Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cells ; 9(9)2020 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-32961767

RESUMO

In eukaryotes, the C20:4 polyunsaturated fatty acid arachidonic acid (AA) plays important roles as a phospholipid component, signaling molecule and precursor of the endocannabinoid-prostanoid axis. Accordingly, the absence of AA causes detrimental effects. Here, compensatory mechanisms involved in AA deficiency in Caenorhabditis elegans were investigated. We show that the ω-3 C20:4 polyunsaturated fatty acid juniperonic acid (JuA) is generated in the C. elegansfat-3(wa22) mutant, which lacks Δ6 desaturase activity and cannot generate AA and ω-3 AA. JuA partially rescued the loss of function of AA in growth and development. Additionally, we observed that supplementation of AA and ω-3 AA modulates lifespan of fat-3(wa22) mutants. We described a feasible biosynthetic pathway that leads to the generation of JuA from α-linoleic acid (ALA) via elongases ELO-1/2 and Δ5 desaturase which is rate-limiting. Employing liquid chromatography mass spectrometry (LC-MS/MS), we identified endocannabinoid-like ethanolamine and glycerol derivatives of JuA and ω-3 AA. Like classical endocannabinoids, these lipids exhibited binding interactions with NPR-32, a G protein coupled receptor (GPCR) shown to act as endocannabinoid receptor in C. elegans. Our study suggests that the eicosatetraenoic acids AA, ω-3 AA and JuA share similar biological functions. This biosynthetic plasticity of eicosatetraenoic acids observed in C. elegans uncovers a possible biological role of JuA and associated ω-3 endocannabinoids in Δ6 desaturase deficiencies, highlighting the importance of ALA.


Assuntos
Caderinas/genética , Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Endocanabinoides/biossíntese , Fator de Crescimento Epidérmico/genética , Ácidos Graxos Insaturados/metabolismo , Longevidade/genética , Acetiltransferases/genética , Acetiltransferases/metabolismo , Animais , Ácido Araquidônico/deficiência , Caderinas/deficiência , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Dessaturase de Ácido Graxo Delta-5 , Fator de Crescimento Epidérmico/deficiência , Ácidos Graxos Dessaturases/genética , Ácidos Graxos Dessaturases/metabolismo , Regulação da Expressão Gênica , Ácido Linoleico/metabolismo , Mutação , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
2.
Tissue Cell ; 63: 101319, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32223947

RESUMO

Salivary epidermal growth factor (EGF) plays an important role in the maintenance of the oral and gastro-esophageal mucosa. Sialoadenectomy delays healing of oral wounds and affects lingual papillae. In this work, we aimed to determine the effect of EGF deficiency induced by sialoadenectomy and evaluate the effect of exogenous EGF administration on the lingual papillae and taste buds in rats. Thirty male adult Wistar albino rats were equally divided into 3 groups; sham-operated control group, sialoadenectomy group and group of sialoadenectomy + EGF. EGF was given 8 weeks after sialoadenectomy in a dose of 1 µg /ml/day in drinking water for 2 weeks. The anterior two-thirds of the tongue was dissected and cut longitudinally into two halves; one half for light microscope and the other for electron microscope examinations. Saliva and blood were collected to determine salivary and plasma EGF. Our results revealed that sialoadenectomy significantly reduced plasma and saliva levels of EGF which resulted in severe disruption of the architecture of lingual papillae. These changes were effectively improved by the exogenous EGF administration. In conclusion, EGF supplementation reversed the effects of sialoadenectomy and restored almost normal architecture of lingual papillae and taste buds.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Glândulas Salivares/metabolismo , Papilas Gustativas/metabolismo , Língua/metabolismo , Animais , Fator de Crescimento Epidérmico/deficiência , Fator de Crescimento Epidérmico/farmacologia , Mucosa Esofágica/efeitos dos fármacos , Mucosa Esofágica/metabolismo , Humanos , Mucosa Bucal/efeitos dos fármacos , Mucosa Bucal/metabolismo , Ratos , Saliva/efeitos dos fármacos , Saliva/metabolismo , Glândulas Salivares/efeitos dos fármacos , Glândulas Salivares/cirurgia , Papilas Gustativas/efeitos dos fármacos , Papilas Gustativas/cirurgia , Língua/efeitos dos fármacos , Língua/patologia , Língua/cirurgia
3.
J Immunoassay Immunochem ; 38(1): 21-33, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27379831

RESUMO

The failure of therapies targeting tumor angiogenesis may be caused by anti-angiogenic resistance mechanisms induced by VEGF and non-VEGF pathways alterations. Anti-angiogenic therapy failure is also attributed to immune system, acting by tumor-associated macrophages that release pro-angiogenic factors and a consequent increase of blood vessels. Recently, in a study by Rheal et al., a new angiogenic receptor, epidermal growth factor, latrophilin, and 7 trans-membrane domain-containing protein 1 on chromosome 1(ELTD1) has been identified as a promising glioma biomarker. In this study we aim to analyse whether this receptor may be used as a target molecule in glioblastoma therapy. Our results showed that small interfering RNA silencing ELTD1 caused cytotoxicity in glioblastoma cells. We also found that PDGFR, VEGFR, and their common PI3K/mTOR intracellular pathway inactivation-induced cytotoxicity in glioblastoma cells. Further, we found high percent of cytotoxicity in a low passage glioblastoma cell line after BEZ235 (a dual inhibitor of PI3K/mTOR pathway) treatment at nanomolar concentrations, compared to AG1433 (a PDGFR inhibitor) and SU1498 (a VEGFR inhibitor) that were only cytotoxic at micromolar ranges. In the future, these could prove as attractive therapeutic targets in single therapy or coupled with classic therapeutic approaches such as chemotherapy of radiotherapy.


Assuntos
Fator de Crescimento Epidérmico/deficiência , Inativação Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , RNA Interferente Pequeno/genética , Receptores Acoplados a Proteínas G/deficiência , Receptores de Peptídeos/deficiência , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/deficiência , Biomarcadores Tumorais/genética , Morte Celular/efeitos dos fármacos , Fator de Crescimento Epidérmico/biossíntese , Fator de Crescimento Epidérmico/genética , Inativação Gênica/efeitos dos fármacos , Glioblastoma/genética , Humanos , RNA Interferente Pequeno/farmacologia , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/biossíntese , Receptores de Peptídeos/genética
4.
J Neurophysiol ; 114(2): 1346-52, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26108956

RESUMO

Chondroitin sulfate proteoglycans (CSPGs) are widely studied in vertebrate systems and are known to play a key role in development, plasticity, and regulation of cortical circuitry. The mechanistic details of this role are still elusive, but increasingly central to the investigation is the homeostatic balance between network excitation and inhibition. Studying a simpler neuronal circuit may prove advantageous for discovering the mechanistic details of the cellular effects of CSPGs. In this study we used a well-established model of homeostatic change after injury in the crab Cancer borealis to show first evidence that CSPGs are necessary for network activity homeostasis. We degraded CSPGs in the pyloric circuit of the stomatogastric ganglion with the enzyme chondroitinase ABC (chABC) and found that removal of CSPGs does not influence the ongoing rhythm of the pyloric circuit but does limit its capacity for recovery after a networkwide perturbation. Without CSPGs, the postperturbation rhythm is slower than in controls and rhythm recovery is delayed. In addition to providing a new model system for the study of CSPGs, this study suggests a wider role for CSPGs, and perhaps the extracellular matrix in general, beyond simply plastic reorganization (as observed in mammals) and into a foundational regulatory role of neural circuitry.


Assuntos
Proteínas de Artrópodes/metabolismo , Braquiúros/fisiologia , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Fator de Crescimento Epidérmico/deficiência , Espaço Extracelular/metabolismo , Fator de Crescimento Insulin-Like I/deficiência , Animais , Far-Western Blotting , Condroitina ABC Liase/metabolismo , Fator de Crescimento Epidérmico/fisiologia , Gânglios dos Invertebrados/metabolismo , Insulina/deficiência , Fator de Crescimento Insulin-Like I/fisiologia , Erros Inatos do Metabolismo , Periodicidade , Técnicas de Cultura de Tecidos , Síndrome de Werner
5.
Nat Commun ; 6: 6188, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25650115

RESUMO

Reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) represents a profound change in cell fate. Here, we show that combining ascorbic acid (AA) and 2i (MAP kinase and GSK inhibitors) increases the efficiency of reprogramming from fibroblasts and synergistically enhances conversion of partially reprogrammed intermediates to the iPSC state. AA and 2i induce differential transcriptional responses, each leading to the activation of specific pluripotency loci. A unique cohort of pluripotency genes including Esrrb require both stimuli for activation. Temporally, AA-dependent histone demethylase effects are important early, whereas Tet enzyme effects are required throughout the conversion. 2i function could partially be replaced by depletion of components of the epidermal growth factor (EGF) and insulin growth factor pathways, indicating that they act as barriers to reprogramming. Accordingly, reduction in the levels of the EGF receptor gene contributes to the activation of Esrrb. These results provide insight into the rewiring of the pluripotency network at the late stage of reprogramming.


Assuntos
Ácido Ascórbico/farmacologia , Cromatina/efeitos dos fármacos , Epigênese Genética , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Diferenciação Celular , Reprogramação Celular/efeitos dos fármacos , Cromatina/química , Cromatina/metabolismo , Embrião de Mamíferos , Fator de Crescimento Epidérmico/deficiência , Fator de Crescimento Epidérmico/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Redes Reguladoras de Genes/efeitos dos fármacos , Genes Reporter , Proteínas de Fluorescência Verde , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Somatomedinas/deficiência , Somatomedinas/genética
6.
Int J Mol Med ; 34(2): 624-31, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24888317

RESUMO

Growth factors are important for regulating a variety of cellular processes and typically act as signaling molecules between cells. In the present study, we examined the mechanisms underlying the inhibitory effects of mixtures of recombinant growth factors (MRGFs) on nitric oxide (NO) and pro-inflammatory cytokine production in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. We also examined whether these effects are mediated through the mitogen­activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) signal transduction pathways. NO production was assessed by measuring nitrite acucmulation using the Greiss reaction. Cytokine concentrations were measured using respective ELISA kits for each cytokine. Our results revealed that the MRGFs significantly attenuated the LPS-induced production of pro-inflammatory cytokines and NO in a dose-dependent manner. To elucidate the mechanisms underlying the inhibitory effects of MRGFs, we examined the effects of the LPS-induced phosphorylation of MAPKs and the activation of the NF-κB signaling pathway on the stabilization of NF-κB nuclear translocation and inhibitory factor-κB (IκB) degradation. Western blot analysis was performed to determine the total and phosphorylated levels of ERK, as well as the nuclear translocation of NF-κB, and IκB phosphorylation and degradation. Our results demonstrated that treatment with MRGFs resulted in a reduction in the phosphorylation of the ERK and NF-κB signaling pathways, whereas the phosphorylation of JNK and p38 was not affected. Taken together, our results suggest that MRGFs inhibit the production of pro-inflammatory cytokines and NO by downregulating inducible NO synthase gene expression and blocking the phosphorylation of the ERK and NF-κB signaling pathways. These findings may provide direct evidence of the potential application of MRGFs in the prevention and treatment of inflammatory diseases.


Assuntos
Citocinas/biossíntese , Fator de Crescimento Epidérmico/deficiência , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Fator de Crescimento Insulin-Like I/deficiência , Insulina/deficiência , Óxido Nítrico/metabolismo , Animais , Citocinas/efeitos dos fármacos , Fator de Crescimento Epidérmico/administração & dosagem , Inflamação/induzido quimicamente , Insulina/administração & dosagem , Fator de Crescimento Insulin-Like I/administração & dosagem , Lipopolissacarídeos/toxicidade , Erros Inatos do Metabolismo , Camundongos , Proteínas Quinases Ativadas por Mitógeno/biossíntese , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , Transdução de Sinais/efeitos dos fármacos
7.
Cell Prolif ; 46(4): 447-56, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23829318

RESUMO

OBJECTIVES: Mesenchymal stem cells (MSCs) are a reliable resource for tissue regeneration, but their molecular mechanisms of differentiation and proliferation remain unclear; this situation has restricted use of MSCs to a limited number of applications. A previous study of ours found a member of the epidermal growth factor family, epiregulin (EREG), to be involved in regulation of MSC differentiation. In the present study, we have used human dental stem cells from the apical papilla (SCAPs) to investigate the role of EREG on proliferation of MSCs. MATERIALS AND METHODS: SCAPs were isolated from apical papillae of immature third molars. Retroviral short hairpin RNA (shRNA) was used to silence EREG gene expression, and human recombinant EREG protein was used to stimulate SCAPs. SCAP proliferation was examined using tetrazolium dye colorimetric assay/cell growth curve. Western blotting was performed to detect expressions of extracellular signal-regulated protein kinases 1 and 2 (Erk1/2), mitogen-activated protein kinases 1 and 2 (MEK1/2), protein kinase B (Akt), p38 mitogen-activated protein kinase (p38 MAPK) and c-Jun N-terminal kinase (JNK). RESULTS: Depletion of EREG with shRNA inhibited SCAP proliferation and repressed phosphorylation of Erk1/2 and JNK. Human recombinant EREG protein promoted cell proliferation and enhanced Erk1/2, MEK and JNK phosphorylation in SCAPs. Furthermore, blocking MEK/Erk signalling with specific Erk1/2 inhibitor PD98059, or JNK signalling with specific inhibitor SP600125, abolished effects of EREG on cell proliferation. CONCLUSION: These findings indicate that EREG could enhance cell proliferation in dental tissue-derived MSCs by activating MEK/Erk and JNK signalling pathways.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Sistema de Sinalização das MAP Quinases , Células-Tronco/metabolismo , Adolescente , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Células Cultivadas , Papila Dentária/citologia , Papila Dentária/efeitos dos fármacos , Papila Dentária/enzimologia , Papila Dentária/fisiologia , Fator de Crescimento Epidérmico/deficiência , Fator de Crescimento Epidérmico/genética , Epirregulina , Humanos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Proteínas Recombinantes/farmacologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia , Adulto Jovem , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Cell Stem Cell ; 11(2): 242-52, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22862949

RESUMO

The cellular signals controlling the formation of cardiomyocytes, vascular smooth muscle, and endothelial cells from stem cell-derived mesoderm are poorly understood. To identify these signals, a mouse embryonic stem cell (ESC)-based differentiation assay was screened against a small molecule library resulting in a 1,4-dihydropyridine inducer of type II TGF-ß receptor (TGFBR2) degradation-1 (ITD-1). ITD analogs enhanced proteasomal degradation of TGFBR2, effectively clearing the receptor from the cell surface and selectively inhibiting intracellular signaling (IC(50) ~0.4-0.8 µM). ITD-1 was used to evaluate TGF-ß involvement in mesoderm formation and cardiopoietic differentiation, which occur sequentially during early development, revealing an essential role in both processes in ESC cultures. ITD-1 selectively enhanced the differentiation of uncommitted mesoderm to cardiomyocytes, but not to vascular smooth muscle and endothelial cells. ITD-1 is a highly selective TGF-ß inhibitor and reveals an unexpected role for TGF-ß signaling in controlling cardiomyocyte differentiation from multipotent cardiovascular precursors.


Assuntos
Di-Hidropiridinas/farmacologia , Regulação para Baixo/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/metabolismo , Proteólise/efeitos dos fármacos , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Di-Hidropiridinas/química , Relação Dose-Resposta a Droga , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fator de Crescimento Epidérmico/deficiência , Fator de Crescimento Epidérmico/metabolismo , Células HEK293 , Humanos , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Peso Molecular , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Relação Estrutura-Atividade
9.
Circ Res ; 111(7): 876-81, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22872153

RESUMO

RATIONALE: The transforming growth factor-ß (TGFß) family member Nodal promotes cardiogenesis, but the mechanism is unclear despite the relevance of TGFß family proteins for myocardial remodeling and regeneration. OBJECTIVE: To determine the function(s) of TGFß family members during stem cell cardiogenesis. METHODS AND RESULTS: Murine embryonic stem cells were engineered with a constitutively active human type I Nodal receptor (caACVR1b) to mimic activation by Nodal and found to secrete a paracrine signal that promotes cardiogenesis. Transcriptome and gain- and loss-of-function studies identified the factor as TGFß2. Both Nodal and TGFß induced early cardiogenic progenitors in embryonic stem cell cultures at day 0 to 2 of differentiation. However, Nodal expression declines by day 4 due to feedback inhibition, whereas TGFß persists. At later stages (days 4-6), TGFß suppresses the formation of cardiomyocytes from multipotent Kdr(+) progenitors while promoting the differentiation of vascular smooth muscle and endothelial cells. CONCLUSIONS: Nodal induces TGFß, and both stimulate the formation of multipotent cardiovascular Kdr(+) progenitors. TGFß, however, becomes uniquely responsible for controlling subsequent lineage segregation by stimulating vascular smooth muscle and endothelial lineages and simultaneously blocking cardiomyocyte differentiation.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Miócitos Cardíacos/citologia , Proteína Nodal/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Células Cultivadas , Células-Tronco Embrionárias/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Fator de Crescimento Epidérmico/deficiência , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/fisiologia , Humanos , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Knockout , Modelos Animais , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Miócitos Cardíacos/fisiologia , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Transdução de Sinais/fisiologia
10.
Mol Ther ; 19(1): 196-203, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20842104

RESUMO

Multipotent stromal cells (MSCs) ameliorate several types of lung injury. The differentiation of MSCs into specific cells at the injury site has been considered as the important process in the MSC effect. However, although MSCs reduce destruction in an elastase-induced lung emphysema model, MSC differentiation is relatively rare, suggesting that MSC differentiation into specific cells does not adequately explain the recuperation observed. Humoral factors secreted by MSCs may also play an important role in ameliorating emphysema. To confirm this hypothesis, emphysema was induced in the lungs of C57BL/6 mice by intratracheal elastase injection 14 days before intratracheal MSC or phosphate-buffered saline (PBS) administration. Thereafter, lungs were collected at several time points and evaluated. Our results showed that MSCs reduced the destruction in elastase-induced emphysema. Furthermore, double immunofluorescence staining revealed infrequent MSC engraftment and differentiation into epithelial cells. Real-time PCR showed increased levels of hepatocyte growth factor (HGF) and epidermal growth factor (EGF). Real-time PCR and western blotting showed enhanced production of secretory leukocyte protease inhibitor (SLPI) in the lung. In-vitro coculture studies confirmed the in vivo observations. Our findings suggest that paracrine factors derived from MSCs is the main mechanism for the protection of lung tissues from elastase injury.


Assuntos
Enfisema/metabolismo , Enfisema/terapia , Células-Tronco Multipotentes/transplante , Animais , Western Blotting/métodos , Linhagem Celular , Enfisema/induzido quimicamente , Enfisema/patologia , Enfisema/prevenção & controle , Fator de Crescimento Epidérmico/deficiência , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Células Epiteliais/efeitos dos fármacos , Imunofluorescência/métodos , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Células-Tronco Multipotentes/citologia , Elastase Pancreática , Alvéolos Pulmonares/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Inibidor Secretado de Peptidases Leucocitárias/metabolismo , Regulação para Cima
11.
Brain Res ; 1358: 20-9, 2010 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-20735994

RESUMO

We hypothesized that one of the mechanisms underlying the protection of brain injury by therapeutic hypothermia is associated with preservation of neural stem cells. We investigated effects of moderate low temperature and the contribution of a cold-inducible molecule for the stemness of neural stem cells. The MEB5 mouse neural stem cell line was cultured in the presence or absence of EGF, and apoptosis, mRNA expression, and immunocytochemistry of the differentiation markers nestin and GFAP were evaluated at 37 or 32°C. We investigated the contribution of the cold-inducible RNA binding protein (CIRP) on apoptosis and differentiation of MEB5 cells at 32°C. EGF deprivation increased the number of apoptotic cells, decreased expression of nestin, and increased expression of GFAP. The moderate low temperature prevented apoptosis and decreases in expression of GFAP in MEB5 by EGF deprivation. The moderate low temperature significantly increased expression of CIRP. siRNA against CIRP significantly increased the apoptotic cell population of MEB5 cells via EGF deprivation at 32°C. These findings suggest that moderate low temperature preserved stemness of neural stem cells and prevented cell apoptosis via the stimulation of CIRP, and one of the mechanisms of rescue of brain injury by the moderate hypothermia is associated with preservation of neural stem cells.


Assuntos
Apoptose/fisiologia , Temperatura Baixa , Regulação da Expressão Gênica/fisiologia , Células-Tronco Neurais/fisiologia , Proteínas de Ligação a RNA/metabolismo , Animais , Apoptose/efeitos dos fármacos , Biotina/metabolismo , Caspases/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Fator de Crescimento Epidérmico/deficiência , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos , Células-Tronco Neurais/efeitos dos fármacos , Preservação Biológica , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Proteínas de Ligação a RNA/genética , Fatores de Tempo
12.
Int J Dev Biol ; 53(4): 549-57, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19247965

RESUMO

During development of the mammalian embryo, there is a complex relation between formation of the mesoderm and the neuroectoderm. In mouse, for example, the role of the node and its mesendoderm derivatives in anterior neural specification is still debated. Mouse Cripto(-/-) embryos could potentially help settle this debate because they lack almost all embryonic endoderm and mesoderm, including the node and its derivatives. In the present paper, we show that Cripto(-/-) embryos can still form functional neural stem cells that are able to differentiate and maintain a neural phenotype both in vivo and in vitro. These data suggest that signals emanating from the mesoderm and endoderm might not be essential for the formation and differentiation of neural stem cells. However, we use grafting experiments to show that the Cripto(-/-) isthmus (the secondary organizer located at the midbrain-hindbrain boundary) loses its inductive ability. We further show that the Cripto(-/-)isthmus expresses lower amounts of the isthmic signalling molecule, Fgf8. Since nearby tissues remain competent to respond to exogenously added Fgf8, this reduction in Fgf8 levels in the Cripto(-/-) isthmus is the potential cause of the loss of patterning ability in graft experiments. Overall, we interpret our data to suggest that the mammalian node and primitive streak are essential for the development of the regional identities that control the specification and formation of the secondary organizers within the developing brain.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Gastrulação/fisiologia , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Placa Neural/anormalidades , Placa Neural/metabolismo , Animais , Diferenciação Celular , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/metabolismo , Fator de Crescimento Epidérmico/deficiência , Fator de Crescimento Epidérmico/genética , Fator 8 de Crescimento de Fibroblasto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Placa Neural/citologia , Transdução de Sinais
13.
Neuroscience ; 158(4): 1378-89, 2009 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-19038313

RESUMO

During embryo neurogenesis, neurons that originate from stem cells located in the forebrain subventricular zone (SVZ) continuously migrate to the olfactory bulb (OB). However, other authors describe the occurrence of resident stem cells in the OB. In the present work we report that the absence of tumor suppressor protein p53 increases the number of neurosphere-forming cells and the proliferation of stem cells derived from 13.5-day embryo OB. Interestingly, differentiation of p53 knockout-derived neurospheres was biased toward neuronal precursors, suggesting a role for p53 in the differentiation process. Moreover, we demonstrate the relevance of p53 in maintaining chromosomal stability in response to genotoxic insult. Finally, our data show that neurosphere stem cells are highly resistant to long-term epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) deprivation in a p53-independent fashion, and they preserve their differentiation potential. Thus, these data demonstrate that p53 controls the proliferation, chromosomal stability and differentiation pattern of embryonic mouse olfactory bulb stem cells.


Assuntos
Diferenciação Celular/genética , Neurogênese/genética , Neurônios/fisiologia , Células-Tronco/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Anexinas/metabolismo , Ciclo Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Células Cultivadas , Aberrações Cromossômicas/efeitos da radiação , Embrião de Mamíferos , Fator de Crescimento Epidérmico/deficiência , Fator 2 de Crescimento de Fibroblastos/deficiência , Citometria de Fluxo/métodos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína Glial Fibrilar Ácida/metabolismo , Marcação In Situ das Extremidades Cortadas/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/efeitos da radiação , Antígenos O/metabolismo , Bulbo Olfatório/citologia , Ligação Proteica , Fatores de Tempo , Tubulina (Proteína)/metabolismo , Proteína Supressora de Tumor p53/deficiência , Raios X/efeitos adversos
14.
Dev Biol ; 315(2): 280-9, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18241853

RESUMO

During early mouse development, the TGFbeta-related protein Nodal specifies the organizing centers that control the formation of the anterior-posterior (A-P) axis. EGF-CFC proteins are important components of the Nodal signaling pathway, most likely by acting as Nodal coreceptors. However, the extent to which Nodal activity depends on EGF-CFC proteins is still debated. Cripto is the earliest EGF-CFC gene expressed during mouse embryogenesis and is involved in both A-P axis orientation and mesoderm formation. To investigate the relation between Cripto and Nodal in the early mouse embryo, we removed the Nodal antagonist Cerberus 1 (Cer1) and simultaneously Cripto, by generating Cer1;Cripto double mouse mutants. We observed that two thirds of the Cer1;Cripto double mutants are rescued in processes that are severely compromised in Cripto(-/-) embryos, namely A-P axis orientation, anterior mesendoderm and posterior neuroectoderm formation. The observed rescue is strongly reduced in Cer1;Cripto;Nodal triple mutants, suggesting that Nodal can signal extensively in the absence of Cripto, if Cer1 is also inhibited. This signaling activity drives A-P axis positioning. Our results provide evidence for the existence of Cripto-independent signaling mechanisms, by which Nodal controls axis specification in the early mouse embryo.


Assuntos
Padronização Corporal/fisiologia , Fator de Crescimento Epidérmico/fisiologia , Glicoproteínas de Membrana/fisiologia , Proteínas de Neoplasias/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/fisiologia , Animais , Padronização Corporal/genética , Citocinas , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Fator de Crescimento Epidérmico/deficiência , Fator de Crescimento Epidérmico/genética , Feminino , Gastrulação/genética , Gastrulação/fisiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Proteína Nodal , Organizadores Embrionários/fisiologia , Gravidez , Proteínas/genética , Proteínas/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/genética
15.
J Cell Physiol ; 215(1): 192-203, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17941089

RESUMO

Human Cripto-1 (CR-1) is a cell membrane protein that is overexpressed in several different types of human carcinomas. In the present study we investigated the mechanisms that regulate the expression of CR-1 gene in cancer cells. We cloned a 2,481 bp 5'-flanking region of the human CR-1 gene into a luciferase reporter vector and transfected NTERA-2 human embryonal carcinoma cells and LS174-T colon cancer cells to test for promoter activity. Activity of CR-1 promoter in both cell lines was modulated by two TGF-beta family members, TGF-beta1 and BMP-4. In particular, TGF-beta1 significantly up-regulated CR-1 promoter activity, whereas a dramatic reduction in CR-1 promoter activity was observed with BMP-4 in NTERA-2 and LS174-T cells. Changes in the CR-1 promoter activity following TGF-beta1 and BMP-4 treatments correlated with changes in CR-1 mRNA and protein expression in NTERA-2 and LS174-T cells. We also identified three Smad binding elements (SBEs) within the CR-1 promoter and point mutation of SBE1 (-2,197/-2,189) significantly reduced response of the CR-1 promoter to both TGF-beta1 and BMP-4 in NTERA-2 and LS174-T cells. Chromatin immunoprecipitation assay also demonstrated binding of Smad-4 to a CR-1 promoter DNA sequence containing SBE1 in LS174-T cells. Finally, BMP-4 inhibited migration of LS174-T cells and F9 mouse embryonal carcinoma cells by downregulation of CR-1 protein. In conclusion, these results suggest a differential modulation of CR-1 gene expression in embryonal and colon cancer cells by two different members of the TGF-beta family.


Assuntos
Proteínas Morfogenéticas Ósseas/farmacologia , Neoplasias do Colo/genética , Células-Tronco de Carcinoma Embrionário/metabolismo , Fator de Crescimento Epidérmico/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicoproteínas de Membrana/genética , Proteínas de Neoplasias/genética , Fator de Crescimento Transformador beta1/farmacologia , Região 5'-Flanqueadora , Animais , Sequência de Bases , Proteína Morfogenética Óssea 4 , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/patologia , Células-Tronco de Carcinoma Embrionário/patologia , Fator de Crescimento Epidérmico/deficiência , Fator de Crescimento Epidérmico/metabolismo , Proteínas Ligadas por GPI , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação/genética , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Smad/metabolismo
16.
Methods Mol Biol ; 329: 151-69, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16845990

RESUMO

Embryonic stem (ES) cells have been suggested as candidate therapeutic tools for regenerative medicine approaches. In this scenario, great efforts are made to define protocols to preferentially direct ES cells toward a defined cell type. To this end, it becomes crucial to characterize the molecular mechanisms as well as the signaling pathways implicated in ES cell differentiation. Findings highlight a key role of cripto, the founding member of a new class of extracellular factors, called EGF-CFC. Indeed, Cripto signaling is strictly required in an early acting window to negatively regulate neural differentiation and to permit differentiation of ES cells to cardiac fate. The protocols defined in this chapter allow preferential direction of ES cell differentiation as embryoid bodies toward either cardiomyocytes or neurons. Although referred to as modulation of Cripto signaling, these methods build the basis for the use of other classes of secreted molecules to control ES cell differentiation.


Assuntos
Embrião de Mamíferos/citologia , Fator de Crescimento Epidérmico/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Fator de Crescimento Epidérmico/deficiência , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/farmacologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/farmacologia , Camundongos , Camundongos Knockout , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Células-Tronco/efeitos dos fármacos
17.
Dev Cell ; 10(5): 667-72, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16678779

RESUMO

Activation of EGFR-Ras-MAPK signaling in vulval precursor cells (VPCs) by LIN-3/EGF from the gonad induces vulval development in C. elegans. The prevailing view is that LIN-3 overcomes an "inhibitory signal" from the adjacent hyp7 hypodermal syncytium. This view originated from observations indicating that inactivation of functionally redundant Synthetic Multivulva (SynMuv) genes in hyp7 can activate EGFR-Ras-MAPK signaling in the VPCs. Many SynMuv genes encode transcription and chromatin-associated factors, including the Rb ortholog. Here, we show that the SynMuv A and SynMuv B gene classes are functionally redundant for transcriptional repression of the key target gene, lin-3/EGF, in the hypodermis. These observations necessitate a revision of the concept of "inhibitory signaling." They also underscore the importance of preventing inappropriate cell signaling during development and suggest that derepression of growth factors may be the mechanism by which tumor suppressor genes such as Rb can have cell nonautonomous effects.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/embriologia , Indução Embrionária , Fator de Crescimento Epidérmico/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes de Helmintos/genética , Vulva/embriologia , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/biossíntese , Fator de Crescimento Epidérmico/biossíntese , Fator de Crescimento Epidérmico/deficiência , Feminino , Perfilação da Expressão Gênica , Células Gigantes/metabolismo , Modelos Biológicos , Mutação/genética , Fenótipo , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Vulva/metabolismo
18.
BMC Neurosci ; 6: 55, 2005 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-16124881

RESUMO

BACKGROUND: The recently discovered adult neural stem cells, which maintain continuous generation of new neuronal and glial cells throughout adulthood, are a promising and expandable source of cells for use in cell replacement therapies within the central nervous system. These cells could either be induced to proliferate and differentiate endogenously, or expanded and differentiated in culture before being transplanted into the damaged site of the brain. In order to achieve these goals effective strategies to isolate, expand and differentiate neural stem cells into the desired specific phenotypes must be developed. However, little is known as yet about the factors and mechanisms influencing these processes. It has recently been reported that pituitary adenylate cyclase-activating polypeptide (PACAP) promotes neural stem cell proliferation both in vivo and in vitro. RESULTS: We used cDNA microarrays with the aim of analysing the transcriptional changes underlying PACAP induced proliferation of neural stem cells. The primary neural stem/progenitor cells used were neurospheres, generated from the lateral ventricle wall of the adult mouse brain. The results were compared to both differentiation and proliferation controls, which revealed an unexpected and significant differential expression relating to withdrawal of epidermal growth factor (EGF) from the neurosphere growth medium. The effect of EGF removal was so pronounced that it masked the changes in gene expression patterns produced by the addition of PACAP. CONCLUSION: Experimental models aiming at transcriptional analysis of induced proliferation in primary neural stem cells need to take into consideration the significant effect on transcription caused by removal of EGF. Alternatively, EGF-free culture conditions need to be developed.


Assuntos
Proliferação de Células/efeitos dos fármacos , Fator de Crescimento Epidérmico/administração & dosagem , Regulação Enzimológica da Expressão Gênica/fisiologia , Neurônios/enzimologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Células-Tronco/enzimologia , Animais , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Fator de Crescimento Epidérmico/deficiência , Fator de Crescimento Epidérmico/fisiologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Camundongos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
19.
Neuron ; 46(2): 233-45, 2005 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-15848802

RESUMO

In an attempt to characterize the molecular components by which electric activity influences the development of synapses, we searched for cell surface proteins modulated by calcium influx and glutamate receptor activity. Here, we report that neuronal depolarization facilitates the conversion of CALEB, which results in a truncated transmembrane form with an exposed EGF domain. To characterize the role of CALEB in synapse development, synaptic features were investigated in slices of the colliculus superior from CALEB-deficient mice. In the absence of CALEB, the number of synapses and their morphological characteristics remained unchanged. However, in CALEB-deficient mice, synapses displayed higher paired-pulse ratios, less depression during prolonged repetitive activation, a lower rate of spontaneous postsynaptic currents, and a lower release probability at early but not mature postnatal stages. Our findings indicate that CALEB provides a molecular basis for maintaining normal release probability at early developmental stages.


Assuntos
Fator de Crescimento Epidérmico/deficiência , Glicoproteínas de Membrana/deficiência , Proteínas do Tecido Nervoso/deficiência , Sinapses/fisiologia , Sequência de Aminoácidos , Animais , Western Blotting , Embrião de Galinha , Potenciais Pós-Sinápticos Excitadores , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Técnicas de Cultura de Órgãos , Colículos Superiores/crescimento & desenvolvimento , Sinapses/ultraestrutura
20.
Plast Reconstr Surg ; 115(1): 128-33, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15622242

RESUMO

The aim of this article was to show the participation of epidermal growth factor (EGF) in the pathogenesis of Dupuytren's disease (palmar contracture). The concentration of EGF in specimens obtained from 68 patients with Dupuytren's contracture and 14 controls was examined immunochemically with the use of enzyme-linked immunosorbent assay. The determined EGF concentration in pathologic aponeurosis with symptoms of Dupuytren's disease (median, 6.29 ng/g; range, 1.67 to 63.09 ng/g) showed significantly different values (p = 0.036) in comparison with the control group (median, 10.1 ng/g; range, 5.13 to 39.81 ng/g). The changes in EGF concentration were shown in tested groups of pathologic tissues that were formed according to the clinical stage of disease progression. The significantly lower concentration than that seen in the control group characterizes tissues with first and third degrees of palmar contracture progression (p = 0.025 and p = 0.018, respectively). In the group of patients with second-degree disease progression, the EGF level increased transiently. Nevertheless, in comparison with the other groups, the difference was not significant. The group with the fourth degree of the disease showed EGF concentrations that resembled the control values. The authors conclude that significant differences in levels of EGF concentration between contractured and normal fasciae may suggest the participation of this cytokine in the pathogenesis of Dupuytren's disease.


Assuntos
Contratura de Dupuytren/etiologia , Fator de Crescimento Epidérmico/análise , Idoso , Progressão da Doença , Contratura de Dupuytren/metabolismo , Contratura de Dupuytren/patologia , Fator de Crescimento Epidérmico/deficiência , Fáscia/química , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fumar/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA