem da Célula , Embrião de Galinha , Proteína Homeobox Nkx-2.2 , Técnicas Imunoenzimáticas , Hibridização In Situ , Integrases/metabolismo , Interneurônios/metabolismo , Camundongos , Neurônios Motores/metabolismo , Retroviridae/genética , Medula Espinal/metabolismo , Células-Tronco/metabolismo , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Proteínas de Peixe-Zebra
14.
PLoS One ; 6(9): e25465, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21980468

RESUMO

During development of the central nervous system, the apical-basal polarity of neuroepithelial cells is critical for homeostasis of proliferation and differentiation of neural stem cells. While adherens junctions at the apical surface of neuroepithelial cells are important for maintaining the polarity, the molecular mechanism regulating integrity of these adherens junctions remains largely unknown. Given the importance of actin cytoskeleton in adherens junctions, we have analyzed the role of mDia, an actin nucleator and a Rho effector, in the integrity of the apical adherens junction. Here we show that mDia1 and mDia3 are expressed in the developing brain, and that mDia3 is concentrated in the apical surface of neuroepithelium. Mice deficient in both mDia1 and mDia3 develop periventricular dysplastic mass widespread throughout the developing brain, where neuroepithelial cell polarity is impaired with attenuated apical actin belts and loss of apical adherens junctions. In addition, electron microscopic analysis revealed abnormal shrinkage and apical membrane bulging of neuroepithelial cells in the remaining areas. Furthermore, perturbation of Rho, but not that of ROCK, causes loss of the apical actin belt and adherens junctions similarly to mDia-deficient mice. These results suggest that actin cytoskeleton regulated by Rho-mDia pathway is critical for the integrity of the adherens junctions and the polarity of neuroepithelial cells, and that loss of this signaling induces aberrant, ectopic proliferation and differentiation of neural stem cells.


Assuntos
Actinas/metabolismo , Proteínas de Transporte/metabolismo , Ventrículos Cerebrais/anormalidades , Ventrículos Cerebrais/patologia , Células Neuroepiteliais/metabolismo , Células Neuroepiteliais/patologia , Junções Aderentes/metabolismo , Junções Aderentes/patologia , Animais , Proteínas de Transporte/genética , Diferenciação Celular/genética , Polaridade Celular/genética , Proliferação de Células , Ventrículos Cerebrais/embriologia , Ventrículos Cerebrais/metabolismo , Líquido Cefalorraquidiano/fisiologia , Feminino , Forminas , Deleção de Genes , Hidrocefalia/etiologia , Hidrocefalia/metabolismo , Hidrocefalia/patologia , Masculino , Camundongos , Células NIH 3T3 , Proteínas rho de Ligação ao GTP/metabolismo
15.
Dev Biol ; 349(2): 504-11, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20951692

RESUMO

In the spinal cord, generation of oligodendrocytes (OLs) is totally dependent on the presence of Olig2, a basic helix-loop-helix transcription factor. However, it also requires Nkx2.2 for its generation, whose expression follows the expression of Olig2. Although it is believed that oligodendrocytes originate from the pMN domain, Nkx2.2 is present in the p3 domain located ventral to the pMN domain. According to recent reports, it is possible that oligodendrocytes are directly derived from the p3 domain in addition to the pMN domain in the chick spinal cord. We examined this hypothesis in this paper. To analyze OL development in the spinal cord, chick embryos are widely used for genetic modification by electroporation or for transplantation experiments, because it is relatively easy to manipulate them compared with mouse embryos. However, genetic modification by electroporation is not appropriate for glial development analyses because glia proliferate vigorously before maturation. In order to overcome these problems, we established a novel method to permanently introduce exogenous gene into a specific cell type. We introduced the CAT1 gene, a murine retroviral receptor, by electroporation followed by injection of murine retrovirus. By using this method, we successfully transduced murine retrovirus into the chick neural tube. We analyzed cell lineage from the p3 domain by restricting CAT1 expression by Nkx2.2-enhancer and found that most of the labeled cells became OLs when the cells were labeled at cE4. Moreover, the labeled OLs were found throughout the white matter in the spinal cord including the most dorsal spinal cord. Thus p3 domain directly generates spinal cord OLs in the chick spinal cord.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Proteínas de Homeodomínio/metabolismo , Oligodendroglia/citologia , Medula Espinal/embriologia , Fatores de Transcrição/metabolismo , Animais , Canais de Cálcio/metabolismo , Embrião de Galinha , Eletroporação , Proteína Homeobox Nkx-2.2 , Imuno-Histoquímica , Hibridização In Situ , Receptores Virais/metabolismo , Retroviridae , Medula Espinal/citologia , Canais de Cátion TRPV/metabolismo , Proteínas de Peixe-Zebra
16.
Genesis ; 46(12): 775-81, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19105217

RESUMO

Tamoxifen-inducible Cre (CreER) has become a powerful tool for in vivo manipulation of the genome. Here, we investigated opposing effects of tamoxifen on apoptosis during embryogenesis using Olig2-CreER knock-in mice, namely, tamoxifen-induced apoptosis through CreER-mediated toxicity and cytoprotective activity of tamoxifen independent of CreER. First, we examined tamoxifen-induced apoptosis; in the homozygous mice, we observed region-specific apoptosis in the ventral neural tube, with no obvious increase in the heterozygotes. Next, we detected a cytoprotective effect on apoptosis in the homozygous dorsal root ganglia (DRG). This apoptosis is a secondary phenotype of Olig2-null mice, as Olig2/CreER is not expressed in the DRG. The cytoprotective effect is DRG-specific, because tamoxifen did not rescue apoptosis in the interdigital mesenchyme. These data indicate that tamoxifen has multiple effects on apoptosis during development and caution that careful examination is necessary when interpreting results obtained from tamoxifen-induced recombination: in Olig2-CreER mice, heterozygotes are usable for lineage-tracing experiment without obvious toxicity, while homozygotes show efficient recombination, despite enhanced apoptosis.


Assuntos
Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Integrases/metabolismo , Tamoxifeno/farmacologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Gânglios Espinais/citologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Fator de Transcrição 2 de Oligodendrócitos
17.
J Neurosci Res ; 86(16): 3494-502, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18816798

RESUMO

Olig2 is a basic helix-loop-helix (bHLH) transcription factor essential for development of motoneurons and oligodendrocytes. It is known that Olig2(+) cells persist in the central nervous system (CNS) from embryonic to adult stages and that the number of Olig2(+) progenitors increases in the injured adult CNS. Recent studies have demonstrated an inhibitory action of Olig2 on neurogenesis in adult CNS, but the fate of Olig2(+) cells in the injured state remains largely unknown. To trace directly the fate of Olig2 cells in the adult cerebral cortex after injury, we employed the CreER/loxP system to target the olig2 locus. In this genetic tracing study, green fluorescent protein (GFP) reporter-positive cells labeled after cryoinjury coexpressed glial fibrillary acidic protein (GFAP), an astrocytic marker. Electron microscopy also showed that GFP(+) cells have the ultrastructural characteristics of astrocytes. Furthermore, GFP(+) cells labeled before injury, most of which had been NG2 cells, also produced bushy astrocytes. Here we show direct evidence that Olig2(+) cells preferentially differentiate into astrocytes, which strongly express GFAP, in response to injury in the adult cerebral cortex. These results suggest that reactive astrocytes, known to be the main contributors to glial scars, originate, at least in part, from Olig2(+) cells.


Assuntos
Astrócitos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Lesões Encefálicas/genética , Linhagem da Célula/genética , Gliose/genética , Proteínas do Tecido Nervoso/genética , Células-Tronco/metabolismo , Animais , Antígenos/metabolismo , Astrócitos/ultraestrutura , Biomarcadores/análise , Biomarcadores/metabolismo , Lesões Encefálicas/metabolismo , Lesões Encefálicas/fisiopatologia , Diferenciação Celular/genética , Mapeamento Cromossômico/métodos , Cicatriz/genética , Cicatriz/metabolismo , Cicatriz/fisiopatologia , Criocirurgia/métodos , Técnicas de Introdução de Genes , Genes Reporter/genética , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/metabolismo , Gliose/fisiopatologia , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Fator de Transcrição 2 de Oligodendrócitos , Proteoglicanas/metabolismo , Células-Tronco/ultraestrutura
18.
J Neurochem ; 100(1): 12-22, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17064358

RESUMO

Cystatin C (CysC) is an endogenous cysteine proteases inhibitor produced by mature astrocytes in the adult brain. Previously we isolated CysC as a factor activating the glial fibrillary acidic protein (GFAP) promoter, and showed that CysC is expressed in astrocyte progenitors during development. Here we show that protease inhibitor activity increased daily in conditioned medium, and that this activity was mainly a result of CysC released from primary cultured cells. Human CysC added to the culture medium of primary brain cells increased the number of GFAP-positive and nestin-positive cells. Human CysC also increased the number of neurospheres formed from embryonic brain, and thus it increases the number of neural stem/precursor cells in a manner similar to glycosylated rat CysC. The addition of a neutralizing antibody, on the other hand, greatly decreased the number of GFAP and glutamate aspartate transporter (GLAST)-positive astrocytes. This decrease was reversed by the addition of CysC but not by another cysteine protease inhibitor. Thus, the promotion of astrocyte development by CysC appears to be independent of its protease inhibitor activity. The antibody increased the number of oligodendrocytes and their precursors. Therefore, CysC modifies glial development in addition to its activity against neural stem/precursor cells.


Assuntos
Cistatinas/farmacologia , Neuroglia/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Fatores Etários , Animais , Anticorpos/farmacologia , Contagem de Células/métodos , Córtex Cerebral/citologia , Cistatina C , Cistatinas/imunologia , Embrião de Mamíferos , Transportador 1 de Aminoácido Excitatório/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica/métodos , Indóis , Proteínas de Filamentos Intermediários/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neuroglia/fisiologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fatores de Tempo
19.
Am J Pathol ; 164(5): 1717-25, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15111318

RESUMO

Olig2 is a recently identified transcription factor involved in the phenotype definition of cells in the oligodendroglial lineage. The expression of Olig2 transcript has been demonstrated in human oligodendroglial tumors, although the protein expression has not been studied extensively. We developed a polyclonal antibody to human Olig2 and analyzed it immunohistochemically. The antibody depicted a single distinct band of predicted molecular weight by Western blotting, and did not cross-react with human Olig1. In normal human brain tissue, the nuclei of oligodendrocytes of interfascicular, perivascular, and perineuronal disposition were clearly labeled by the antibody. Similarly, the nuclei of oligodendroglial tumors were labeled. There was no apparent correlation between the staining intensity and histological grade. Astrocytic components within the tumors were generally less or not stained. Astrocytic tumors were also positive with the Olig2 antiserum to a lesser extent, and the difference between oligodendroglial and astrocytic tumors was demonstrated by a statistical analysis. Olig2 and glial fibrillary acidic protein were expressed in a mutually exclusive manner, and Olig2 expression was cell-cycle related. Neither central neurocytoma nor schwannoma cases were stained. Our antibody was demonstrated to be useful in recognizing normal oligodendrocytes on paraffin sections, and applicable in diagnosis of some brain tumors.


Assuntos
Glioma/patologia , Imuno-Histoquímica/métodos , Proteínas do Tecido Nervoso/química , Oligodendroglia/patologia , Animais , Anticorpos/química , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Ciclo Celular , Linhagem da Célula , Núcleo Celular/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Glioma/imunologia , Humanos , Camundongos , Células NIH 3T3 , Proteínas do Tecido Nervoso/imunologia , Fator de Transcrição 2 de Oligodendrócitos , Oligodendroglia/imunologia , Parafina/metabolismo
20.
Development ; 130(24): 6027-35, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14573516

RESUMO

Drosophila glial cells missing (gcm) is a key gene that determines the fate of stem cells within the nervous system. Two mouse gcm homologs have been identified, but their function in the nervous system remains to be elucidated. To investigate their function, we constructed retroviral vectors harboring Drosophila gcm and two mouse Gcm genes. Expression of these genes appeared to influence fibroblast features. In particular, mouse Gcm1 induced the expression of astrocyte-specific Ca(2+)-binding protein, S100beta, in those cells. Introduction of the mouse Gcm1 gene in cultured cells from embryonic brains resulted in the induction of an astrocyte lineage. This effect was also observed by in utero injection of retrovirus harboring mouse Gcm1 into the embryonic brain. However, cultures from mouse Gcm1-deficient mouse brains did not exhibit significant reductions in the number of astrocytes. Furthermore, in situ hybridization analysis of mouse Gcm1 mRNA revealed distinct patterns of expression in comparison with other well-known glial markers. The mammalian homolog of Drosophila gcm, mouse Gcm1, exhibits the potential to induce gliogenesis, but may function in the generation of a minor subpopulation of glial cells.


Assuntos
Diferenciação Celular/fisiologia , Drosophila melanogaster/fisiologia , Neuroglia/fisiologia , Neuropeptídeos/metabolismo , Transativadores/metabolismo , Animais , Células Cultivadas , Proteínas de Ligação a DNA , Proteínas de Drosophila , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hibridização In Situ , Camundongos , Camundongos Knockout , Neuroglia/citologia , Neurônios/citologia , Neurônios/metabolismo , Neuropeptídeos/genética , Proteínas Nucleares , Retroviridae/genética , Retroviridae/metabolismo , Transativadores/genética , Fatores de Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA