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 Rep ; 12(1): 3242, 2022 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-35217706

RESUMO

Blastocyst complementation is an intriguing way of generating humanized animals for organ preparation in regenerative medicine and establishing novel models for drug development. Confirming that complemented organs and cells work normally in chimeric animals is critical to demonstrating the feasibility of blastocyst complementation. Here, we generated thymus-complemented chimeric mice, assessed the efficacy of anti-PD-L1 antibody in tumor-bearing chimeric mice, and then investigated T-cell function. Thymus-complemented chimeric mice were generated by injecting C57BL/6 (B6) embryonic stem cells into Foxn1nu/nu morulae or blastocysts. Flow cytometry data showed that the chimeric mouse thymic epithelial cells (TECs) were derived from the B6 cells. T cells appeared outside the thymi. Single-cell RNA-sequencing analysis revealed that the TEC gene-expression profile was comparable to that in B6 mice. Splenic T cells of chimeric mice responded very well to anti-CD3 stimulation in vitro; CD4+ and CD8+ T cells proliferated and produced IFNγ, IL-2, and granzyme B, as in B6 mice. Anti-PD-L1 antibody treatment inhibited MC38 tumor growth in chimeric mice. Moreover, in the chimeras, anti-PD-L1 antibody restored T-cell activation by significantly decreasing PD-1 expression on T cells and increasing IFNγ-producing T cells in the draining lymph nodes and tumors. T cells produced by complemented thymi thus functioned normally in vitro and in vivo. To successfully generate humanized animals by blastocyst complementation, both verification of the function and gene expression profiling of complemented organs/cells in interspecific chimeras will be important in the near future.


Assuntos
Blastocisto , Linfócitos T CD8-Positivos , Animais , Blastocisto/metabolismo , Quimera/genética , Células-Tronco Embrionárias , Camundongos , Camundongos Endogâmicos C57BL
2.
Eur J Neurosci ; 44(5): 2272-84, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27422015

RESUMO

Synaptic efficacy is determined by various factors, including the quantal size, which is dependent on the amount of neurotransmitters in synaptic vesicles at the presynaptic terminal. It is essential for stable synaptic transmission that the quantal size is kept within a constant range and that synaptic efficacy during and after repetitive synaptic activation is maintained by replenishing release sites with synaptic vesicles. However, the mechanisms for these fundamental properties have still been undetermined. We found that the active zone protein CAST (cytomatrix at the active zone structural protein) played pivotal roles in both presynaptic regulation of quantal size and recycling of endocytosed synaptic vesicles. In the CA1 region of hippocampal slices of the CAST knockout mice, miniature excitatory synaptic responses were increased in size, and synaptic depression after prolonged synaptic activation was larger, which was attributable to selective impairment of synaptic vesicle trafficking via the endosome in the presynaptic terminal likely mediated by Rab6. Therefore, CAST serves as a key molecule that regulates dynamics and neurotransmitter contents of synaptic vesicles in the excitatory presynaptic terminal in the central nervous system.


Assuntos
Região CA1 Hipocampal/metabolismo , Proteínas do Citoesqueleto/metabolismo , Endocitose , Vesículas Sinápticas/metabolismo , Animais , Região CA1 Hipocampal/citologia , Proteínas do Citoesqueleto/genética , Potenciais Pós-Sinápticos Excitadores , Exocitose , Camundongos , Camundongos Endogâmicos C57BL , Potenciais Pós-Sinápticos em Miniatura , Terminações Pré-Sinápticas/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
3.
PLoS One ; 8(11): e80356, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24236178

RESUMO

Adherens junctions (AJs) play a role in mechanically connecting adjacent cells to maintain tissue structure, particularly in epithelial cells. The major cell-cell adhesion molecules at AJs are cadherins and nectins. Afadin binds to both nectins and α-catenin and recruits the cadherin-ß-catenin complex to the nectin-based cell-cell adhesion site to form AJs. To explore the role of afadin in radial glial and ependymal cells in the brain, we generated mice carrying a nestin-Cre-mediated conditional knockout (cKO) of the afadin gene. Newborn afadin-cKO mice developed hydrocephalus and died neonatally. The afadin-cKO brain displayed enlarged lateral ventricles and cerebral aqueduct, resulting from stenosis of the caudal end of the cerebral aqueduct and obliteration of the ventral part of the third ventricle. Afadin deficiency further caused the loss of ependymal cells from the ventricular and aqueductal surfaces. During development, radial glial cells, which terminally differentiate into ependymal cells, scattered from the ventricular zone and were replaced by neurons that eventually covered the ventricular and aqueductal surfaces of the afadin-cKO midbrain. Moreover, the denuded ependymal cells were only occasionally observed in the third ventricle and the cerebral aqueduct of the afadin-cKO midbrain. Afadin was co-localized with nectin-1 and N-cadherin at AJs of radial glial and ependymal cells in the control midbrain, but these proteins were not concentrated at AJs in the afadin-cKO midbrain. Thus, the defects in the afadin-cKO midbrain most likely resulted from the destruction of AJs, because AJs in the midbrain were already established before afadin was genetically deleted. These results indicate that afadin is essential for the maintenance of AJs in radial glial and ependymal cells in the midbrain and is required for normal morphogenesis of the cerebral aqueduct and ventral third ventricle in the midbrain.


Assuntos
Junções Aderentes/patologia , Células Ependimogliais/metabolismo , Células Ependimogliais/patologia , Deleção de Genes , Hidrocefalia/genética , Hidrocefalia/patologia , Proteínas dos Microfilamentos/genética , Animais , Aqueduto do Mesencéfalo/metabolismo , Aqueduto do Mesencéfalo/patologia , Modelos Animais de Doenças , Mesencéfalo/metabolismo , Mesencéfalo/patologia , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Fenótipo , Transporte Proteico , Terceiro Ventrículo/metabolismo , Terceiro Ventrículo/patologia
4.
PLoS One ; 8(6): e68134, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23840823

RESUMO

Afadin is an intracellular binding partner of nectins, cell-cell adhesion molecules, and plays important roles in the formation of cell-cell junctions. Afadin-knockout mice show early embryonic lethality, therefore little is known about the function of afadin during organ development. In this study, we generated mice lacking afadin expression in endothelial cells, and found that the majority of these mice were embryonically lethal as a result of severe subcutaneous edema. Defects in the lymphatic vessels of the skin were observed, although the morphology in the blood vessels was almost normal. Severe disruption of VE-cadherin-mediated cell-cell junctions occurred only in lymphatic endothelial cells, but not in blood endothelial cells. Knockout of afadin did not affect the differentiation and proliferation of lymphatic endothelial cells. Using in vitro assays with blood and lymphatic microvascular endothelial cells (BMVECs and LMVECs, respectively), knockdown of afadin caused elongated cell shapes and disruption of cell-cell junctions among LMVECs, but not BMVECs. In afadin-knockdown LMVECs, enhanced F-actin bundles at the cell periphery and reduced VE-cadherin immunostaining were found, and activation of RhoA was strongly increased compared with that in afadin-knockdown BMVECs. Conversely, inhibition of RhoA activation in afadin-knockdown LMVECs restored the cell morphology. These results indicate that afadin has different effects on blood and lymphatic endothelial cells by controlling the levels of RhoA activation, which may critically regulate the lymphangiogenesis of mouse embryos.


Assuntos
Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Endotélio Vascular/citologia , Linfangiogênese/fisiologia , Proteínas dos Microfilamentos/fisiologia , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Adesão Celular/fisiologia , Proliferação de Células , Células Cultivadas , Embrião de Mamíferos/metabolismo , Endotélio Vascular/metabolismo , Humanos , Junções Intercelulares/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos
5.
Mol Cell Biol ; 33(4): 644-60, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23207902

RESUMO

Polycomb-group (PcG) complex 1 acts as an E3 ubiquitin ligase both for histone H2A to silence transcription and for geminin to regulate its stability. Scmh1 is a substoichiometric component of PcG complex 1 that provides the complex with an interaction domain for geminin. Scmh1 is unstable and regulated through the ubiquitin-proteasome system, but its molecular roles are unknown, so we generated Scmh1-deficient mice to elucidate its function. Loss of Scmh1 caused derepression of Hoxb4 and Hoxa9, direct targets of PcG complex 1-mediated transcriptional silencing in hematopoietic cells. Double knockdown of Hoxb4 and Hoxa9 or transduction of a dominant-negative Hoxb4N→A mutant caused geminin accumulation. Age-related transcriptional downregulation of derepressed Hoxa9 also leads to geminin accumulation. Transduction of Scmh1 lacking a geminin-binding domain restored derepressed expression of Hoxb4 and Hoxa9 but did not downregulate geminin like full-length Scmh1. Each of Hoxb4 and Hoxa9 can form a complex with Roc1-Ddb1-Cul4a to act as an E3 ubiquitin ligase for geminin. We suggest that geminin dysregulation may be restored by derepressed Hoxb4 and Hoxa9 in Scmh1-deficient mice. These findings suggest that PcG and a subset of Hox genes compose a homeostatic regulatory system for determining expression level of geminin.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas Nucleares/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas de Ciclo Celular/genética , Linhagem Celular , Regulação para Baixo , Geminina , Técnicas de Inativação de Genes , Genes Homeobox , Loci Gênicos , Hematopoese , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas Nucleares/genética , Fenótipo , Proteínas do Grupo Polycomb/química , Proteínas do Grupo Polycomb/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Ubiquitina/metabolismo
6.
J Neurosci ; 32(35): 12192-203, 2012 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-22933801

RESUMO

How size and shape of presynaptic active zones are regulated at the molecular level has remained elusive. Here we provide insight from studying rod photoreceptor ribbon-type active zones after disruption of CAST/ERC2, one of the cytomatrix of the active zone (CAZ) proteins. Rod photoreceptors were present in normal numbers, and the a-wave of the electroretinogram (ERG)--reflecting their physiological population response--was unchanged in CAST knock-out (CAST(-/-)) mice. Using immunofluorescence and electron microscopy, we found that the size of the rod presynaptic active zones, their Ca(2+) channel complement, and the extension of the outer plexiform layer were diminished. Moreover, we observed sprouting of horizontal and bipolar cells toward the outer nuclear layer indicating impaired rod transmitter release. However, rod synapses of CAST(-/-) mice, unlike in mouse mutants for the CAZ protein Bassoon, displayed anchored ribbons, normal vesicle densities, clustered Ca(2+) channels, and essentially normal molecular organization. The reduction of the rod active zone size went along with diminished amplitudes of the b-wave in scotopic ERGs. Assuming, based on the otherwise intact synaptic structure, an unaltered function of the remaining release apparatus, we take our finding to suggest a scaling of release rate with the size of the active zone. Multielectrode-array recordings of retinal ganglion cells showed decreased contrast sensitivity. This was also observed by optometry, which, moreover, revealed reduced visual acuity. We conclude that CAST supports large active zone size and high rates of transmission at rod ribbon synapses, which are required for normal vision.


Assuntos
Proteínas do Citoesqueleto/deficiência , Proteínas do Citoesqueleto/genética , Deleção de Genes , Terminações Pré-Sinápticas/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Percepção Visual/fisiologia , Potenciais de Ação/fisiologia , Animais , Quimera , Feminino , Masculino , Camundongos , Camundongos Knockout , Estimulação Luminosa/métodos , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia
7.
J Cell Sci ; 124(Pt 13): 2231-40, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21652626

RESUMO

Afadin interacts with the cytoplasmic region of nectins, which are immunoglobulin-like cell adhesion molecules at adherens junctions, and links them to the actin cytoskeleton. Afadin regulates activities of cells in culture such as directional motility, proliferation and survival. We used Cre-loxP technology to generate mice conditionally lacking afadin specifically in the intestinal epithelia after birth. The loss of afadin caused increased paracellular permeability in the intestinal mucosa and enhanced susceptibility to the tissue destruction induced by dextran sulfate sodium. The junctional architecture of the intestinal epithelia appeared to be preserved, whereas the deficiency of afadin caused the mislocalization of nectin-2 and nectin-3 from adherens junctions to basolateral membrane domains but not that of other components of apical junctions. By contrast, such phenotypic changes were undetected in mice lacking nectin-2, nectin-3 or both. These findings suggest that afadin plays crucial roles, independently of the role as the nectin-afadin module, in barrier function and homeostasis of the intestinal epithelia once the epithelial structure has been established.


Assuntos
Mucosa Intestinal/metabolismo , Proteínas dos Microfilamentos/metabolismo , Animais , Adesão Celular , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Sulfato de Dextrana/farmacologia , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Mucosa Intestinal/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Nectinas , Permeabilidade , Fenótipo
8.
Biochem Biophys Res Commun ; 385(4): 539-44, 2009 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-19481057

RESUMO

In the hippocampus, synapses are formed between mossy fiber terminals and CA3 pyramidal cell dendrites and comprise highly developed synaptic junctions (SJs) and puncta adherentia junctions (PAJs). Dynamic remodeling of synapses in the hippocampus is implicated in learning and memory. Components of both the nectin-afadin and cadherin-catenin cell adhesion systems exclusively accumulate at PAJs. We investigated the role of afadin at synapses in mice in which the afadin gene was conditionally inactivated in hippocampal neurons. In these mutant mice, the signals for not only nectins, but also N-cadherin and beta-catenin, were hardly detected in the CA3 area, in addition to loss of the signal for afadin, resulting in disruption of PAJs. Ultrastructural analysis revealed an increase in the number of perforated synapses, suggesting the instability of SJs. These results indicate that afadin is involved not only in the assembly of nectins and cadherins at synapses, but also in synaptic remodeling.


Assuntos
Hipocampo/enzimologia , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas dos Microfilamentos/metabolismo , Sinapses/enzimologia , Animais , Caderinas/metabolismo , Moléculas de Adesão Celular/metabolismo , Hipocampo/citologia , Histona-Lisina N-Metiltransferase/genética , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Nectinas , Células Piramidais/enzimologia , Células Piramidais/ultraestrutura , beta Catenina/metabolismo
9.
Cancer Sci ; 100(4): 608-16, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19215226

RESUMO

LIM-domain only (LMO) 7 is a multifunctional protein that is predicted to regulate the actin cytoskeleton, assembly of adherens junctions in epithelial cells, and gene expression. LMO7 was highly expressed in the mouse lung and predominantly localized to the apical membrane domain of bronchiolar epithelial cells. Although mice lacking LMO7 were viable and fertile in specific pathogen-free conditions, they developed protruding epithelial lesions in the terminal and respiratory bronchioles and alveolar ducts at 14-15 weeks of age. Furthermore, they tended to develop spontaneous adenocarcinoma in the lung at over 90 weeks of age. The cumulative incidence ratios of lung cancer were 22% in LMO7(-/-) mice and 13% in LMO7(+/-) mice whereas no primary lung cancer was observed in wild-type mice. Ex vivo analyses of the cancer cells showed numerical chromosome abnormalities and tumorigenicity in nude mice. These results suggest that LMO7 can act as a tumor suppressor whose deficiency confers a genetic predisposition to naturally occurring lung cancer.


Assuntos
Suscetibilidade a Doenças/metabolismo , Neoplasias Pulmonares/genética , Fatores de Transcrição/deficiência , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma/ultraestrutura , Animais , Proteínas de Homeodomínio/genética , Imuno-Histoquímica , Proteínas com Domínio LIM , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/ultraestrutura , Camundongos , Camundongos Knockout , Camundongos Nus , Modelos Biológicos , Fatores de Transcrição/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Cell Biol ; 183(2): 323-37, 2008 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-18936251

RESUMO

Neurotransmitter release from presynaptic nerve terminals is regulated by soluble NSF attachment protein receptor (SNARE) complex-mediated synaptic vesicle fusion. Tomosyn inhibits SNARE complex formation and neurotransmitter release by sequestering syntaxin-1 through its C-terminal vesicle-associated membrane protein (VAMP)-like domain (VLD). However, in tomosyn-deficient mice, the SNARE complex formation is unexpectedly decreased. In this study, we demonstrate that the N-terminal WD-40 repeat domain of tomosyn catalyzes the oligomerization of the SNARE complex. Microinjection of the tomosyn N-terminal WD-40 repeat domain into neurons prevented stimulated acetylcholine release. Thus, tomosyn inhibits neurotransmitter release by catalyzing oligomerization of the SNARE complex through the N-terminal WD-40 repeat domain in addition to the inhibitory activity of the C-terminal VLD.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Neurotransmissores/metabolismo , Proteínas R-SNARE/metabolismo , Proteínas SNARE/metabolismo , Animais , Masculino , Camundongos , Fibras Musgosas Hipocampais/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/deficiência , Plasticidade Neuronal , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteínas R-SNARE/química , Proteínas R-SNARE/deficiência , Ratos , Ratos Wistar , Sequências Repetitivas de Aminoácidos , Proteínas SNARE/química , Proteínas SNARE/ultraestrutura , Relação Estrutura-Atividade , Sinapses/metabolismo , Transmissão Sináptica , Proteína 25 Associada a Sinaptossoma/metabolismo , Sintaxina 1/metabolismo
11.
J Immunol ; 180(7): 4774-84, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18354201

RESUMO

The Doc2 family comprises the brain-specific Doc2alpha and the ubiquitous Doc2beta and Doc2gamma. With the exception of Doc2gamma, these proteins exhibit Ca(2+)-dependent phospholipid-binding activity in their Ca(2+)-binding C2A domain and are thought to be important for Ca(2+)-dependent regulated exocytosis. In excitatory neurons, Doc2alpha interacts with Munc13-1, a member of the Munc13 family, through its N-terminal Munc13-1-interacting domain and the Doc2alpha-Munc13-1 system is implicated in Ca(2+)-dependent synaptic vesicle exocytosis. The Munc13 family comprises the brain-specific Munc13-1, Munc13-2, and Munc13-3, and the non-neuronal Munc13-4. We previously showed that Munc13-4 is involved in Ca(2+)-dependent secretory lysosome exocytosis in mast cells, but the involvement of Doc2 in this process is not determined. In the present study, we found that Doc2alpha but not Doc2beta was endogenously expressed in the RBL-2H3 mast cell line. Doc2alpha colocalized with Munc13-4 on secretory lysosomes, and interacted with Munc13-4 through its two regions, the N terminus containing the Munc13-1-interacting domain and the C terminus containing the Ca(2+)-binding C2B domain. In RBL-2H3 cells, Ca(2+)-dependent secretory lysosome exocytosis was inhibited by expression of the Doc2alpha mutant lacking either of the Munc13-4-binding regions and the inhibition was suppressed by coexpression of Munc13-4. Knockdown of endogenous Doc2alpha also reduced Ca(2+)-dependent secretory lysosome exocytosis, which was rescued by re-expression of human Doc2alpha but not by its mutant that could not bind to Munc13-4. Moreover, Ca(2+)-dependent secretory lysosome exocytosis was severely reduced in bone marrow-derived mast cells from Doc2alpha knockout mice. These results suggest that the Doc2alpha-Muunc13-4 system regulates Ca(2+)-dependent secretory lysosome exocytosis in mast cells.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Lisossomos/metabolismo , Mastócitos/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Antígenos/imunologia , Medula Óssea/imunologia , Medula Óssea/metabolismo , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular , Chlorocebus aethiops , Exocitose/imunologia , Regulação da Expressão Gênica , Humanos , Ionóforos/farmacologia , Lisossomos/imunologia , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , Proteínas de Membrana/genética , Camundongos , Proteínas do Tecido Nervoso/genética , Ésteres de Forbol/farmacologia , Ligação Proteica , Ratos
12.
Mol Cell Biol ; 27(12): 4388-405, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17438139

RESUMO

Synaptic scaffolding molecule (S-SCAM) interacts with a wide variety of molecules at excitatory and inhibitory synapses. It comprises three alternative splicing variants, S-SCAMalpha, -beta, and -gamma. We generated mutant mice lacking specifically S-SCAMalpha. S-SCAMalpha-deficient mice breathe and feed normally but die within 24 h after birth. Primary cultured hippocampal neurons from mutant mice have abnormally elongated dendritic spines. Exogenously expressed S-SCAMalpha corrects this abnormal morphology, while S-SCAMbeta and -gamma have no effect. Active RhoA decreases in cortical neurons from mutant mice. Constitutively active RhoA and ROCKII shift the length of dendritic spines toward the normal level, whereas ROCK inhibitor (Y27632) blocks the effect by S-SCAMalpha. S-SCAMalpha fails to correct the abnormal spine morphology under the treatment of N-methyl-d-aspartate (NMDA) receptor inhibitor (AP-5), Ca(2+)/calmodulin kinase inhibitor (KN-62), or tyrosine kinase inhibitor (PP2). NMDA treatment increases active RhoA in dendrites in wild-type hippocampal neurons, but not in mutant neurons. The ectopic expression of S-SCAMalpha, but not -beta, recovers the NMDA-responsive accumulation of active RhoA in dendrites. Phosphorylation of extracellular signal-regulated kinase 1/2 and Akt and calcium influx in response to NMDA are not impaired in mutant neurons. These data indicate that S-SCAMalpha is a scaffold required to activate RhoA protein in response to NMDA receptor signaling in dendrites.


Assuntos
Dendritos/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Blastocisto/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Córtex Cerebral/ultraestrutura , Espinhas Dendríticas/patologia , Eletroporação , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Hipocampo/citologia , Hipocampo/metabolismo , Hipocampo/ultraestrutura , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Neurônios/citologia , Neurônios/metabolismo , Neurônios/ultraestrutura , Gravidez , Isoformas de Proteínas/química , Estrutura Terciária de Proteína , Recombinação Genética , Sinapses/metabolismo
13.
Proc Natl Acad Sci U S A ; 104(1): 252-7, 2007 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-17190817

RESUMO

Myeloid dendritic cells (mDCs) recognize and respond to polyI:C, an analog of dsRNA, by endosomal Toll-like receptor (TLR) 3 and cytoplasmic receptors. Natural killer (NK) cells are activated in vivo by the administration of polyI:C to mice and in vivo are reciprocally activated by mDCs, although the molecular mechanisms are as yet undetermined. Here, we show that the TLR adaptor TICAM-1 (TRIF) participates in mDC-derived antitumor NK activation. In a syngeneic mouse tumor implant model (C57BL/6 vs. B16 melanoma with low H-2 expresser), i.p. administration of polyI:C led to the retardation of tumor growth, an effect relied on by NK activation. This NK-dependent tumor regression did not occur in TICAM-1(-/-) or IFNAR(-/-) mice, whereas a normal NK antitumor response was induced in PKR(-/-), MyD88(-/-), IFN-beta(-/-), and wild-type mice. IFNAR was a prerequisite for the induction of IFN-alpha/beta and TLR3. The lack of TICAM-1 did not affect IFN production but resulted in unresponsiveness to IL-12 production, mDC maturation, and polyI:C-mediated NK-antitumor activity. This NK activation required NK-mDC contact but not IL-12 function in in vivo transwell analysis. Implanted tumor growth in IFNAR(-/-) mice was retarded by adoptively transferring polyI:C-treated TICACM-1-positive mDCs but not TICAM-1(-/-) mDCs. Thus, TICAM-1 in mDCs critically facilitated mDC-NK contact and activation of antitumor NK, resulting in the regression of low MHC-expressing tumors.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Melanoma Experimental/imunologia , Células Mieloides/imunologia , Receptor 3 Toll-Like/fisiologia , Animais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Poli I-C/farmacologia , Receptor de Interferon alfa e beta/fisiologia
14.
J Immunol ; 177(12): 8512-21, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17142749

RESUMO

Rho family small GTP-binding proteins, including Rho, Rac, and Cdc42, are key determinants of cell movement and actin-dependent cytoskeletal morphogenesis. Rho GDP-dissociation inhibitor (GDI) alpha and Rho GDIbeta (or D4/Ly-GDI), closely related regulators for Rho proteins, are both expressed in hemopoietic cell lineages. Nevertheless, the functional contributions of Rho GDIs remain poorly understood in vivo. In this study, we report that combined disruption of both the Rho GDIalpha and Rho GDIbeta genes in mice resulted in reduction of marginal zone B cells in the spleen, retention of mature T cells in the thymic medulla, and a marked increase in eosinophil numbers. Furthermore, these mice showed lower CD3 expression and impaired CD3-mediated proliferation of T cells. While B cells showed slightly enhanced chemotactic migration in response to CXCL12, peripheral T cells showed markedly reduced chemotactic migration in response to CCL21 and CCL19 associated with decreased receptor levels of CCR7. Overall, Rho protein levels were reduced in the bone marrow, spleen, and thymus but sustained activation of the residual part of RhoA, Rac1, and Cdc42 was detected mainly in the bone marrow and spleen. Rho GDIalpha and Rho GDIbeta thus play synergistic roles in lymphocyte migration and development by modulating activation cycle of the Rho proteins in a lymphoid organ-specific manner.


Assuntos
Linfócitos B/fisiologia , Quimiotaxia , Inibidores de Dissociação do Nucleotídeo Guanina/fisiologia , Proteínas/fisiologia , Animais , Contagem de Células , Células Cultivadas , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Linfonodos/citologia , Masculino , Camundongos , Camundongos Endogâmicos , Antígenos de Histocompatibilidade Menor , Especificidade de Órgãos , Baço/citologia , Linfócitos T/fisiologia , Timo/citologia , Proteínas rho de Ligação ao GTP/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho , Inibidor beta de Dissociação do Nucleotídeo Guanina rho , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico
15.
Genes Cells ; 11(9): 1125-32, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16923130

RESUMO

Seminiferous epithelia of the testes contain two types of intercellular junctions: Sertoli-Sertoli junctions and Sertoli-spermatid junctions. The former junctions are equipped with tight and adherens junctions while the latter junctions are not. Ca2+ -independent immunoglobulin-like cell-cell adhesion molecules, nectin-2 and nectin-3, asymmetrically localize at the Sertoli cell side and at the spermatid side of Sertoli-spermatid junctions, respectively. They heterophilically trans-interact to make contact between the two cells. Nectin-2(-/-) mice have shown male-specific infertility, disrupted Sertoli-spermatid junctions and morphologically impaired spermatid development. Here we report testicular phenotypes of nectin-3(-/-) mice exhibiting male-specific infertility. Nectin-3(-/-) mice had defects in the later steps of sperm morphogenesis including distorted nuclei and abnormal distribution of mitochondria, as well as in localization of nectin-2 at the Sertoli-spermatid junctions. Transplantation of wild-type spermatogenic stem cells into the nectin-3(-/-) testes partially rescued these defects in sperm morphogenesis. These results indicate that the heterophilic trans-interaction between nectin-2 and nectin-3 is essential for the formation and maintenance of Sertoli-spermatid junctions that plays a critical role in spermatid development.


Assuntos
Moléculas de Adesão Celular/metabolismo , Espermátides/crescimento & desenvolvimento , Animais , Moléculas de Adesão Celular/deficiência , Feminino , Infertilidade Masculina , Junções Intercelulares/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nectinas , Fenótipo , Transporte Proteico , Células de Sertoli/patologia , Células de Sertoli/ultraestrutura , Espermátides/citologia , Espermátides/patologia , Espermátides/ultraestrutura , Transplante de Células-Tronco
16.
Proc Natl Acad Sci U S A ; 103(26): 10029-34, 2006 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-16782817

RESUMO

Rab3A small G protein is a member of the Rab family and is most abundant in the brain, where it is localized on synaptic vesicles. Evidence is accumulating that Rab3A plays a key role in neurotransmitter release and synaptic plasticity. Rab3A cycles between the GDP-bound inactive and GTP-bound active forms, and this change in activity is associated with the trafficking cycle of synaptic vesicles at nerve terminals. Rab3 GTPase-activating protein (GAP) stimulates the GTPase activity of Rab3A and is expected to determine the timing of the dissociation of Rab3A from synaptic vesicles, which may be coupled with synaptic vesicle exocytosis. Rab3 GAP consists of two subunits: the catalytic subunit p130 and the noncatalytic subunit p150. Recently, mutations in p130 were found to cause Warburg Micro syndrome with severe mental retardation. Here, we generated p130-deficient mice and found that the GTP-bound form of Rab3A accumulated in the brain. Loss of p130 in mice resulted in inhibition of Ca(2+)-dependent glutamate release from cerebrocortical synaptosomes and altered short-term plasticity in the hippocampal CA1 region. Thus, Rab3 GAP regulates synaptic transmission and plasticity by limiting the amount of the GTP-bound form of Rab3A.


Assuntos
Plasticidade Neuronal/genética , Transmissão Sináptica/genética , Sinaptossomos/enzimologia , Proteínas rab3 de Ligação ao GTP/fisiologia , Proteína rab3A de Ligação ao GTP/metabolismo , Animais , Cálcio/metabolismo , Ácido Glutâmico/metabolismo , Hipocampo/enzimologia , Camundongos , Camundongos Mutantes , Sinapses/enzimologia , Proteínas rab3 de Ligação ao GTP/análise , Proteínas rab3 de Ligação ao GTP/genética , Proteína rab3A de Ligação ao GTP/análise
17.
Mol Cell Neurosci ; 31(2): 315-25, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16300961

RESUMO

Synapses are specialized intercellular junctions whose specificity and plasticity are mediated by synaptic cell adhesion molecules. In hippocampus, the mossy fibers form synapses on the apical dendrites of the CA3 pyramidal cells where synaptic and puncta adherentia junctions (PAJs) are highly developed. Synaptic junctions are the sites of neurotransmission, while PAJs are regarded as mechanical adhesion sites. Cell-cell adhesion molecules nectin-1 and nectin-3 asymmetrically localize at the pre- and post-synaptic sides of PAJs, respectively. To reveal the definitive role of nectins, we analyzed nectin-1-/- and nectin-3(-/-) mice. In both the mutant mice, the number of PAJs at the synapses between the mossy fiber terminals and the dendrites of the CA3 pyramidal cells was reduced. In addition, the abnormal mossy fiber trajectory was observed. These results indicate that nectins are involved in the formation of PAJs, which maintain the proper mossy fiber trajectory.


Assuntos
Moléculas de Adesão Celular/metabolismo , Hipocampo/citologia , Junções Intercelulares/metabolismo , Fibras Musgosas Hipocampais/metabolismo , Sinapses/metabolismo , Animais , Animais Recém-Nascidos , Caderinas , Moléculas de Adesão Celular/genética , Eletrofisiologia , Hipocampo/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Fibras Musgosas Hipocampais/ultraestrutura , Nectinas , Plasticidade Neuronal/fisiologia , Sinapses/ultraestrutura , Transmissão Sináptica/fisiologia
18.
Cancer Lett ; 220(1): 95-9, 2005 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-15737692

RESUMO

We have previously shown that transforming growth factor-beta1 (TGF-beta1) markedly stimulates the invasive capacity of rat ascites hepatoma AH130 W1 cells in vitro and in vivo. A differential hybridization procedure was used to isolate genes that were specifically up-regulated in TGF-beta1 treated W1 cells. Among ten independent cDNA clones, we focused on LMO7 and a variant isoform, LMO7S, that was generated by alternative splicing. LMO7 had PDZ and LIM domains, while LMO7S had only PDZ domain. TGF-beta1 up-regulated expression levels of LMO7 and LMO7S. LMO7 expression was up-regulated in the highly metastatic clone MM1.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proteínas de Homeodomínio/biossíntese , Neoplasias Hepáticas/metabolismo , Fatores de Transcrição/biossíntese , Fator de Crescimento Transformador beta/farmacologia , Animais , Ascite/metabolismo , Invasividade Neoplásica , Ratos , Fator de Crescimento Transformador beta1 , Células Tumorais Cultivadas , Regulação para Cima
19.
Development ; 132(7): 1525-37, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15728677

RESUMO

Nectins are Ca2+-independent immunoglobulin-like cell-cell-adhesion molecules consisting of four members. Nectins homophilically and heterophilically trans-interact to form a variety of cell-cell junctions, including cadherin-based adherens junctions in epithelial cells and fibroblasts in culture, synaptic junctions in neurons, and Sertoli cell-spermatid junctions in the testis, in cooperation with, or independently of, cadherins. To further explore the function of nectins, we generated nectin 1-/- and nectin 3-/-)mice. Both nectin 1-/- and nectin 3-/- mice showed a virtually identical ocular phenotype, microphthalmia, accompanied by a separation of the apex-apex contact between the pigment and non-pigment cell layers of the ciliary epithelia. Immunofluorescence and immunoelectron microscopy revealed that nectin 1 and nectin 3, but not nectin 2, localized at the apex-apex junctions between the pigment and non-pigment cell layers of the ciliary epithelia. However, nectin 1-/- and nectin 3-/- mice showed no impairment of the apicolateral junctions between the pigment epithelia where nectin 1, nectin 2 and nectin 3 localized, or of the apicolateral junctions between the non-pigment epithelia where nectin 2 and nectin 3, but not nectin 1, localized. These results indicate that the heterophilic trans-interaction between nectin 1 and nectin 3 plays a sentinel role in establishing the apex-apex adhesion between the pigment and non-pigment cell layers of the ciliary epithelia that is essential for the morphogenesis of the ciliary body.


Assuntos
Moléculas de Adesão Celular/fisiologia , Corpo Ciliar/embriologia , Animais , Moléculas de Adesão Celular/genética , Corpo Ciliar/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microftalmia/genética , Microftalmia/patologia , Nectinas , RNA Mensageiro/fisiologia
20.
J Clin Invest ; 114(6): 857-66, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15372110

RESUMO

A serine/threonine protein kinase, Cot/Tpl2, is indispensable for extracellular signal-regulated kinase (ERK) activation and production of TNF-alpha and PGE2 in LPS-stimulated macrophages. We show here that Cot/Tpl2 is also activated by other Toll-like receptor (TLR) ligands. Bacterial DNA rich in the dinucleotide CG (CpG-DNA), unlike LPS or synthetic lipopeptide, activated ERK in a Cot/Tpl2-independent manner. Peritoneal macrophages and bone marrow-derived DCs from Cot/Tpl2-/- mice produced significantly more IL-12 in response to CpG-DNA than those from WT mice. Enhanced IL-12 production in Cot/Tpl2-/- macrophages is, at least partly, regulated at the transcriptional level, and the elevated IL-12 mRNA level in Cot/Tpl2-/- macrophages is accompanied by decreased amounts of IL-12 repressors, such as c-musculoaponeurotic fibrosarcoma (c-Maf) and GATA sequence in the IL-12 promoter-binding protein (GA-12-binding protein; GAP-12) in the nucleus. Consistently, Cot/Tpl2-/- mice showed Th1-skewed antigen-specific immune responses upon OVA immunization and Leishmania major infection in vivo. These results indicate that Cot/Tpl2 is an important negative regulator of Th1-type adaptive immunity, that it achieves this regulation by inhibiting IL-12 production from accessory cells, and that it might be a potential target molecule in CpG-DNA-guided vaccination.


Assuntos
DNA Bacteriano/imunologia , Interleucina-12/biossíntese , MAP Quinase Quinase Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Células Th1/imunologia , Animais , Sequência de Bases , Técnicas de Cultura de Células , Primers do DNA , Éxons , MAP Quinase Quinase Quinases/deficiência , MAP Quinase Quinase Quinases/genética , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...