Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Clin Exp Immunol ; 162(2): 348-61, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21077278

RESUMO

Single immunoglobulin (Ig) interleukin-1R-related molecule (SIGIRR) is an Ig-like membrane protein critical for negative regulation of Toll-like receptor (TLR)-4-mediated signalling. We investigated SIGIRR expression and its regulation mechanism in intestinal epithelial cells (IECs) during inflammation. Endoscopic biopsy specimens were obtained from active and inactive colonic mucosa of ulcerative colitis (UC) patients, then SIGIRR expression was examined using real-time polymerase chain reaction (PCR) and immunohistochemistry (IH). Mice experimental colitis models were established by administrations of sulphonic acid (TNBS) and dextran sodium sulphate (DSS), and epithelial expression of SIGIRR was examined using real-time PCR, IH and flow cytometry. The effects of lipopolysaccharide (LPS) and tumour necrosis factor (TNF)-α on SIGIRR expression were evaluated in vitro using cultured IECs. To elucidate SIGIRR expression regulation in IECs, binding ability of the transcription factor SP1 at the responsive element of the SIGIRR promoter was examined using gel-shift and chromatin immunoprecipitation (ChIP) assays. In human colonic samples, SIGIRR was expressed mainly in IECs at levels significantly higher in inactive compared to active mucosa. In the mice, SIGIRR colonic expression decreased rapidly after colitis development and returned gradually to basal levels. Experimental colitis-mediated down-regulation of SIGIRR in IECs was also confirmed by IH and flow cytometry results. Further, inflammatory conditions induced by TLR ligands and TNF-α caused significant down-regulation of SIGIRR expression in IECs, which was dependent upon decreased SP1 binding at the responsive element of the SIGIRR promoter. We found that SIGIRR is expressed in IECs and serves as a negative regulator to maintain gut innate immunity, which is down-regulated during inflammation by inhibition of an SP1-mediated pathway.


Assuntos
Colite/metabolismo , Regulação para Baixo/imunologia , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Receptores de Interleucina-1/metabolismo , Adulto , Idoso , Animais , Linhagem Celular Tumoral , Colite/induzido quimicamente , Colite Ulcerativa/metabolismo , Colo/metabolismo , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Humanos , Inflamação/metabolismo , Intestino Grosso/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , RNA Interferente Pequeno/genética , Receptores de Interleucina-1/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Fator de Transcrição Sp1/metabolismo , Organismos Livres de Patógenos Específicos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia , Adulto Jovem
2.
Clin Exp Immunol ; 159(2): 185-98, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19912257

RESUMO

Several negative regulatory mechanisms control Toll-like receptor (TLR)-mediated inflammatory responses and restore immune system balance, including the zinc-finger protein A20, a negative regulator of TLR signalling that inhibits nuclear factor kappa B (NF-kappaB) activity. In the present study, we investigated TLR-5-mediated A20 expression and its role in intestinal epithelial cells (IECs) during inflammation. HCT-15 and HT-29 cells were stimulated with flagellin, then the expressions of A20, interleukin-1 receptor-associated kinase (IRAK-M) and Tollip were evaluated using RNase protection assay. Furthermore, experimental colitis was induced in tlr4-deficient CH3/HeJ mice by administration of dextran sodium sulphate (DSS), then flagellin was injected anally, and the colonic expression of A20 was examined by real-time polymerase chain reaction (PCR) and immunohistochemistry. To confirm flagellin-induced expression of A20, we employed an organ culture system. The role of A20 in flagellin-induced tolerance induction was evaluated in vitro, using a gene knock-down method targeting A20. A20 expression increased rapidly and peaked at 1 h after flagellin stimulation in cultured IECs, then declined gradually to the basal level. In vivo, anal injection of flagellin induced epithelial expression of A20 in injured colonic tissue, whereas flagellin did not cause a significant increase in A20 expression in non-injured normal tissue, which was also confirmed in vitro using the organ culture system. Gene knock-down using A20 siRNA did not influence tolerance induced by restimulation with flagellin. A20 is an early response negative regulator of TLR-5 signalling in IECs that functions during intestinal inflammation. Our results provide new insights into the negative feedback regulation of TLR-5 signalling that maintains the innate immune system in the gut.


Assuntos
Células Epiteliais/metabolismo , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Receptor 5 Toll-Like/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Proteínas de Ligação a DNA , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Flagelina/administração & dosagem , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Células HT29 , Humanos , Imuno-Histoquímica , Inflamação/patologia , Intestinos/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lipopolissacarídeos/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Proteínas Nucleares/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Receptor 5 Toll-Like/genética , Proteína 3 Induzida por Fator de Necrose Tumoral alfa
3.
J Oral Rehabil ; 35(6): 408-14, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18422514

RESUMO

Many papers have been published on surgical mandibulectomy and reconstruction. However, only a few reports refer to masticatory function after prosthodontic treatment in mandibulectomy patients. The aim of this study was to investigate the masticatory function of mandibulectomy patients. Twenty-three subjects (10 males and 13 females, with an average age of 63 years) participated in this study: 11 subjects who had undergone unilateral marginal mandibulectomy, six subjects with unilateral segmental mandibulectomy with reconstruction and six subjects with hemimandibulectomy without reconstruction. Mixing Ability Index (MAI) was used to measure masticatory function on the non-defect side and on the defect side with a prosthesis installed. Comparisons were carried out among the marginal, segmental and hemimandibular groups and between the non-defect side and the defect side. Consequently, our study indicates these results. On the non-defect side, a significant difference was found between the marginal and the segmental groups, and between the marginal and the hemimandibular groups. In the marginal and the segmental groups, a significant difference was found between the non-defect and the defect sides. In conclusion, our study suggests that MAI is an adequate tool to study the masticatory function in mandibulectomy patients, the masticatory function of the mandibulectomy patients is more impaired than that of the ordinary removable partial denture patients, and that surgical intervention affects the masticatory function on not only the defect side but also the non-defect side in mandibulectomy patients.


Assuntos
Mandíbula/cirurgia , Mastigação/fisiologia , Idoso , Feminino , Humanos , Masculino , Mandíbula/fisiopatologia , Neoplasias Mandibulares/fisiopatologia , Neoplasias Mandibulares/cirurgia , Pessoa de Meia-Idade , Período Pós-Operatório , Estatísticas não Paramétricas
4.
Am J Pathol ; 143(3): 804-13, 1993 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8395772

RESUMO

Microphthalmia and cerebral atrophies were induced in mouse embryos after injection of murine cytomegalovirus (MCMV) into the conceptus at midgestation. The concepti of ICR mice on day 8.5 of gestation were injected with MCMV through the uterine wall, then pregnancies were allowed to continue. On day 15.5 of gestation, microphthalmia was observed in 19.2% of the MCMV-injected embryos (1 x 10(4) plaque-forming units). As the survival rate decreased when pregnancies were allowed to continue further, incidence of microphthalmia decreased, whereas cerebral atrophies, determined by examining the histological sections, were observed in 17.6% of the surviving mouse fetuses on day 18.5 of gestation. Microphthalmia was confirmed by microscopically measuring the eyes on the serial coronal sections. There were two types of microphthalmia: one with marked hypoplastic eye with periglobular mesenchymal proliferation, the other with small eye and lens without the mesenchymal proliferation. Immunohistochemical analysis was performed using antibodies specific to the nuclear antigen of MCMV. Viral antigen-positive cells were widely distributed in the mesenchymes around the oral and nasal cavities and in the mesenchymes around the brain, especially in the endothelial cells of the vessels and the perivascular mesodermal cells. In the eyes, viral antigen-positive cells were observed in mononuclear blood cells in the cavities of the vitreous bodies. These results suggest that the primary target of congenital cytomegalovirus infection may be the mesenchymal cells; then the infection extends to the eyes and brain. In addition, the mesenchymal infection may also disrupt their organogenesis, resulting in microphthalmia and cerebral atrophy. This experimental system may provide a model similar to congenital cytomegalovirus infection in humans.


Assuntos
Córtex Cerebral/patologia , Infecções por Citomegalovirus/complicações , Microftalmia/microbiologia , Animais , Antígenos Virais/análise , Atrofia/embriologia , Atrofia/microbiologia , Atrofia/patologia , Córtex Cerebral/microbiologia , Olho/microbiologia , Olho/patologia , Idade Gestacional , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos ICR , Microftalmia/embriologia
5.
J Gen Virol ; 73 ( Pt 4): 975-81, 1992 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1353108

RESUMO

The human embryonal carcinoma cell line NT2/D1 is known to be non-permissive for human cytomegalovirus (HCMV) but becomes permissive after being induced to differentiate by retinoic acid (RA). Because homeobox genes have been reported to be specifically activated in the RA-differentiated NT2/D1 cells, we investigated the possible correlation between expression of homeobox (HOX) 2 genes and expression of the immediate early (IE) genes of HCMV both in NT2/D1 cells and in HCMV permissive human embryonic lung (HEL) cells. HCMV infection did not induce activation of the HOX2A, HOX2E and HOX2I genes in undifferentiated NT2/D1 cells nor affect their activation in the RA-differentiated NT2/D1 cells. By in situ hybridization using a HOX2A RNA probe, HOX2A transcript-positive cells appeared as clusters in RA-differentiated NT2/D1 cells. Viral antigen-positive cells detected by immunofluorescence using an antibody specific for the IE-1 antigen of HCMV appeared as clusters among the population of cells in which the HOX2A transcript was detected. The HOX2A gene only was expressed in HEL cells, however none of the HOX2 genes was expressed in non-permissive HeLa, Raji or mouse embryonic cells. These results suggest that activation of the HOX2A may be necessary for the expression of IE genes. HCMV infection markedly increased the expression of the HOX2E gene in HEL cells in the presence, but not in the absence, of cycloheximide. Ultraviolet-inactivated HCMV also displayed this effect. On the other hand, HCMV infection suppressed expression of the HOX2A gene to some degree at the early and late phases of infection in HEL cells. Activation of the HOX2E gene by HCMV might possibly have a role in virus-induced abnormal embryogenesis.


Assuntos
Citomegalovirus/genética , Genes Homeobox/genética , Genes Virais/genética , Proteínas Imediatamente Precoces , Transcrição Gênica , Animais , Antígenos Virais/isolamento & purificação , Diferenciação Celular , Regulação da Expressão Gênica , Histocitoquímica , Humanos , Imuno-Histoquímica , Camundongos , Hibridização de Ácido Nucleico , RNA Mensageiro/isolamento & purificação , Células Tumorais Cultivadas
6.
Arch Virol ; 127(1-4): 37-48, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1333757

RESUMO

The susceptibility of mouse embryonic cells to murine cytomegalovirus (MCMV) infection was studied by injecting the virus in the early and mid-gestation stages. For the early stage, blastocysts from BDF1 mice were injected with MCMV or minimal essential medium (MEM) by micromanipulator and returned to the uteri of pseudopregnant ICR mice. On day 11 of gestation, the embryos were examined immunohistochemically, using antibody specific to the early antigen of MCMV, and the placentae were examined by plaque assay. No infection was detected by either method. Furthermore, no infection was detected in MCMV-infected blastocysts that were cultured and examined for infection by immunofluorescence. For mid-gestation embryos, the conceptus was injected with MCMV on day 8.5 of gestation and was subjected to immunohistochemical analysis from day 10.5 to 12.5 of gestation. Viral antigen-positive cells were first observed in the placentae, then antigen-positive cells appeared among the blood cells, endothelial and mesodermal cells of the embryos. On day 12.5 of gestation, clusters of viral antigen-positive cells were sometimes observed in the hearts and livers. Although the incidence was lower, viral antigen-positive cells were also observed in the neuroectoderm and the eyes. These results suggest that MCMV does not infect early embryos and that infection first occurs in the placenta of postimplantation embryos, whence it extends through the blood cells to the endothelial and mesodermal cells of different embryonic regions, eventually extending to the neuroectoderm.


Assuntos
Infecções por Citomegalovirus/embriologia , Citomegalovirus/patogenicidade , Embrião de Mamíferos/microbiologia , Animais , Antígenos Virais/metabolismo , Blastocisto/citologia , Blastocisto/microbiologia , Células Cultivadas , Idade Gestacional , Técnicas Imunoenzimáticas , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos , Placenta/microbiologia
7.
Acta Pathol Jpn ; 41(9): 661-7, 1991 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1663688

RESUMO

To investigate the pathogenesis of brain abnormalities caused by congenital cytomegalovirus (CMV) infection, we previously reported experimental murine models of brain damage induced by intraventricular injection of murine CMV (MCMV) at the late stage of gestation. In the present study, viral DNA-positive cells in the damaged brain at different postnatal stages detected by in situ hybridization were compared with viral antigen-positive cells detected by an immunoperoxidase method using a monoclonal antibody against the immediate early antigen. At birth, the number of viral DNA-positive cells almost equalled that of viral antigen-positive cells. Seven to ten days after birth, the number of viral DNA-positive cells in the brain of MCMV-injected mice was one-fifth that of viral antigen-positive cells. Viral DNA-positive cells were more numerous in the hippocampus than in the cortex, and their density was dependent on the presence of viral antigen-positive cells. Dotted reaction products were observed in the nuclei of viral DNA-positive cells. These cells were rarely detected in lesions of later stages such as atrophy of the cortex and hippocampus, or the wall of the cystic lesions. These results suggest that viral DNA-positive cells detected by in situ hybridization are infected cells in which viral DNA replication is occurring actively.


Assuntos
Encéfalo/microbiologia , Infecções por Citomegalovirus/microbiologia , Citomegalovirus/isolamento & purificação , DNA Viral/análise , Animais , Animais Recém-Nascidos/microbiologia , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Idade Gestacional , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos ICR , Hibridização de Ácido Nucleico
8.
Acta Neuropathol ; 82(6): 435-41, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1664630

RESUMO

Mouse embryos were infected with murine cytomegalovirus (MCMV) by injecting the virus into the cerebral ventricles at the late gestation. After deliveries, offspring were fed by the mothers until 4 weeks. Cystic brain lesions, regarded as porencephaly or paraventricular cysts, were observed in about 20% of the MCMV-injected offspring 3 to 4 weeks after birth. The porencephaly involved the cerebral cortex and the white matter, and sometimes opened to the ventricles, while the paraventricular cysts involved the white matter. The inner surfaces of the cysts were covered with thin monolayer cells. Around the cystic lesions, perivascular cuffings were sometimes observed in the meninges and the basal regions. Viral antigen-positive cells were observed in the cortex and the hippocampus but were hardly observed along the cystic walls. Immunohistochemical double staining using antibodies specific for the viral antigen and specific for factor VIII-related antigen showed that the brain capillary endothelial cells had susceptibility to MCMV infection, and in addition that some neurons in the cortex and the hippocampus had the same susceptibility. These findings suggest that there are at least two ways by which MCMV induce abnormalities in the developing mouse brains; migration of MCMV-infected neurons and affinity to the endothelial cells of the brain vessels to this virus.


Assuntos
Encefalopatias/patologia , Cistos/patologia , Infecções por Citomegalovirus/patologia , Animais , Animais Recém-Nascidos , Antígenos Virais/análise , Vasos Sanguíneos/microbiologia , Encefalopatias/imunologia , Encefalopatias/microbiologia , Circulação Cerebrovascular , Cistos/imunologia , Infecções por Citomegalovirus/imunologia , Doenças Fetais/patologia , Camundongos , Camundongos Endogâmicos ICR
9.
Endocrinol Jpn ; 37(1): 113-20, 1990 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-2166653

RESUMO

The effects of testosterone (T) on the secretion of progesterone (P) by ovarian granulosa cells obtained from immature rats pre-treated with pregnant mare's serum gonadotropin were examined in vitro. T (10 nM-10 microM) enhanced both basal and FSH- or cAMP-stimulated secretion of P in a dose-dependent manner. Furthermore, T augmented FSH-stimulated cAMP production. The biphasic secretory pattern of P induced by continuous superfusion of granulosa cells with FSH was much exaggerated in the cultures supplemented with T. A stimulatory effect of T on secretion of P was observed only in the medium that contained serum. T affected neither the basal nor the FSH-stimulated secretion on 20 alpha-dihydroprogesterone. Androsterone, a non-aromatizable and low-potency androgen, at a similar concentration as T mimicked the effects of T on the secretion of progesterone. These results indicate that androgen stimulates mature granulosa cells to enhance the secretion of P. This androgen action extends either up- or down-stream of cAMP in the process of steroidogenesis.


Assuntos
Androgênios/fisiologia , Células da Granulosa/metabolismo , Folículo Ovariano/metabolismo , Progesterona/metabolismo , 20-alfa-Di-Hidroprogesterona/metabolismo , Androsterona/farmacologia , Animais , Células Cultivadas , AMP Cíclico/biossíntese , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Hormônio Foliculoestimulante/fisiologia , Técnicas In Vitro , Ratos , Ratos Endogâmicos
10.
Biol Reprod ; 40(5): 935-41, 1989 May.
Artigo em Inglês | MEDLINE | ID: mdl-2765617

RESUMO

To estimate whether melatonin is involved in gonadal activity in the male quail, the dynamics of plasma melatonin at an early stage of the photoperiodic response were investigated. Nocturnal levels of melatonin were manipulated by treatment with anti-melatonin (anti-M). By means of 4 additional hours of photic stimulation of the brain (provided by a red light-emitting diode inserted through the back of the head) after the environmental lights (8L:16D, lights-on, 1000 h) were turned off, the elevation of levels of melatonin after lights-off was significantly suppressed on Days 1 and 2 (p less than 0.01); after 5 days of brain-lighting, gonadal growth first became noticeable. However, 4 h of brain-lighting before lights-on elicited no change in levels of melatonin or in gonadal growth. The injections of anti-M just before lights-off (at 1800 h) for the first 3 days caused significant gonadal growth (p less than 0.01), whereas injections at 2200, 0200, or 0600 h were without effect. In addition, 4 h of brain-lighting before lights-on became gonadostimulatory (p less than 0.01) when it was accompanied by the injection of anti-M at 1800 h, but remained without effect when anti-M was injected at 0600 h. These results suggest that melatonin is involved in the initial stage of photoperiodism in birds, and the timing of suppression of the elevation of melatonin levels is critical in gonadal development.


Assuntos
Ritmo Circadiano , Coturnix/fisiologia , Melatonina/fisiologia , Codorniz/fisiologia , Animais , Masculino , Melatonina/sangue , Tamanho do Órgão , Estimulação Luminosa , Testículo/crescimento & desenvolvimento
11.
Mol Cell Endocrinol ; 59(3): 213-20, 1988 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2846383

RESUMO

Granulosa cells from immature female rats, pretreated with pregnant mare's serum gonadotropin, were cultured with microcarrier beads for 24 h, and superfused with culture medium. Progesterone was transiently released following a 10-min pulse of FSH (100 ng/ml), and there was a self-priming effect of FSH. 10-min pulses of 8-bromo-adenosine 3',5'-cyclic monophosphate (8Br-cAMP) (1 mg/ml) mimicked the effects of follicle-stimulating hormone (FSH). Continuous superfusion with FSH induced biphasic secretion of progesterone, which was composed of a parabolic (the first) and a plateau (the second) phase. By contrast, the pattern of secretion induced by continuous superfusion with 8Br-cAMP was monophasic. FSH-stimulated secretion of progesterone was rapidly inhibited by the addition of 10 microM cycloheximide (CX), but secretion recovered upon removal of this inhibitor. In the second phase, the recovery of secretion was accompanied by an overshoot of the plateau value. The present results suggest that: (1) the generation of the time-related biphasic pattern of secretion cannot be interpreted by cAMP alone; (2) FSH stimulates the secretion of progesterone by a mechanism that involves newly synthesized protein.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/metabolismo , Progesterona/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/administração & dosagem , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Cálcio/farmacologia , AMP Cíclico/biossíntese , Cicloeximida/farmacologia , Relação Dose-Resposta a Droga , Feminino , Hormônio Foliculoestimulante/administração & dosagem , Células da Granulosa/efeitos dos fármacos , Ratos , Ratos Endogâmicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...