Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(49): e2305713120, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-38015851

RESUMO

The tumor suppressor protein p53 accumulates in response to cellular stress and consequently orchestrates the expression of multiple genes in a p53-level and time-dependent manner to overcome stress consequences, for which a molecular mechanism is currently unknown. Previously, we reported that DNA torsional flexibility distinguishes among p53 response elements (REs) and that transactivation at basal p53 levels is correlated with p53 REs flexibility. Here, we calculated the flexibility of ~200 p53 REs. By connecting functional outcomes of p53-target genes' activation to the calculated flexibility of their REs, we show that genes known to belong to pathways that are activated rapidly upon stress contain REs that are significantly more flexible relative to REs of genes known to be involved in pathways that are activated later in the response to stress. The global structural properties of several p53 REs belonging to different pathways were experimentally validated. Additionally, reporter-gene expression driven by flexible p53 REs occurred at lower p53 levels and with faster rates than expression from rigid REs. Furthermore, analysis of published endogenous mRNA levels of p53-target genes as a function of REs' flexibility showed that early versus late genes differ significantly in their flexibility properties of their REs and that highly flexible p53 REs enable high-activation level exclusively to early-response genes. Overall, we demonstrate that DNA flexibility of p53 REs contributes significantly to functional selectivity in the p53 system by facilitating the initial steps of p53-dependent target-genes expression, thereby contributing to survival versus death decisions in the p53 system.


Assuntos
Elementos de Resposta , Proteína Supressora de Tumor p53 , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ativação Transcricional , DNA/genética
2.
Cell Signal ; 86: 110085, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34280495

RESUMO

Inflammation and cancer are intimately linked. A key mediator of inflammation is the transcription-factor NF-κB/RelA:p50. SEF (also known as IL-17RD) is a feedback antagonist of NF-κB/RelA:p50 that is emerging as an important link between inflammation and cancer. SEF acts as a buffer to prevent excessive NF-κB activity by sequestering NF-κB/RelA:p50 in the cytoplasm of unstimulated cells, and consequently attenuating the NF-κB response upon pro-inflammatory cytokine stimulation. SEF contributes to cancer progression also via modulating other signaling pathways, including those triggered by growth-factors. Despite its important role in human physiology and pathology, mechanisms that regulate SEF biochemical properties and inhibitory activity are unknown. Here we show that human SEF is an intrinsically labile protein that is stabilized via CK2-mediated phosphorylation, and identified the residues whom phosphorylation by CK2 stabilizes hSEF. Unlike endogenous SEF, ectopic SEF was rapidly degraded when overexpressed but was stabilized in the presence of excess CK2, suggesting a mechanism for limiting SEF levels depending upon CK2 processivity. Additionally, phosphorylation by CK2 potentiated hSef interaction with NF-κB in cell-free binding assays. Most importantly, we identified a CK2 phosphorylation site that was indispensable for SEF inhibition of pro-inflammatory cytokine signaling but was not required for SEF inhibition of growth-factor signaling. To our knowledge, this is the first demonstration of post-translational modifications that regulate SEF at multiple levels to optimize its inhibitory activity in a specific signaling context. These findings may facilitate the design of SEF variants for treating cytokine-dependent pathologies, including cancer and chronic inflammation.


Assuntos
Caseína Quinase II , Caseínas , Caseína Quinase II/metabolismo , Caseínas/metabolismo , Humanos , NF-kappa B/metabolismo , Fosforilação , Transdução de Sinais , Fator de Transcrição RelA/metabolismo
4.
Cell Signal ; 59: 110-121, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30862497

RESUMO

The classical NF-κB transcription factor (RelA:p50) and the tumor suppressor Sef axis constitute a negative regulatory loop in which Sef, a target of NF-κB/RelA:p50, fine-tunes NF-κB/RelA:p50 transcriptional-activation in response to inflammatory stimuli trough binding to p50. Similar to the inhibitor IκBα, Sef sequesters NF-κB/RelA:p50 in the cytoplasm of unstimulated cells. Despite its key roles in regulating multiple cellular processes and its potential role as mediator between inflammation and cancer, Sef structural domains required to fulfill its tasks are poorly characterized, and how Sef specificity towards RelA:p50 is achieved is unknown. In-vitro binding assays using bacterially expressed Sef and Co-IP experiments, revealed that in addition to p50, Sef directly interacts with IκBα, and the IKKß subunit of the IKK complex which mediates RelA:p50 induction by inflammatory stimuli. These interactions are ligand-independent and do not require Sef post-translational modifications. Deletion mutagenesis mapped binding site to IKKß in a 74- residue segment juxtaposing Sef transmembrane domain, whereas several Sef regions seem to interact with IκBα. Moreover, we identified two new sites which together with the previously identified conserved tyrosine constitute three discontinuous Sef regions each indispensable for Sef binding to RelA:p50 and inhibiting its cytokine induced transcriptional activation. Contrary to IκBα, endogenous Sef is not degraded upon cytokine-stimulation, and its targeting in different cell types markedly enhances cytokine-induced NF-κB nuclear translocation. These results reveal Sef as the first scaffold that brings together the components of NF-κB/RelA:p50 signaling-module. Sef scaffolding function explains the basis for Sef specificity towards inhibiting inflammatory cytokine-induction of NF-κB/RelA:p50.


Assuntos
Subunidade p50 de NF-kappa B/metabolismo , Receptores de Interleucina/química , Receptores de Interleucina/metabolismo , Fator de Transcrição RelA/metabolismo , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/metabolismo , Sítios de Ligação , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células HEK293 , Humanos , Quinase I-kappa B/metabolismo , Inibidor de NF-kappaB alfa/metabolismo , Ligação Proteica
5.
Sci Rep ; 7(1): 15060, 2017 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-29118380

RESUMO

Carcinomas constitute over 80% of all human cancer types with no effective therapy for metastatic disease. Here, we demonstrate, for the first time, the efficacy of therapeutic-ultrasound (TUS) to deliver a human tumor suppressor gene, hSef-b, to prostate tumors in vivo. Sef is downregulated in various human carcinomas, in a manner correlating with tumor aggressiveness. In vitro, hSef-b inhibited proliferation of TRAMP C2 cells and attenuated activation of ERK/MAPK and the master transcription factor NF-κB in response to FGF and IL-1/TNF, respectively. In vivo, transfection efficiency of a plasmid co-expressing hSef-b/eGFP into TRAMP C2 tumors was 14.7 ± 2.5% following a single TUS application. Repeated TUS treatments with hSef-b plasmid, significantly suppressed prostate tumor growth (60%) through inhibition of cell proliferation (60%), and reduction in blood vessel density (56%). In accordance, repeated TUS-treatments with hSef-b significantly inhibited in vivo expression of FGF2 and MMP-9. FGF2 is a known mitogen, and both FGF2/MMP-9 are proangiogenic factors. Taken together our results strongly suggest that hSef-b acts in a cell autonomous as well as non-cell autonomous manner. Moreover, the study demonstrates the efficacy of non-viral TUS-based hSef-b gene delivery approach for the treatment of prostate cancer tumors, and possibly other carcinomas where Sef is downregulated.


Assuntos
Técnicas de Transferência de Genes , Neovascularização Patológica/prevenção & controle , Neoplasias da Próstata/terapia , Receptores de Interleucina/genética , Carga Tumoral/genética , Terapia por Ultrassom/métodos , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos Endogâmicos C57BL , Neovascularização Patológica/genética , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/genética , Receptores de Interleucina/metabolismo
6.
Semin Cell Dev Biol ; 53: 101-14, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26903404

RESUMO

Fibroblast growth factor (FGF) signaling is involved in a multitude of biological processes, while impairment of FGF signaling is implicated in a variety of human diseases including developmental disorders and cancer. Therefore, it is not surprising that FGF activity is regulated at multiple and distinct levels. This review focuses on positive and negative modulation of the FGF signal exemplified by recently identified protein modulators anosmin-1, fibronectin-leucine-rich transmembrane protein 3 (FLRT3) and similar expression to FGF (Sef). We examine how these proteins regulate FGF signaling at multiple levels and across species. Finally, we describe the role of these regulators in human disease.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Animais , Desenvolvimento Embrionário , Fatores de Crescimento de Fibroblastos/química , Humanos , Modelos Animais , Modelos Biológicos , Neoplasias/metabolismo
7.
Reproduction ; 148(5): 507-17, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25118304

RESUMO

Similar expression to FGF (Sef or IL17-RD), is a tumor suppressor and an inhibitor of growth factors as well as of pro-inflammatory cytokine signaling. In this study, we examined the regulation of Sef expression by gonadotropins during ovarian folliculogenesis. In sexually immature mice, in situ hybridization (ISH) localized Sef gene expression to early developing oocytes and granulosa cells (GC) but not to theca cells. Sef was also expressed in mouse ovarian endothelial cells, in the fallopian tube epithelium as well as in adipose tissue venules. SEF protein expression, determined by immunohistochemistry (IHC), correlated well with Sef mRNA expression in GC, while differential expression was noticed in oocytes. High Sef mRNA but undetectable SEF protein levels were observed in the oocytes of primary/secondary follicles, while an inverse correlation was found in the oocytes of preantral and small antral follicles. Sef mRNA expression dropped after pregnant mare's serum gonadotropin (PMSG) administration, peaked at 6-8 h after human chorionic gonadotropin (hCG) treatment, and declined by 12 h after this treatment. ISH and IHC localized the changes to oocytes and mural GC following PMSG treatment, whereas Sef expression increased in mural GC and declined in granulosa-lutein cells upon hCG treatment. The ovarian expression of SEF was confirmed using human samples. ISH localized SEF transcripts to human GC of antral follicles but not to corpora lutea. Furthermore, SEF mRNA was detected in human GC recovered from preovulatory follicles. These results are the first to demonstrate SEF expression in a healthy ovary during folliculogenesis. Hormonal regulation of its expression suggests that SEF may be an important factor involved in intra-ovarian control mechanisms.


Assuntos
Células da Granulosa/metabolismo , Proteínas de Membrana/metabolismo , Oócitos/metabolismo , Receptores de Interleucina/metabolismo , Animais , Gonadotropina Coriônica/farmacologia , Feminino , Fármacos para a Fertilidade/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Gonadotropinas Equinas/farmacologia , Células da Granulosa/efeitos dos fármacos , Humanos , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Oócitos/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptores de Interleucina/genética
8.
Dev Cell ; 23(3): 611-23, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22975329

RESUMO

The NF-κB transcription factor controls diverse biological processes. According to the classical model, NF-κB is retained in the cytoplasm of resting cells via binding to inhibitory, IκB proteins and translocates into the nucleus upon their ligand-induced degradation. Here we reveal that Sef, a known tumor suppressor and inhibitor of growth factor signaling, is a spatial regulator of NF-κB. Sef expression is regulated by the proinflammatory cytokines tumor necrosis factor and interleukin-1, and Sef specifically inhibits "classical" NF-κB (p50:p65) activation by these ligands. Like IκBs, Sef sequesters NF-κB in the cytoplasm of resting cells. However, contrary to IκBs, Sef continues to constrain NF-κB nuclear entry upon ligand stimulation. Accordingly, endogenous Sef knockdown markedly enhances stimulus-induced NF-κB nuclear translocation and consequent activity. This study establishes Sef as a feedback antagonist of proinflammatory cytokines and highlights its potential to regulate the crosstalk between proinflammatory cytokine receptors and receptor tyrosine kinases.


Assuntos
Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Citoplasma/metabolismo , Inflamação , NF-kappa B/metabolismo , Receptores de Interleucina/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Células HEK293 , Células HeLa , Humanos , Camundongos , NF-kappa B/antagonistas & inibidores , Células NIH 3T3
9.
Int J Biochem Cell Biol ; 40(10): 2040-52, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18450498

RESUMO

The strength and duration of intracellular signals must be precisely regulated, since inappropriate signaling can cause disease. Negative feedback mechanisms provide an effective means of controlling growth factor-mediated signaling, either by restricting the incoming signal or by inducing counter-regulatory mechanisms affecting signal propagation. Sef proteins represent a new class of feedback antagonists capable of regulating receptor tyrosine kinase signaling. The involvement of Sef in development, as well as in other biological processes, was demonstrated by biochemical and genetic approaches.


Assuntos
Retroalimentação Fisiológica , Proteínas de Membrana/metabolismo , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Transdução de Sinais , Sequência de Aminoácidos , Animais , Desenvolvimento Embrionário , Regulação da Expressão Gênica , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Dados de Sequência Molecular , Receptores Proteína Tirosina Quinases/metabolismo
10.
J Biol Chem ; 283(14): 9308-17, 2008 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-18230614

RESUMO

FGF10, a heparan sulfate (HS)-binding growth factor, is required for branching morphogenesis of mouse submandibular glands (SMGs). HS increases the affinity of FGF10 for FGFR2b, which forms an FGF10.FGFR2b.HS ternary signaling complex, and results in diverse biological outcomes, including proliferation and epithelial morphogenesis. Defining the HS structures involved in specific FGF10-mediated events is critical to understand how HS modulates growth factor signaling in specific developmental contexts. We used HS-deficient BaF3/FGFR2b cells, which require exogenous HS to proliferate, to investigate the HS requirements for FGF10-mediated proliferation and primary SMG epithelia to investigate the structural requirements of HS for FGF10-mediated epithelial morphogenesis. In BaF3/FGFR2b cells, heparin with at least 10 saccharides and 6-O-, 2-O-, and N-sulfates were required for maximal proliferation. During FGF10-mediated SMG epithelial morphogenesis, HS increased proliferation and end bud expansion. Defined heparin decasaccharide libraries showed that 2-O-sulfation with either an N-or 6-O-sulfate induced end bud expansion, whereas decasaccharides with 6-O-sulfation alone induced duct elongation. End bud expansion resulted from increased FGFR1b signaling, with increased FGFR1b, Fgf1, and Spry1 as well as increased Aqp5 expression, a marker of end bud differentiation. Duct elongation was associated with expression of Cp2L1, a marker of developing ducts. Collectively, these findings show that the size and sulfate patterns of HS modulate specific FGF10-mediated events, such as proliferation, duct elongation, end bud expansion, and differentiation, and provide mechanistic insight as to how the developmental localization of specific HS structures in tissues influences FGF10-mediated morphogenesis and differentiation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Epitélio/embriologia , Fator 10 de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Heparitina Sulfato/metabolismo , Morfogênese/efeitos dos fármacos , Glândula Submandibular/embriologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Aquaporina 5/biossíntese , Diferenciação Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fator 1 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos ICR , Morfogênese/fisiologia , Complexos Multiproteicos/metabolismo , Oligossacarídeos/metabolismo , Fosfoproteínas/biossíntese , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/efeitos dos fármacos
11.
Acta Histochem ; 110(2): 155-62, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17980404

RESUMO

Fibroblast growth factors (FGFs) signal via four distinct high affinity cell surface tyrosine kinase receptors, termed FGFR1-FGFR4 (FGFR-FGF-receptor). Recently, a new modulator of the FGF signaling pathway, the transmembrane protein 'similar expression to FGF genes' (Sef), has been identified in zebrafish and subsequently in mammals. Sef from mouse and human inhibits FGF mitogenic activity. In the present study, we analyzed the expression of Sef in distinct rat brain areas, in the spinal cord and in peripheral nerves and spinal ganglia using semi-quantitative RT-PCR. Furthermore, we studied the cellular expression pattern of Sef in intact spinal ganglia and sciatic nerves and, in addition, after crush lesion, using in situ hybridization and immunohistochemistry. Sef transcripts were expressed in all brain areas evaluated and in the spinal cord. A neuronal expression was found in both intact and injured spinal ganglia. Intact sciatic nerves, however, showed little or no Sef expression. Seven days after injury, high Sef expression was concentrated to the crush site, and Schwann cells seemed to be the source of Sef. The labeling pattern of up-regulated Sef was complementary to the patterns of FGF-2 and FGFR1-3, which were localized proximal and distal to the crush site. These results suggest an involvement of Sef during the nerve regeneration process, possibly by fine-tuning the effects of FGF signaling.


Assuntos
Proteínas de Membrana/metabolismo , Sistema Nervoso/metabolismo , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Regulação para Cima , Animais , Encéfalo/metabolismo , Feminino , Gânglios Espinais/lesões , Gânglios Espinais/metabolismo , Perfilação da Expressão Gênica , Imuno-Histoquímica , Proteínas de Membrana/genética , Compressão Nervosa , Sistema Nervoso Periférico/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células de Schwann/metabolismo , Nervo Isquiático/metabolismo , Medula Espinal/metabolismo
12.
Biochem Biophys Res Commun ; 350(4): 825-33, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17049492

RESUMO

Growth alterations within the gastric mucosa during chronic gastric inflammation are key steps in gastric cancer development. FGF7, a specific mitogen for epithelial cells, is implicated in epithelial tissue repair and cancer. We investigated FGF7 expression in normal human stomach, and in 35 cases from various gastric pathologies including 23 gastritis and 8 adenocarcinoma cases. Modest FGF7 protein levels were detected in the normal mucosal gland epithelium and in stromal fibroblasts. FGF7 protein levels, however, were markedly increased in the mucosal epithelium of all gastric inflammation cases. A similar elevated expression was also observed in gastric adenocarcinoma. Upregulation of FGF7 protein was associated with a modest increase in FGF7 mRNA expression. Interestingly, high levels of FGF7 anti-sense (AS) RNA were observed in the gastric pathologies, at the same sites where FGF7 protein was upregulated. Altogether, these findings suggest a role for FGF7 in maintaining gastric mucosa integrity, and that FGF7 protein levels are regulated mainly by posttranscriptional mechanisms. The elevated FGF7 protein levels in gastric inflammation and gastric cancer, together with the known oncogenic potential of FGF7, implicate excessive FGF7 signaling in gastric tumorigenesis, and point to FGF7 as an attractive target for gastric cancer prevention and treatment.


Assuntos
Adenocarcinoma/metabolismo , Biomarcadores Tumorais/metabolismo , Fator 7 de Crescimento de Fibroblastos/metabolismo , Mucosa Gástrica/metabolismo , Gastrite/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Gástricas/metabolismo , Expressão Gênica , Humanos , Gastropatias/metabolismo , Distribuição Tecidual
13.
J Biol Chem ; 281(51): 39225-35, 2006 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-17035228

RESUMO

Negative feedback is among the key mechanisms for regulating receptor tyrosine kinase (RTK) signaling. Human Sef, a recently identified inhibitor of RTK signaling, encodes different isoforms, including a membrane spanning (hSef-a) and a cytosolic (hSef-b) isoform. Previously, we reported that hSef-b inhibited fibroblast proliferation and prevented the activation of mitogen-activated protein kinase (MAPK), without affecting protein kinase B/Akt or p38 MAPK. Conflicting results were reported concerning hSef-a inhibition of MAPK activation, and the effect of hSef-a on other RTK-induced signaling pathways is unknown. Here we show that, in fibroblasts, similar to hSef-b, ectopic expression of hSef-a inhibited fibroblast growth factor-induced cell proliferation. Unlike hSef-b, however, the growth arrest was mediated via a MAPK-independent mechanism, and was accompanied by elevated p38 MAPK phosphorylation and inhibition of protein kinase B/Akt. In addition, hSef-a, but not hSef-b, mediated apoptosis in fibroblast growth factor-stimulated cells. Chemical inhibitor of p38 MAPK abrogated the effect of hSef-a on apoptosis. In epithelial cells, ectopic expression of hSef-a inhibited the activation of MAPK, whereas down-regulation of endogenous hSef-a significantly increased MAPK activation and accelerated growth factor-dependent cell proliferation. These results indicate that hSef-a is a multifunctional negative modulator of RTK signaling and clearly demonstrate that hSef-a can inhibit the activation of MAPK, although in a cell type-specific manner. Moreover, the differences between the activities of hSef-a and hSef-b suggest that hSef isoforms can control signal specificity and subsequent cell fate by utilizing different mechanisms to modulate RTK signaling.


Assuntos
Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Interleucina/fisiologia , Transdução de Sinais , Animais , Apoptose , Proliferação de Células , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Células HeLa , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Células NIH 3T3 , Isoformas de Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Interleucina/química , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
J Biol Chem ; 281(8): 5178-87, 2006 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-16269412

RESUMO

Heparin-binding growth factors are crucial for the formation of human epidermis, but little is known about the role of heparan sulfate proteoglycans in this process. Here we investigated the role of the heparan sulfate proteoglycan, perlecan, in the formation of human epidermis, by utilizing in vitro engineered human skin. By disrupting perlecan expression either in the dermis or the epidermis, we found that epidermally derived perlecan is essential for epidermal formation. Perlecan-deficient keratinocytes formed a strikingly thin and poorly organized epidermis because of premature apoptosis and failure to complete their stratification program. Exogenous perlecan fully restored epidermal formation. Perlecan deposition in the basement membrane zone correlated with formation of multilayered epidermis. Perlecan deficiency, however, had no effect on the lining and deposition of major basement membrane components as was evident by a continuous linear staining of laminin and collagen IV. Similarly, perlecan deficiency did not affect the distribution of beta1 integrin. Addition of the perlecan ligand, fibroblast growth factor 7, protected perlecan-deficient keratinocytes from cell death and improved the thickness of the epidermis. Taken together, our results revealed novel roles for perlecan in epidermal formation. Perlecan regulates both the survival and terminal differentiation steps of keratinocytes. Our results suggested a model whereby perlecan regulates these processes via controlling the bioavailability of perlecan-binding soluble factors involved in epidermal morphogenesis.


Assuntos
Epiderme/metabolismo , Proteoglicanas de Heparan Sulfato/química , Proteoglicanas de Heparan Sulfato/fisiologia , Queratinócitos/metabolismo , Células 3T3 , Animais , Apoptose , Membrana Basal/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Clonagem Molecular , Colágeno Tipo IV/química , Meios de Cultivo Condicionados/farmacologia , Derme/metabolismo , Fator 7 de Crescimento de Fibroblastos/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Hibridização In Situ , Laminina/química , Ligantes , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Hibridização de Ácido Nucleico , Oligonucleotídeos Antissenso/química , Ligação Proteica , Proteínas/química , Pele/metabolismo , Fatores de Tempo , Engenharia Tecidual
15.
Dev Dyn ; 233(2): 301-12, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15844098

RESUMO

The signaling pathways leading to growth and patterning of various organs are tightly controlled during the development of any organism. These control mechanisms usually involve the utilization of feedback- and pathway-specific antagonists where the pathway induces the expression of its own antagonist. Sef is a feedback antagonist of fibroblast growth factor (FGF) signaling, which has been identified recently in zebrafish and mammals. Here, we report the isolation of chicken Sef (cSef) and demonstrate the conserved nature of the regulatory relationship with FGF signaling. In chick embryos, Sef is expressed in a pattern that coincides with many known sites of FGF signaling. In the developing limb, cSef is expressed in the mesoderm underlying the apical ectodermal ridge (AER) in the region known as the progress zone. cSef message first appeared after limb budding and AER formation. Expression was intense at stages of rapid limb outgrowth, and gradually decreased to almost undetectable levels when differentiation was clearly apparent. Gain- and loss-of-function experiments showed that FGFs differentially regulate the expression of cSef in various tissues. Thus, removal of the AER down-regulated cSef expression, and FGF2 but not FGF4 or FGF8 beads substituted for the AER in maintaining cSef expression. At sites where cSef is not normally expressed, FGF4 and FGF2, but not FGF8 beads, induced cSef expression. Our results demonstrate the complexity of cSef regulation by FGFs and point to FGF2 as a prime candidate in regulating cSef expression during normal limb development. The spatiotemporal pattern of cSef expression during limb development suggests a role for cSef in regulating limb outgrowth but not limb initiation.


Assuntos
Proteínas Aviárias/metabolismo , Extremidades/embriologia , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/metabolismo , Sequência de Aminoácidos , Animais , Proteínas Aviárias/química , Proteínas Aviárias/genética , Embrião de Galinha , Clonagem Molecular , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 4 de Crescimento de Fibroblastos , Fator 8 de Crescimento de Fibroblasto , Humanos , Cinética , Botões de Extremidades/citologia , Botões de Extremidades/embriologia , Botões de Extremidades/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Mesoderma/metabolismo , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas/metabolismo , Alinhamento de Sequência , Transdução de Sinais
16.
Biochem J ; 389(Pt 1): 145-50, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15769253

RESUMO

HS (heparan sulphate) proteoglycans bind secreted signalling proteins, including FGFs (fibroblast growth factors) through their HS side chains. Such chains contain a wealth of differentially sulphated saccharide epitopes. Whereas specific HS structures are commonly believed to modulate FGF-binding and activity, selective binding of defined HS epitopes to FGFs has generally not been demonstrated. In the present paper, we have identified a series of sulphated HS octasaccharide epitopes, derived from authentic HS or from biosynthetic libraries that bind with graded affinities to FGF4, FGF7 and FGF8b. These HS species, along with previously identified oligosaccharides that interact with FGF1 and FGF2, constitute the first comprehensive survey of FGF-binding HS epitopes based on carbohydrate sequence analysis. Unexpectedly, our results demonstrate that selective modulation of FGF activity cannot be explained in terms of binding of individual FGFs to specific HS target epitopes. Instead, different FGFs bind to identical HS epitopes with similar relative affinities and low selectivity, such that the strength of these interactions increases with increasing saccharide charge density. We conclude that FGFs show extensive sharing of binding sites in HS. This conclusion challenges the current notion of specificity in HS-FGF interactions, and instead suggests that a set of common HS motifs mediates cellular targeting of different FGFs.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Heparitina Sulfato/química , Heparitina Sulfato/metabolismo , Sítios de Ligação , Epitopos , Fator 1 de Crescimento de Fibroblastos/química , Fator 1 de Crescimento de Fibroblastos/genética , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 4 de Crescimento de Fibroblastos/química , Fator 4 de Crescimento de Fibroblastos/genética , Fator 4 de Crescimento de Fibroblastos/metabolismo , Fator 7 de Crescimento de Fibroblastos/química , Fator 7 de Crescimento de Fibroblastos/genética , Fator 7 de Crescimento de Fibroblastos/metabolismo , Fator 8 de Crescimento de Fibroblasto/química , Fator 8 de Crescimento de Fibroblasto/genética , Fator 8 de Crescimento de Fibroblasto/metabolismo , Fatores de Crescimento de Fibroblastos/química , Fatores de Crescimento de Fibroblastos/genética , Ligação Proteica
17.
J Biol Chem ; 280(14): 13457-64, 2005 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-15695515

RESUMO

The hypothesis that neuropilin-1 (Npn-1) may interact with heparin-binding proteins other than vascular endothelial growth factor has been tested using an optical biosensor-based binding assay. The results show that fibroblast growth factor (FGF) 1, 2, 4, and 7, FGF receptor 1, hepatocyte growth factor/scatter factor (HGF/SF), FGF-binding protein, normal protease sensitive form of prion protein, antithrombin III, and Npn-1 itself are all able to interact with Npn-1 immobilized on the sensor surface. FGF-2, FGF-4, and HGF/SF are also shown to interact with Npn-1 in a solution assay. Moreover, these protein-protein interactions are dependent on the ionic strength of the medium and are inhibited by heparin, and the kinetics of binding of FGF-2, FGF-4 and HGF/SF to Npn-1 are characterized by fast association rate constants (270,000-1,600,000 m(-1) s(-1)). These results suggest that Npn-1 possesses a "heparin" mimetic site that is able to interact at least in part through ionic bonding with the heparin binding site on many of the proteins studied. Npn-1 was also found to potentiate the growth stimulatory activity of FGF-2 on human umbilical vein endothelial cells, indicating that Npn-1 may not just bind but also regulate the activity of heparin-binding proteins.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Heparina/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Neuropilina-1/metabolismo , Animais , Sítios de Ligação , Técnicas Biossensoriais , Linhagem Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Modelos Moleculares , Neuropilina-1/química , Polissacarídeos/metabolismo , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Receptores Proteína Tirosina Quinases/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo
18.
J Biol Chem ; 280(10): 9735-44, 2005 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-15637081

RESUMO

Sprouty proteins are evolutionarily conserved negative feedback regulators of multiple receptor tyrosine kinases. Mammalian versions of these proteins differentially regulate signaling induced by the fibroblast and the epidermal growth factors (FGF and EGF, respectively). Herein we show that, although both growth factors elevate expression of Sprouty-2, FGF- and not EGF-induced activation of the Erk/MAPK pathway is inhibited by Sprouty-2. Attenuation of FGF-signaling is accompanied by the induction of Sprouty-2 phosphorylation on the amino-terminal as well as carboxyl-terminal tyrosine residues, which are less effectively modified upon EGF treatment. Mutagenesis of carboxyl-terminal tyrosines, especially a newly identified phosphorylation site, tyrosine 227, impaired the inhibitory activity of Sprouty-2. These results attribute a novel role for carboxyl-terminal tyrosine residues and yet unidentified phosphotyrosine-binding proteins in the differential regulation of Sprouty-2 activity.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Fosfotirosina/metabolismo , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células CHO , Células COS , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Cisteína , Proteína Adaptadora GRB2 , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas do Tecido Nervoso/genética , Fosforilação , Ligação Proteica , Interferência de RNA , Ratos , Proteínas Recombinantes/metabolismo , Transfecção
19.
Proc Natl Acad Sci U S A ; 101(5): 1229-34, 2004 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-14742870

RESUMO

Receptor tyrosine kinases (RTKs) control a multitude of biological processes and are therefore subjected to multiple levels of regulation. Negative feedback is one of the mechanisms that provide an effective means to control RTK-mediated signaling. Sef has recently been identified as a specific antagonist of fibroblast growth factor (FGF) signaling in zebrafish and subsequently in mouse and human. Sef encodes a putative type I transmembrane protein that antagonizes the Ras/mitogen-activated protein kinase pathway in all three species. Mouse Sef was also shown to inhibit the phosphatidylinositol 3-kinase pathway. We show here that an alternative splicing mechanism generates an isoform of human Sef, hSef-b, which unlike the previously reported Sef (hSef-a) is a cytosolic protein. Contrary to hSef-a, which is ubiquitously expressed, hSef-b transcripts display a restricted pattern of expression in human tissues. hSef-b inhibits FGF-induced cell proliferation and prevents the activation of mitogen-activated protein kinase without affecting the upstream component MAPK kinase. Furthermore, hSef-b does not antagonize FGF induction of the phosphatidylinositol 3-kinase pathway. In addition to the effects on FGF signaling, hSef-b inhibited cellular response to platelet-derived growth factor but not other RTK ligands. Therefore, alternative splicing of the hSef gene expands the Sef feedback inhibition repertoire of RTK signaling.


Assuntos
Receptores de Interleucina/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , Divisão Celular , Clonagem Molecular , Ciclina D1/análise , Sistema de Sinalização das MAP Quinases , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Especificidade de Órgãos , Isoformas de Proteínas
20.
FEBS Lett ; 552(2-3): 150-4, 2003 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-14527678

RESUMO

Receptor binding specificity is an essential element in regulating the diverse activities of fibroblast growth factors (FGFs). FGF7 is ideal to study how this specificity is conferred at the structural level, as it interacts exclusively with one isoform of the FGF-receptor (FGFR) family, known as FGFR2IIIb. Previous mutational analysis suggested the importance of the beta4/beta5 loop of FGF7 in specific receptor recognition. Here a theoretical model of FGFR2IIIb/FGF7 complex showed that this loop interacts with the FGFR2IIIb unique exon. In addition, the model revealed new residues that either directly interact with the FGFR2IIIb unique exon (Asp63, Leu142) or facilitate this interaction (Arg65). Mutations in these residues reduced both receptor binding affinity and biological activity of FGF7. Altogether, these results provide the basis for understanding how receptor-binding specificity of FGF7 is conferred at the structural level.


Assuntos
Fatores de Crescimento de Fibroblastos/química , Fatores de Crescimento de Fibroblastos/genética , Receptores Proteína Tirosina Quinases/química , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/química , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Células 3T3 , Processamento Alternativo , Animais , Sítios de Ligação , Linhagem Celular , Éxons , Fator 7 de Crescimento de Fibroblastos , Fatores de Crescimento de Fibroblastos/metabolismo , Cinética , Substâncias Macromoleculares , Camundongos , Modelos Moleculares , Mutagênese , Mutação Puntual , Estrutura Terciária de Proteína , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...