Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 78(1): 287-298, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32189007

RESUMO

Unveiling the key pathways underlying postnatal beta-cell proliferation can be instrumental to decipher the mechanisms of beta-cell mass plasticity to increased physiological demand of insulin during weight gain and pregnancy. Using transcriptome and global Serine Threonine Kinase activity (STK) analyses of islets from newborn (10 days old) and adult rats, we found that highly proliferative neonatal rat islet cells display a substantially elevated activity of the mitogen activated protein 3 kinase 12, also called dual leucine zipper-bearing kinase (Dlk). As a key upstream component of the c-Jun amino terminal kinase (Jnk) pathway, Dlk overexpression was associated with increased Jnk3 activity and was mainly localized in the beta-cell cytoplasm. We provide the evidence that Dlk associates with and activates Jnk3, and that this cascade stimulates the expression of Ccnd1 and Ccnd2, two essential cyclins controlling postnatal beta-cell replication. Silencing of Dlk or of Jnk3 in neonatal islet cells dramatically hampered primary beta-cell replication and the expression of the two cyclins. Moreover, the expression of Dlk, Jnk3, Ccnd1 and Ccnd2 was induced in high replicative islet beta cells from ob/ob mice during weight gain, and from pregnant female rats. In human islets from non-diabetic obese individuals, DLK expression was also cytoplasmic and the rise of the mRNA level was associated with an increase of JNK3, CCND1 and CCND2 mRNA levels, when compared to islets from lean and obese patients with diabetes. In conclusion, we find that activation of Jnk3 signalling by Dlk could be a key mechanism for adapting islet beta-cell mass during postnatal development and weight gain.


Assuntos
Células Secretoras de Insulina/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Transdução de Sinais , Animais , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina D2/genética , Ciclina D2/metabolismo , Feminino , Glucose/farmacologia , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 10 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 10 Ativada por Mitógeno/genética , Obesidade/metabolismo , Obesidade/patologia , Pâncreas/crescimento & desenvolvimento , Pâncreas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
2.
Lab Invest ; 99(2): 210-230, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30413814

RESUMO

In order to understand the pathobiology of neurotrophic keratopathy, we established a mouse model by coagulating the first branch of the trigeminal nerve (V1 nerve). In our model, the sensory nerve in the central cornea disappeared and remaining fibers were sparse in the peripheral limbal region. Impaired corneal epithelial healing in the mouse model was associated with suppression of both cell proliferation and expression of stem cell markers in peripheral/limbal epithelium as well as a reduction of transient receptor potential vanilloid 4 (TRPV4) expression in tissue. TRPV4 gene knockout also suppressed epithelial repair in mouse cornea, although it did not seem to directly modulate migration of epithelium. In a co-culture experiment, TRPV4-introduced KO trigeminal ganglion upregulated nerve growth factor (NGF) in cultured corneal epithelial cells, but ganglion with a control vector did not. TRPV4 gene introduction into a damaged V1 nerve rescues the impairment of epithelial healing in association with partial recovery of the stem/progenitor cell markers and upregulation of cell proliferation and of NGF expression in the peripheral/limbal epithelium. Gene transfer of TRPV4 did not accelerate the regeneration of nerve fibers. Sensory nerve TRPV4 is critical to maintain stemness of peripheral/limbal basal cells, and is one of the major mechanisms of homeostasis maintenance of corneal epithelium.


Assuntos
Epitélio Corneano , Células-Tronco , Canais de Cátion TRPV/metabolismo , Nervo Trigêmeo/metabolismo , Cicatrização/fisiologia , Animais , Células Cultivadas , Epitélio Corneano/citologia , Epitélio Corneano/lesões , Epitélio Corneano/metabolismo , Técnicas de Inativação de Genes , Camundongos , Células-Tronco/citologia , Células-Tronco/metabolismo , Canais de Cátion TRPV/genética , Nervo Trigêmeo/química
3.
Proc Natl Acad Sci U S A ; 110(10): 4045-50, 2013 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23431148

RESUMO

Glaucoma, a major cause of blindness worldwide, is a neurodegenerative optic neuropathy in which vision loss is caused by loss of retinal ganglion cells (RGCs). To better define the pathways mediating RGC death and identify targets for the development of neuroprotective drugs, we developed a high-throughput RNA interference screen with primary RGCs and used it to screen the full mouse kinome. The screen identified dual leucine zipper kinase (DLK) as a key neuroprotective target in RGCs. In cultured RGCs, DLK signaling is both necessary and sufficient for cell death. DLK undergoes robust posttranscriptional up-regulation in response to axonal injury in vitro and in vivo. Using a conditional knockout approach, we confirmed that DLK is required for RGC JNK activation and cell death in a rodent model of optic neuropathy. In addition, tozasertib, a small molecule protein kinase inhibitor with activity against DLK, protects RGCs from cell death in rodent glaucoma and traumatic optic neuropathy models. Together, our results establish a previously undescribed drug/drug target combination in glaucoma, identify an early marker of RGC injury, and provide a starting point for the development of more specific neuroprotective DLK inhibitors for the treatment of glaucoma, nonglaucomatous forms of optic neuropathy, and perhaps other CNS neurodegenerations.


Assuntos
MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/fisiologia , Células Ganglionares da Retina/enzimologia , Células Ganglionares da Retina/patologia , Animais , Morte Celular/genética , Morte Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo , Glaucoma/tratamento farmacológico , Glaucoma/etiologia , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , Masculino , Camundongos , Doenças do Nervo Óptico/etiologia , Doenças do Nervo Óptico/patologia , Traumatismos do Nervo Óptico/tratamento farmacológico , Traumatismos do Nervo Óptico/enzimologia , Traumatismos do Nervo Óptico/patologia , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Ratos , Ratos Wistar , Células Ganglionares da Retina/efeitos dos fármacos , Transdução de Sinais , Regulação para Cima
4.
BMC Cell Biol ; 14: 12, 2013 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-23496950

RESUMO

BACKGROUND: The regulatory mechanisms of motor protein-dependent intracellular transport are still not fully understood. The kinesin-1-binding protein, JIP1, can function as an adaptor protein that links kinesin-1 and other JIP1-binding "cargo" proteins. However, it is unknown whether these "cargo" proteins influence the JIP1-kinesin-1 binding. RESULTS: We show here that JIP1-kinesin-1 binding in Neuro2a cells was dependent on conserved amino acid residues in the JIP1-phosphotyrosine binding (PTB) domain, including F687. In addition, mutation of F687 severely affected the neurite tip localization of JIP1. Proteomic analysis revealed another kinesin-1 binding protein, JIP3, as a major JIP1 binding protein. The association between JIP1 and JIP3 was dependent on the F687 residue in JIP1, and this association induced the formation of a stable ternary complex with kinesin-1. On the other hand, the binding of JIP1 and JIP3 was independent of kinesin-1 binding. We also show that other PTB binding proteins can interrupt the formation of the ternary complex. CONCLUSIONS: The formation of the JIP1-kinesin-1 complex depends on the protein binding-status of the JIP1 PTB domain. This may imply a regulatory mechanism of kinesin-1-dependent intracellular transport.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cinesinas/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/química , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/química , Animais , Linhagem Celular Tumoral , Células HEK293 , Humanos , Camundongos , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteômica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética
5.
Am J Hum Genet ; 86(6): 881-91, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20493457

RESUMO

A de novo 9q33.3-q34.11 microdeletion involving STXBP1 has been found in one of four individuals (group A) with early-onset West syndrome, severe hypomyelination, poor visual attention, and developmental delay. Although haploinsufficiency of STXBP1 was involved in early infantile epileptic encephalopathy in a previous different cohort study (group B), no mutations of STXBP1 were found in two of the remaining three subjects of group A (one was unavailable). We assumed that another gene within the deletion might contribute to the phenotype of group A. SPTAN1 encoding alpha-II spectrin, which is essential for proper myelination in zebrafish, turned out to be deleted. In two subjects, an in-frame 3 bp deletion and a 6 bp duplication in SPTAN1 were found at the initial nucleation site of the alpha/beta spectrin heterodimer. SPTAN1 was further screened in six unrelated individuals with WS and hypomyelination, but no mutations were found. Recombinant mutant (mut) and wild-type (WT) alpha-II spectrin could assemble heterodimers with beta-II spectrin, but alpha-II (mut)/beta-II spectrin heterodimers were thermolabile compared with the alpha-II (WT)/beta-II heterodimers. Transient expression in mouse cortical neurons revealed aggregation of alpha-II (mut)/beta-II and alpha-II (mut)/beta-III spectrin heterodimers, which was also observed in lymphoblastoid cells from two subjects with in-frame mutations. Clustering of ankyrinG and voltage-gated sodium channels at axon initial segment (AIS) was disturbed in relation to the aggregates, together with an elevated action potential threshold. These findings suggest that pathological aggregation of alpha/beta spectrin heterodimers and abnormal AIS integrity resulting from SPTAN1 mutations were involved in pathogenesis of infantile epilepsy.


Assuntos
Deficiências do Desenvolvimento/genética , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Células Cultivadas , Humanos , Lactente , Camundongos , Dados de Sequência Molecular , Bainha de Mielina/metabolismo , Fenótipo , Quadriplegia/genética , Espasmos Infantis/genética , Espectrina/genética , Transfecção
6.
J Neurosci ; 31(17): 6468-80, 2011 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-21525288

RESUMO

Studies using cultured neurons have established the critical role of microtubule regulators in neuronal polarization. The c-Jun N-terminal kinase (JNK) pathway is one of the candidate signaling pathways driving microtubule regulation during neuronal polarization. However, the significance of the JNK pathway in axon formation, a fundamental step in neuronal polarization, in vivo, remains unclear. Here, we provide evidence supporting the notion that the JNK pathway contributes to axon formation, in vivo, by identifying the genetic interactions between mouse JNK1 and dual leucine zipper kinase (DLK). Double mutants exhibited severe defects in axon formation in the cerebral neocortex. Moreover, RNA interference rescue experiments, in vitro, showed that DLK and JNK1 function in a common pathway to support neuronal polarization by promoting short-neurite and axon formation. Defects in axon formation caused by perturbations of the DLK-JNK pathway were significantly improved by Taxol. However, defects in short-neurite formation caused by perturbations of the DLK-JNK pathway were enhanced by Taxol. Together, these in vivo and in vitro observations indicate that the DLK-JNK pathway facilitates axon formation in neocortical neurons via stage-specific regulation of microtubule stability.


Assuntos
Axônios/fisiologia , MAP Quinase Quinase Quinases/metabolismo , Microtúbulos/fisiologia , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Neocórtex/citologia , Neurônios/citologia , Animais , Anticorpos Monoclonais/metabolismo , Axônios/efeitos dos fármacos , Polaridade Celular/genética , Células Cultivadas , Interações Medicamentosas , Embrião não Mamífero , Inibidores Enzimáticos/farmacologia , Feminino , Gânglios Espinais/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde/genética , Técnicas In Vitro , MAP Quinase Quinase Quinases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microtúbulos/efeitos dos fármacos , Proteína Quinase 8 Ativada por Mitógeno/genética , Modelos Biológicos , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Proteínas de Neurofilamentos/metabolismo , Paclitaxel/farmacologia , Estimulação Física , Interferência de RNA/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Transfecção/métodos , Moduladores de Tubulina/farmacologia
7.
J Neurosci ; 31(34): 12094-103, 2011 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-21865452

RESUMO

Dendritic spines are postsynaptic structures that receive excitatory synaptic input from presynaptic terminals. Actin and its regulatory proteins play a central role in morphogenesis of dendritic spines. In addition, recent studies have revealed that microtubules are indispensable for the maintenance of mature dendritic spine morphology by stochastically invading dendritic spines and regulating dendritic localization of p140Cap, which is required for actin reorganization. However, the regulatory mechanisms of microtubule dynamics remain poorly understood. Partitioning-defective 1b (PAR1b), a cell polarity-regulating serine/threonine protein kinase, is thought to regulate microtubule dynamics by inhibiting microtubule binding of microtubule-associated proteins. Results from the present study demonstrated that PAR1b participates in the maintenance of mature dendritic spine morphology in mouse hippocampal neurons. Immunofluorescent analysis revealed PAR1b localization in the dendrites, which was concentrated in dendritic spines of mature neurons. PAR1b knock-down cells exhibited decreased mushroom-like dendritic spines, as well as increased filopodia-like dendritic protrusions, with no effect on the number of protrusions. Live imaging of microtubule plus-end tracking proteins directly revealed decreases in distance and duration of microtubule growth following PAR1b knockdown in a neuroblastoma cell line and in dendrites of hippocampal neurons. In addition, reduced accumulation of GFP-p140Cap in dendritic protrusions was confirmed in PAR1b knock-down neurons. In conclusion, the present results suggested a novel function for PAR1b in the maintenance of mature dendritic spine morphology by regulating microtubule growth and the accumulation of p140Cap in dendritic spines.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Diferenciação Celular/genética , Espinhas Dendríticas/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Microtúbulos/ultraestrutura , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Forma Celular/fisiologia , Células Cultivadas , Espinhas Dendríticas/ultraestrutura , Feminino , Masculino , Camundongos , Camundongos Endogâmicos ICR , Microtúbulos/metabolismo , Neurônios/ultraestrutura , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética
8.
J Neurosci ; 26(51): 13357-62, 2006 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-17182786

RESUMO

Collapsin response mediator protein 1 (CRMP1) is one of the CRMP family members that mediates signal transduction of axon guidance molecules. Here, we show evidence that CRMP1 is involved in Reelin (Reln) signaling to regulate neuronal migration in the cerebral cortex. In crmp1-/- mice, radial migration of cortical neurons was retarded. This phenotype was not observed in the sema3A-/- and crmp1+/-;sema3A+/- cortices. However, CRMP1 was colocalized with disabled-1 (Dab1), an adaptor protein in Reln signaling. In the Reln(rl/rl) cortex, CRMP1 and Dab1 were expressed at a higher level, yet tyrosine phosphorylated at a lower level. Loss of crmp1 in a dab1 heterozygous background led to the disruption of hippocampal lamination, a Reeler-like phenotype. In addition to axon guidance, CRMP1 regulates neuronal migration by mediating Reln signaling.


Assuntos
Moléculas de Adesão Celular Neuronais/fisiologia , Movimento Celular/fisiologia , Córtex Cerebral/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Fosfoproteínas/fisiologia , Serina Endopeptidases/fisiologia , Transdução de Sinais/fisiologia , Animais , Moléculas de Adesão Celular Neuronais/genética , Linhagem Celular , Movimento Celular/genética , Córtex Cerebral/citologia , Proteínas da Matriz Extracelular/genética , Humanos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes Neurológicos , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Fosfoproteínas/genética , Proteína Reelina , Serina Endopeptidases/genética , Transdução de Sinais/genética
9.
J Neurosci ; 26(46): 11992-2002, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17108173

RESUMO

Mammalian corticogenesis substantially depends on migration and axon projection of newborn neurons that are coordinated by a yet unidentified molecular mechanism. Dual leucine zipper kinase (DLK) induces activation of c-Jun N-terminal kinase (JNK), a molecule that regulates morphogenesis in various organisms. We show here, using gene targeting in mice, that DLK is indispensable for establishing axon tracts, especially those originating from neocortical pyramidal neurons of the cerebrum. Direct and quantitative analysis of radial migration of pyramidal neurons using slice culture and a time-lapse imaging system revealed that acceleration around the subplate was affected by DLK gene disruption and by administration of a JNK inhibitor. Phosphorylation of JNK substrates, including c-Jun and doublecortin, and of JNK itself at the activation loop were partially affected in brains of DLK-deficient mouse embryos. These data suggest that DLK plays a significant role in the coordinated regulation of radial migration and axon projection by modulating JNK activity.


Assuntos
Movimento Celular/fisiologia , Córtex Cerebral/embriologia , Córtex Cerebral/enzimologia , Cones de Crescimento/enzimologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Animais , Diferenciação Celular/fisiologia , Córtex Cerebral/citologia , Quimera , Proteínas do Domínio Duplacortina , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Feminino , Cones de Crescimento/ultraestrutura , MAP Quinase Quinase Quinases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos ICR , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Vias Neurais/citologia , Vias Neurais/embriologia , Vias Neurais/enzimologia , Neuropeptídeos/metabolismo , Técnicas de Cultura de Órgãos , Proteínas Proto-Oncogênicas c-jun/metabolismo , Células Piramidais/citologia , Células Piramidais/enzimologia
10.
J Invest Dermatol ; 137(1): 132-141, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27519653

RESUMO

Dual leucine zipper-bearing kinase (DLK) is an inducer of keratinocyte differentiation, a complex process also involving microtubule reorganization to the cell periphery. However, signaling mechanisms involved in this process remain to be elucidated. Here, we demonstrate that DLK enhances and is required for microtubule reorganization to the cell periphery in human cell culture models and in Dlk knockout mouse embryos. In tissue-engineered skins with reduced DLK expression, cortical distribution of two microtubule regulators, LIS1 and HSP27, is impaired as well as desmosomal and tight junction integrity. Altered cortical distribution of desmosomal and tight junction proteins was also confirmed in Dlk knockout mouse embryos. Finally, desmosomal and tight junction defects were also observed after microtubule disruption in nocodazole-treated tissue-engineered skins, thus confirming a role for microtubules in the maintenance of these types of cell junctions. Globally, this study demonstrates that DLK is a key regulator of microtubule reorganization to the cell periphery during keratinocyte differentiation and that this process is required for the maintenance of desmosomal and tight junction integrity.


Assuntos
Diferenciação Celular/fisiologia , Desmossomos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Microtúbulos/metabolismo , Nocodazol/farmacologia , Junções Íntimas/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Imunofluorescência , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Microscopia Eletrônica de Transmissão , Microtúbulos/efeitos dos fármacos , Fosforilação , Interferência de RNA , Papel (figurativo) , Estatísticas não Paramétricas , Junções Íntimas/efeitos dos fármacos
11.
Gene Expr Patterns ; 5(4): 517-23, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15749080

RESUMO

C-Jun N-terminal kinase (JNK) is implicated in regulating the various cellular events during neural development that include differentiation, apoptosis and migration. MUK/DLK/ZPK is a MAP kinase kinase kinase (MAPKKK) enzyme that activates JNK via MAP kinase kinases (MAPKK) such as MKK7. We show here that the expression of MUK/DLK/ZPK protein in the developing mouse embryo is almost totally specific for the neural tissues, including central, peripheral, and autonomic nervous systems. The only obvious exception is the liver, in which the protein is temporally expressed at around E11. The expression becomes obvious in the neurons of the brain and neural crest tissues at embryonic day 10 (E10) after neuron production is initiated. By E14, MUK/DLK/ZPK proteins are found in various neural tissues including the brain, spinal cord, sensory ganglia (such as trigeminal and dorsal root ganglia), and the sympathetic and visceral nerves. The localization of MUK/DLK/ZPK protein in neural cells almost consistently overlapped that of betaIII-tubulin, a neuron specific tubulin isoform, and both proteins were more concentrated in axons than in cell bodies and dendrites. The intensely overlapping localization of betaIII-tubulin and MUK/DLK/ZPK indicated that this protein kinase is tightly associated with the microtubules of neurons.


Assuntos
Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Sistema Nervoso/embriologia , Proteínas Serina-Treonina Quinases/genética , Animais , Axônios/enzimologia , Ativação Enzimática , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase 4 , MAP Quinase Quinase Quinases , Camundongos , Microtúbulos/enzimologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Sistema Nervoso/enzimologia , Neurônios/enzimologia
12.
Oncol Rep ; 13(6): 1033-41, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15870918

RESUMO

Von Hippel-Lindau (VHL) disease is associated with various missense germline mutations in the VHL tumor suppressor gene. Some are associated with type 1 VHL disease, renal cell carcinoma (RCC) without pheochromocytoma, while others are associated with type 2A or 2B VHL disease, pheochromocytoma without and with RCC, respectively. These mutations may cause substitutions of specific amino acid residue and functional change of VHL protein (pVHL), which leads to the oncogenesis of the particular tumor types that characterize the different VHL disease types. To investigate, we transfected a pVHL-null RCC cell line with plasmids expressing wild-type pVHL (WT) or pVHL bearing 1 of 3 point mutations. These occur in the pVHL regions that bind hypoxia-inducible factor alpha (HIF-alpha ) or Elongin C. Microarray analysis showed that the clones bearing a mutation in the elongin-binding region (mutant 167) were unique, as many more genes were suppressed than up-regulated. The other two mutant groups, which bear a mutation in the HIF-alpha -binding region (mutants 98 and 111), showed the opposite pattern. The 167 mutation is associated with type 2B VHL disease. Real-time PCR analysis confirmed the altered expression of selected genes in the clones. Relative to WT, stratifin (14-3-3 sigma) and lysyl oxidase-like 1 were down-regulated in the 167 mutants, while the transforming growth factor beta-induced protein (beta ig-h3) was up-regulated in the 111 mutants. Thus, the location of pVHL mutations results in distinct gene expression patterns. Moreover, a mutation in the elongin-binding domain may induce type 2B tumors through different molecular pathways compared to those induced by type 1- or 2A-associated mutations in the HIF-alpha -binding region.


Assuntos
Carcinoma de Células Renais/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Neoplasias Renais/genética , Mutação/genética , Feocromocitoma/genética , Proteínas Supressoras de Tumor/genética , Ubiquitina-Proteína Ligases/genética , Neoplasias das Glândulas Suprarrenais/genética , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Elonguina , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor Von Hippel-Lindau , Doença de von Hippel-Lindau/genética
13.
PLoS One ; 10(5): e0126942, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25978062

RESUMO

In mouse cerebral corticogenesis, neurons are generated from radial glial cells (RGCs) or from their immediate progeny, intermediate neuronal precursors (INPs). The balance between self-renewal of these neuronal precursors and specification of cell fate is critical for proper cortical development, but the signaling mechanisms that regulate this progression are poorly understood. EphA4, a member of the receptor tyrosine kinase superfamily, is expressed in RGCs during embryogenesis. To illuminate the function of EphA4 in RGC cell fate determination during early corticogenesis, we deleted Epha4 in cortical cells at E11.5 or E13.5. Loss of EphA4 at both stages led to precocious in vivo RGC differentiation toward neurogenesis. Cortical cells isolated at E14.5 and E15.5 from both deletion mutants showed reduced capacity for neurosphere formation with greater differentiation toward neurons. They also exhibited lower phosphorylation of ERK and FRS2α in the presence of FGF. The size of the cerebral cortex at P0 was smaller than that of controls when Epha4 was deleted at E11.5 but not when it was deleted at E13.5, although the cortical layers were formed normally in both mutants. The number of PAX6-positive RGCs decreased at later developmental stages only in the E11.5 Epha4 deletion mutant. These results suggest that EphA4, in cooperation with an FGF signal, contributes to the maintenance of RGC self-renewal and repression of RGC differentiation through the neuronal lineage. This function of EphA4 is especially critical and uncompensated in early stages of corticogenesis, and thus deletion at E11.5 reduces the size of the neonatal cortex.


Assuntos
Diferenciação Celular/fisiologia , Células Ependimogliais/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Regeneração Nervosa/fisiologia , Receptor EphA4/fisiologia , Animais , Apoptose/fisiologia , Western Blotting , Encéfalo/anatomia & histologia , Encéfalo/embriologia , Encéfalo/fisiologia , Linhagem da Célula/fisiologia , Células Cultivadas , Camundongos , Transdução de Sinais/fisiologia
14.
Sex Dev ; 8(5): 297-310, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24503953

RESUMO

The study of the external genitalia is divided into 2 developmental stages: the formation and growth of a bipotential genital tubercle (GT) and the sexual differentiation of the male and female GT. The sexually dimorphic processes, which occur during the second part of GT differentiation, are suggested to be governed by androgen signaling and more recently crosstalk with other signaling factors. The process of elucidating the regulatory mechanisms of hormone signaling towards other signaling networks in the GT is still in its early stages. Nevertheless, it is becoming a productive area of research. This review summarizes various studies on the development of the murine GT and the defining characteristics of a masculinized GT and presents the different signaling pathways possibly involved during masculinization.


Assuntos
Genitália/embriologia , Diferenciação Sexual/fisiologia , Transdução de Sinais/fisiologia , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genitália/citologia , Genitália/metabolismo , Masculino , Camundongos
15.
Dev Neurobiol ; 70(14): 929-42, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20715151

RESUMO

Doublecortin (DCX) is expressed in young neurons and functions as a microtubule-associated protein. DCX is essential for neuronal migration because humans with mutations in the DCX gene exhibit cortical lamination defects known as lissencephaly in males and subcortical laminar heterotopia (or double cortex syndrome) in females. Phosphorylation of DCX alters its affinity for tubulin and may modulate neurite extension and neuronal migration. Previous in vitro phosphorylation experiments revealed that cyclin-dependent kinase 5 (Cdk5) phosphorylates multiple sites of DCX, including Ser332, (S332). However, phosphorylation at only Ser297 has been shown in vivo. In the present study, we examined phosphorylation of S332 of DCX in the Cdk5-/- mouse brain and results found, unexpectedly, indicate an increased DCX phosphorylation at S332. We found that JNK, not Cdk5, phosphorylates DCX at S332 in vivo. To examine the physiological significance of S332 phosphorylation of DCX in neuronal cells, we transfected cells with either GFP, GFP-DCX-WT, or GFP-DCX-S332A and analyzed neurite extension and migration. Introduction of GFP-DCX-WT enhanced neurite extension and migration. These effects of DCX introduction were suppressed when we used GFP-DCX-S332A. Treatment of neurons with JNK inhibitor increased the amount of DCX that bound to tubulin. Interestingly, amount of DCX that bound to tubulin decreased in Cdk5-/- brain homogenates, which indicates that phosphorylation of DCX by JNK is critical for the regulation of DCX binding to tubulin. These results suggest the physiological importance of phosphorylation of DCX for its function.


Assuntos
Movimento Celular/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Neuritos/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Fosforilação/fisiologia , Animais , Western Blotting , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Quinase 5 Dependente de Ciclina/genética , Quinase 5 Dependente de Ciclina/metabolismo , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Imuno-Histoquímica , Imunoprecipitação , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Camundongos , Camundongos Endogâmicos ICR , Neurônios/citologia , Transfecção
16.
Intern Med ; 47(11): 1027-31, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18520115

RESUMO

A 72-year-old man patient was referred to our institution for evaluation and treatment of right pleural effusion. Eosinophilic pleural effusion and peripheral eosinophilia were identified during the course of hospitalization. Pulmonary paragonimiasis was confirmed by the presence of paragonimus-specific IgG antibodies for Paragonimus (P.) westermani and P. miyazakii in his serum. Although Praziquantel, a highly effective agent for the treatment of lung flukes was repeatedly administered, the pleural effusion did not subside and the patient's condition gradually deteriorated until his death due to circulatory insufficiency. Postmortem examination revealed malignant mesothelioma of the sarcomatous type encasing the right lung and heart. Cardiac involvement accompanied with old and recent-onset myocardial ischemic changes resulted in death of this patient. Here, we report a very rare case of malignant mesothelioma with a concomitant infection of parasitic lung fluke.


Assuntos
Neoplasias Cardíacas/complicações , Pneumopatias Parasitárias/complicações , Pneumopatias/complicações , Mesotelioma/complicações , Paragonimíase/complicações , Idoso , Animais , Anti-Helmínticos/uso terapêutico , Anticorpos Anti-Helmínticos/sangue , Eosinofilia/complicações , Neoplasias Cardíacas/diagnóstico , Humanos , Pneumopatias/diagnóstico , Pneumopatias Parasitárias/tratamento farmacológico , Pneumopatias Parasitárias/parasitologia , Masculino , Mesotelioma/diagnóstico , Paragonimíase/tratamento farmacológico , Paragonimíase/parasitologia , Paragonimus/imunologia , Paragonimus/isolamento & purificação , Paragonimus westermani/imunologia , Paragonimus westermani/isolamento & purificação , Derrame Pleural/etiologia , Praziquantel/uso terapêutico
17.
Nat Genet ; 40(6): 782-8, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18469812

RESUMO

Early infantile epileptic encephalopathy with suppression-burst (EIEE), also known as Ohtahara syndrome, is one of the most severe and earliest forms of epilepsy. Using array-based comparative genomic hybridization, we found a de novo 2.0-Mb microdeletion at 9q33.3-q34.11 in a girl with EIEE. Mutation analysis of candidate genes mapped to the deletion revealed that four unrelated individuals with EIEE had heterozygous missense mutations in the gene encoding syntaxin binding protein 1 (STXBP1). STXBP1 (also known as MUNC18-1) is an evolutionally conserved neuronal Sec1/Munc-18 (SM) protein that is essential in synaptic vesicle release in several species. Circular dichroism melting experiments revealed that a mutant form of the protein was significantly thermolabile compared to wild type. Furthermore, binding of the mutant protein to syntaxin was impaired. These findings suggest that haploinsufficiency of STXBP1 causes EIEE.


Assuntos
Epilepsias Mioclônicas/genética , Proteínas Munc18/genética , Mutação de Sentido Incorreto/genética , Adulto , Sequência de Aminoácidos , Encéfalo/diagnóstico por imagem , Deleção Cromossômica , Cromossomos Humanos Par 9/genética , Dicroísmo Circular , Eletroencefalografia , Epilepsias Mioclônicas/patologia , Feminino , Genoma Humano , Heterozigoto , Humanos , Hibridização in Situ Fluorescente , Lactente , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino , Análise em Microsséries , Dados de Sequência Molecular , Proteínas Munc18/química , Proteínas Munc18/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Hibridização de Ácido Nucleico , Conformação Proteica , Proteínas Qa-SNARE/metabolismo , Radiografia , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas
18.
Biochem Biophys Res Commun ; 352(1): 84-90, 2007 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-17109820

RESUMO

Studies of islet neogenesis have suggested that the regeneration of islet cells can be achieved through redifferentiation of pre-existing islet cells. However, this hypothesis is largely unproven and fails to account for the diversity of observed islet neogenesis. Here we show that cultured neonatal pancreatic cells dedifferentiate into betaIII tubulin-expressing precursors, which then expand and redifferentiate into both neural and pancreatic lineage progenies. Redifferentiation happens not only in the islet cells, but also in the ductal cells that may represent what are called ductal origin "pancreatic stem cells". The in vitro redifferentiation of neonatal pancreatic cells recapitulates the embryonic development by sequential endocrine differentiation accompanied by the coexpression of neuronal marker betaIII tubulin with endocrine hormones until terminal differentiation. The neuronal differentiation of pancreatic cells, however, occurs prior to endocrine differentiation and gradually becomes dominant, thus implying a default neuronal lineage specification for cultured pancreatic cells.


Assuntos
Diferenciação Celular , Linhagem da Célula , Neurônios/citologia , Pâncreas/citologia , Animais , Animais Recém-Nascidos , Biomarcadores , Separação Celular , Células Cultivadas , Insulina/genética , Camundongos , Camundongos Endogâmicos C57BL , Pâncreas/metabolismo , Regiões Promotoras Genéticas/genética , Células-Tronco/citologia , Células-Tronco/metabolismo
19.
Cell Tissue Res ; 325(1): 189-95, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16520976

RESUMO

JNK is one of the key molecules regulating cell differentiation and migration in a variety of cell types, including cerebral cortical neurons. MUK/DLK/ZPK belongs to the MAP kinase-kinase-kinase class of protein kinases for the JNK pathway and is expressed predominantly in neural tissue. We have determined the expression pattern of MUK/DLK/ZPK and active JNK in the cerebellum at different stages of postnatal development. Quantitative analysis by Western blotting has showed that high expression levels of MUK/DLK/ZPK and active JNK are maintained during the postnatal development of the cerebellum, and that these levels decrease in the adult cerebellum. Immunohistochemical staining has revealed, however, that their distribution in the developing cerebellum is considerably different. Although active JNK is highly concentrated in the premigratory zone of the external germinal layer (EGL), high expression of MUK/DLK/ZPK has been observed in the molecular layer and in the premigratory zone. Neither the active JNK nor MUK protein has been detected in the proliferative zone of the EGL. These observations suggest that during the postnatal development of the cerebellum, the MUK-JNK signaling pathway contributes to the regulation of granule cell differentiation and migration; further, the activity of MUK/DLK/ZPK is tightly regulated by posttranslational mechanisms and by its expression level.


Assuntos
Cerebelo/embriologia , Cerebelo/enzimologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Animais , Ativação Enzimática , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Modelos Biológicos , Neurônios/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo
20.
Development ; 133(9): 1735-44, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16571631

RESUMO

In developing mammalian telencephalon, the loss of adherens junctions and cell cycle exit represent crucial steps in the differentiation of neuroepithelial cells into neurons, but the relationship between these cellular events remains obscure. Atypical protein kinase C (aPKC) is known to contribute to junction formation in epithelial cells and to cell fate determination for Drosophila neuroblasts. To elucidate the functions of aPKClambda, one out of two aPKC members, in mouse neocortical neurogenesis, a Nestin-Cre mediated conditional gene targeting system was employed. In conditional aPKClambda knockout mice, neuroepithelial cells of the neocortical region lost aPKClambda protein at embryonic day 15 and demonstrated a loss of adherens junctions, retraction of apical processes and impaired interkinetic nuclear migration that resulted in disordered neuroepithelial tissue architecture. These results are evidence that aPKClambda is indispensable for the maintenance of adherens junctions and may function in the regulation of adherens junction integrity upon differentiation of neuroepithelial cells into neurons. In spite of the loss of adherens junctions in the neuroepithelium of conditional aPKClambda knockout mice, neurons were produced at a normal rate. Therefore, we concluded that, at least in the later stages of neurogenesis, regulation of cell cycle exit is independent of adherens junctions.


Assuntos
Neocórtex/citologia , Neocórtex/fisiologia , Células Neuroepiteliais/metabolismo , Neurônios/metabolismo , Proteína Quinase C/fisiologia , Junções Aderentes/ultraestrutura , Animais , Células Cultivadas , Imuno-Histoquímica , Isoenzimas/genética , Isoenzimas/fisiologia , Camundongos , Camundongos Knockout , Neocórtex/embriologia , Neocórtex/ultraestrutura , Neurônios/ultraestrutura , Técnicas de Cultura de Órgãos , Proteína Quinase C/genética , Telencéfalo/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA