Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Vascul Pharmacol ; 153: 107246, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38040222

RESUMO

CTHRC1 is transiently expressed by activated fibroblasts during tissue repair and in certain cancers, and CTHRC1 derived from osteocytes is detectable in circulation. Because its biological activity is poorly understood, we investigated whether the N terminus of CTHRC1 encodes a propeptide requiring cleavage to become activated. The effects of full-length versus cleaved recombinant CTHRC1 on endothelial cell metabolism and gene expression were examined in vitro. Respirometry was performed on Cthrc1 null and wildtype mice to obtain evidence for biological activity of CTHRC1 in vivo. Cleavage of the propeptide observed in vitro was attenuated in the presence of protease inhibitors, and cleaved CTHRC1 significantly promoted glycolysis whereas full-length CTHRC1 was less effective. The respiratory exchange ratio was significantly higher in wildtype mice compared to Cthrc1 null mice, supporting the findings of CTHRC1 promoting glycolysis in vivo. Key enzymes involved in glycolysis were significantly upregulated in endothelial cells in response to treatment with CTHRC1. In healthy human subjects, 58% of the cohort had detectable levels of circulating full-length CTHRC1, whereas all subjects with undetectable levels of full-length CTHRC1 (with one exception) had measurable levels of truncated CTHRC1 (88 pg/ml to >400 ng/ml). Our findings support a concept where CTHRC1 induction in activated fibroblasts at sites of ischemia such as tissue injury or cancer functions to increase glycolysis for ATP production under hypoxic conditions, thereby promoting cell survival and tissue repair. By promoting glycolysis under normoxic conditions, CTHRC1 may also be a contributor to the Warburg effect characteristically observed in many cancers.


Assuntos
Proteínas da Matriz Extracelular , Neoplasias , Animais , Humanos , Camundongos , Angiogênese , Células Endoteliais/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Camundongos Knockout
2.
Transfusion ; 63 Suppl 3: S168-S176, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37070378

RESUMO

INTRODUCTION: Tranexamic acid (TXA) is widely used as an antifibrinolytic agent in hemorrhagic trauma patients. The beneficial effects of TXA exceed the suppression of blood loss and include the ability to decrease inflammation and edema. We found that TXA suppresses the release of mitochondrial DNA and enhances mitochondrial respiration. These results allude that TXA could operate through plasmin-independent mechanisms. To address this hypothesis, we compared the effects of TXA on lipopolysaccharide (LPS)-induced expression of proinflammatory cytokines in plasminogen (Plg) null and Plg heterozygous mice. METHODS: Plg null and Plg heterozygous mice were injected with LPS and TXA or LPS only. Four hours later, mice were sacrificed and total RNA was prepared from livers and hearts. Real time quantitative polymerase chain reaction with specific primers was used to assess the effects of LPS and TXA on the expression of pro-inflammatory cytokines. RESULTS: LPS enhanced the expression of Tnfα in the livers and hearts of recipient mice. The co-injection of TXA significantly decreased the effect of LPS both in Plg null and heterozygous mice. A similar trend was observed with LPS-induced Il1α expression in hearts and livers. CONCLUSIONS: The effects of TXA on the endotoxin-stimulated expression of Tnfα and Il1α in mice do not depend on the inhibition of plasmin generation. These results indicate that TXA has other biologically important target(s) besides plasminogen/plasmin. Fully understanding the molecular mechanisms behind the extensive beneficial effects of TXA and future identification of its targets may lead to improvement in the use of TXA in trauma, cardiac, and orthopedic surgical patients.


Assuntos
Antifibrinolíticos , Ácido Tranexâmico , Camundongos , Animais , Ácido Tranexâmico/farmacologia , Fibrinolisina , Fibrinolíticos , Endotoxinas , Fator de Necrose Tumoral alfa/genética , Lipopolissacarídeos/farmacologia , Antifibrinolíticos/farmacologia , Plasminogênio/genética , Plasminogênio/metabolismo , Hemorragia
3.
J Surg Res ; 251: 287-295, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32199337

RESUMO

BACKGROUND: The endothelial glycocalyx (EG) is involved in critical regulatory mechanisms that maintain endothelial vascular integrity. We hypothesized that prolonged cardiopulmonary bypass (CPB) may be associated with EG degradation. We performed an analysis of soluble syndecan-1 levels in relation to duration of CPB, as well as factors associated with cell stress and damage, such as mitochondrial DNA (mtDNA) and inflammation. METHODS: Blood samples from subjects undergoing cardiac surgery with CPB (n = 54) were obtained before and during surgery, 4-8 h and 24 h after completion of CPB, and on postoperative day 4. Flow cytometry was used to determine subpopulations of white blood cells. Plasma levels of mtDNA were determined using quantitative polymerase chain reaction and plasma content of shed syndecan-1 was measured. To determine whether syndecan-1 was signaling white blood cells, the effect of recombinant syndecan-1 on mobilization of neutrophils from bone marrow was tested in mice. RESULTS: CPB is associated with increased mtDNA during surgery, increased syndecan-1 blood levels at 4-8 h, and increased white blood cell count at 4-8 h and 24 h. Correlation analysis revealed significant positive associations between time on CPB and syndecan-1 (rs = 0.488, P < 0.001) and level of syndecan-1 and neutrophil count (rs = 0.351, P = 0.038) at 4-8 h. Intravenous administration of recombinant syndecan-1 in mice resulted in a 2.5-fold increase in the number of circulating neutrophils, concurrent with decreased bone marrow neutrophil number. CONCLUSIONS: Longer duration of CPB is associated with increased plasma levels of soluble syndecan-1, a signal for EG degradation, which can induce neutrophil egress from the bone marrow. Development of therapy targeting EG shedding may be beneficial in patients with prolonged CPB.


Assuntos
Ponte Cardiopulmonar/efeitos adversos , Endotélio/ultraestrutura , Glicocálix/fisiologia , Duração da Cirurgia , Idoso , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Ponte Cardiopulmonar/métodos , DNA Mitocondrial/sangue , Feminino , Humanos , Interleucina-6/sangue , Contagem de Leucócitos , Masculino , Camundongos , Pessoa de Meia-Idade , Neutrófilos/patologia , Proteínas Recombinantes/farmacologia , Sindecana-1/sangue , Sindecana-1/farmacologia
4.
J Trauma Acute Care Surg ; 86(4): 617-624, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30589751

RESUMO

BACKGROUND: Severe burn injuries are known to initiate a profound systemic inflammatory response (SIRS) that may lead to burn shock and other SIRS-related complications. Damage-associated molecular patterns (DAMPs) are important early signaling molecules that initiate SIRS after burn injury. Previous work in a rodent model has shown that application of a topical immune modulator (p38MAPK inhibitor) applied directly to the burn wound decreases cytokine expression, reduces pulmonary inflammation and edema. Our group has demonstrated that tranexamic acid (TXA)-in addition to its use as an antifibrinolytic-has cell protective in vitro effects. We hypothesized that administration of TXA after burn injury would attenuate DAMP release and reduce lung inflammation. METHODS: C57/BL6 male mice underwent a 40% Total Body Surface Area (TBSA) scald burn. Sham animals underwent the same procedure in room temperature water. One treatment group received the topical application of p38MAPK inhibitor after burn injury. The other treatment group received an intraperitoneal administration of TXA after burn injury. Animals were sacrificed at 5 hours. Plasma was collected by cardiac puncture. MtDNA levels in plasma were determined by quantitative Polymerase Chain Reaction (qPCR). Syndecan-1 levels in plasma were measured by ELISA. Lungs were harvested, fixed, and paraffin-embedded. Sections of lungs were stained for antigen to detect macrophages. RESULTS: Topical p38MAPK inhibitor and TXA significantly attenuated mtDNA release. Both TXA and the topical p38MAPK inhibitor reduced lung inflammation as represented by decreased macrophage infiltration. Syndecan-1 levels showed no difference between burn and treatment groups. CONCLUSION: Both p38 MAPK inhibitor and TXA demonstrated the ability to attenuate burn-induced DAMP release and lung inflammation. Beyond its role as an antifibrinolytic, TXA may have significant anti-inflammatory effects pertinent to burn resuscitation. Further study is required; however, TXA may be a useful adjunct in burn resuscitation.


Assuntos
Alarminas/efeitos dos fármacos , Queimaduras/tratamento farmacológico , Queimaduras/fisiopatologia , Modelos Animais de Doenças , Mitocôndrias/efeitos dos fármacos , Pneumonia/tratamento farmacológico , Ácido Tranexâmico/farmacologia , Administração Tópica , Animais , DNA Mitocondrial/antagonistas & inibidores , DNA Mitocondrial/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
5.
J Cell Biochem ; 119(3): 2636-2645, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29058774

RESUMO

Highly variable expression of mesoderm-specific transcript (Mest) in adipose tissue among genetically homogeneous mice fed an obesogenic diet, and its positive association with fat mass expansion, suggests that Mest is an epigenetic determinant for the development of obesity. Although the mechanisms by which MEST augments fat accumulation in adipocytes have not been elucidated, it has sequence homology and catalytic peptide motifs which suggests that it functions as an epoxide hydrolase or as a glycerol- or acylglycerol-3-phosphate acyltransferase. To better understand MEST function, detailed studies were performed to precisely define the intracellular organelle localization of MEST using immunofluorescence confocal microscopy. Lentiviral-mediated expression of a C-terminus Myc-DDK-tagged MEST fusion protein expressed in 3T3-L1 preadipocytes/adipocytes, and ear-derived mesenchymal stem cells (EMSC) from mice was observed in the endoplasmic reticulum (ER) membranes and is consistent with previous studies showing endogenous MEST in the membrane fraction of adipose tissue. MEST was not associated with the Golgi apparatus or mitochondria; however, frequent contacts were observed between MEST-positive ER and mitochondria. MEST-positive domains were also shown on the plasma membrane (PM) of non-permeabilized cells but they did not co-localize with ER-PM bridges. Post-adipogenic differentiated 3T3-L1 adipocytes and EMSC showed significant co-localization of MEST with the lipid droplet surface marker perilipin at contact points between the ER and lipid droplet. Identification of MEST as an ER-specific protein that co-localizes with lipid droplets in cells undergoing adipogenic differentiation supports a function for MEST in the facilitation of lipid accumulation and storage in adipocytes.


Assuntos
Adipócitos/metabolismo , Retículo Endoplasmático/metabolismo , Gotículas Lipídicas/metabolismo , Proteínas/metabolismo , Células 3T3-L1 , Adipócitos/patologia , Animais , Diferenciação Celular/fisiologia , Hipertrofia/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Knockout , Obesidade/metabolismo
6.
Biochemistry ; 55(7): 1159-67, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26836284

RESUMO

Fibroblast growth factor 1 (FGF1), a ubiquitously expressed pro-angiogenic protein that is involved in tissue repair, carcinogenesis, and maintenance of vasculature stability, is released from the cells via a stress-dependent nonclassical secretory pathway. FGF1 secretion is a result of transmembrane translocation of this protein. It correlates with the ability of FGF1 to permeabilize membranes composed of acidic phospholipids. Like several other nonclassically exported proteins, FGF1 exhibits ß-barrel folding. To assess the role of folding of FGF1 in its secretion, we applied targeted mutagenesis in combination with a complex of biophysical methods and molecular dynamics studies, followed by artificial membrane permeabilization and stress-induced release experiments. It has been demonstrated that a mutation of proline 135 located in the C-terminus of FGF1 results in (i) partial unfolding of FGF1, (ii) a decrease in FGF1's ability to permeabilize bilayers composed of phosphatidylserine, and (iii) drastic inhibition of stress-induced FGF1 export. Thus, folding of FGF1 is critical for its nonclassical secretion.


Assuntos
Permeabilidade da Membrana Celular , Fator 1 de Crescimento de Fibroblastos/química , Modelos Moleculares , Dobramento de Proteína , Substituição de Aminoácidos , Animais , Varredura Diferencial de Calorimetria , Fator 1 de Crescimento de Fibroblastos/genética , Fator 1 de Crescimento de Fibroblastos/metabolismo , Células HEK293 , Humanos , Cinética , Bicamadas Lipídicas/química , Membranas Artificiais , Camundongos , Simulação de Dinâmica Molecular , Mutação , Células NIH 3T3 , Permeabilidade , Fosfatidilserinas/química , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
7.
J Cell Biochem ; 116(8): 1522-31, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25560297

RESUMO

FGF1 is a nonclassically released growth factor that regulates carcinogenesis, angiogenesis, and inflammation. In vitro and in vivo, FGF1 export is stimulated by cell stress. Upon stress, FGF1 is transported to the plasma membrane where it localizes prior to transmembrane translocation. To determine which proteins participate in the submembrane localization of FGF1 and its export, we used immunoprecipitation mass spectrometry to identify novel proteins that associate with FGF1 during heat shock. The heat shock-dependent association of FGF1 with the large protein AHNAK2 was observed. Heat shock induced the translocation of FGF1 and AHNAK2 to the cytoskeletal fraction. In heat-shocked cells, FGF1 and the C-terminal fragment of AHNAK2 colocalized with F-actin in the vicinity of the cell membrane. Depletion of AHNAK2 resulted in a drastic decrease of stress-induced FGF1 export but did not affect spontaneous FGF2 export and FGF1 release induced by the inhibition of Notch signaling. Thus, AHNAK2 is an important element of the FGF1 nonclassical export pathway.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Via Secretória , Estresse Fisiológico , Actinas/metabolismo , Animais , Membrana Celular/metabolismo , Proteínas do Citoesqueleto/química , Humanos , Espectrometria de Massas , Camundongos , Células NIH 3T3 , Temperatura
8.
J Cell Biochem ; 115(5): 874-88, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24375433

RESUMO

FGF applied as a single growth factor to quiescent mouse fibroblasts induces a round of DNA replication, however continuous stimulation results in arrest in the G1 phase of the next cell cycle. We hypothesized that FGF stimulation induces the establishment of cell memory, which prevents the proliferative response to repeated or continuous FGF application. When a 2-5 days quiescence period was introduced between primary and repeated FGF treatments, fibroblasts failed to efficiently replicate in response to secondary FGF application. The establishment of "FGF memory" during the first FGF stimulation did not require DNA synthesis, but was dependent on the activity of FGF receptors, MEK, p38 MAPK and NFκB signaling, and protein synthesis. While secondary stimulation resulted in strongly decreased replication rate, we did not observe any attenuation of morphological changes, Erk1/2 phosphorylation and cyclin D1 induction. However, secondary FGF stimulation failed to induce the expression of cyclin A, which is critical for the progression from G1 to S phase. Treatment of cells with a broad range histone deacetylase inhibitor during the primary FGF stimulation rescued the proliferative response to the secondary FGF treatment suggesting that the establishment of "FGF memory" may be based on epigenetic changes. We suggest that "FGF memory" can prevent the hyperplastic response to cell damage and inflammation, which are associated with an enhanced FGF production and secretion. "FGF memory" may present a natural obstacle to the efficient application of recombinant FGFs for the treatment of ulcers, ischemias, and wounds.


Assuntos
Ciclo Celular/genética , Movimento Celular/genética , Proliferação de Células , Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Ciclina D1/genética , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/genética , Fatores de Crescimento de Fibroblastos/administração & dosagem , Fase G1/genética , Histona Desacetilases/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Camundongos , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Mol Cancer Res ; 10(2): 230-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22172337

RESUMO

UNLABELLED: Prostate carcinoma is among the most common causes of cancer-related death in men, representing 15% of all male malignancies in developed countries. Neuroendocrine differentiation (NED) has been associated with tumor progression, poor prognosis, and with the androgen-independent status. Currently, no successful therapy exists for advanced, castration-resistant disease. Because hypoxia has been linked to prostate cancer progression and unfavorable outcome, we sought to determine whether hypoxia would impact the degree of neuroendocrine differentiation of prostate cancer cells in vitro. RESULTS: Exposure of LNCaP cells to low oxygen tension induced a neuroendocrine phenotype, associated with an increased expression of the transcription factor neurogenin3 and neuroendocrine markers, such as neuron-specific enolase, chromogranin A, and ß3-tubulin. Moreover, hypoxia triggered a significant decrease of Notch 1 and Notch 2 mRNA and protein expression, with subsequent downregulation of Notch-mediated signaling, as shown by reduced levels of the Notch target genes, Hes1 and Hey1. NED was promoted by attenuation of Hes1 transcription, as cells expressing a dominant-negative form of Hes1 displayed increased levels of neuroendocrine markers under normoxic conditions. Although hypoxia downregulated Notch 1 and Notch 2 mRNA transcription and receptor activation also in the androgen-independent cell lines, PC-3 and Du145, it did not change the extent of NED in these cultures, suggesting that androgen sensitivity may be required for transdifferentiation to occur. CONCLUSIONS: Hypoxia induces NED of LNCaP cells in vitro, which seems to be driven by the inhibition of Notch signaling with subsequent downregulation of Hes1 transcription.


Assuntos
Androgênios/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Hipóxia , Proteínas do Tecido Nervoso/metabolismo , Neoplasias da Próstata/metabolismo , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Proteínas do Tecido Nervoso/genética , Tumores Neuroendócrinos/metabolismo , Oxigênio/administração & dosagem , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Receptor Notch1/genética , Receptor Notch2/genética , Fatores de Transcrição HES-1
10.
J Cell Physiol ; 226(11): 3064-75, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21302306

RESUMO

FGF1, a widely expressed proangiogenic factor involved in tissue repair and carcinogenesis, is released from cells through a non-classical pathway independent of endoplasmic reticulum and Golgi. Although several proteins participating in FGF1 export were identified, genetic mechanisms regulating this process remained obscure. We found that FGF1 export and expression are regulated through Notch signaling mediated by transcription factor CBF1 and its partner MAML. The expression of a dominant negative (dn) form of CBF1 in 3T3 cells induces transcription of FGF1 and sphingosine kinase 1 (SphK1), which is a component of FGF1 export pathway. dnCBF1 expression stimulates the stress-independent release of transduced FGF1 from NIH 3T3 cells and endogenous FGF1 from A375 melanoma cells. NIH 3T3 cells transfected with dnCBF1 form colonies in soft agar and produce rapidly growing highly angiogenic tumors in nude mice. The transformed phenotype of dnCBF1 transfected cells is efficiently blocked by dn forms of FGF receptor 1 and S100A13, which is a component of FGF1 export pathway. FGF1 export and acceleration of cell growth induced by dnCBF1 depend on SphK1. Similar to dnCBF1, dnMAML transfection induces FGF1 expression and release, and accelerates cell proliferation. The latter effect is strongly decreased in FGF1 null cells. We suggest that the regulation of FGF1 expression and release by CBF1-mediated Notch signaling can play an important role in tumor formation.


Assuntos
Transformação Celular Neoplásica/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Receptores Notch/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Humanos , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Camundongos , Camundongos Nus , Células NIH 3T3 , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Proteínas Nucleares/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas S100/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Transfecção
11.
Mol Biol Cell ; 19(11): 4863-74, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18784255

RESUMO

Angiogenesis is controlled by several regulatory mechanisms, including the Notch and fibroblast growth factor (FGF) signaling pathways. FGF1, a prototype member of FGF family, lacks a signal peptide and is released through an endoplasmic reticulum-Golgi-independent mechanism. A soluble extracellular domain of the Notch ligand Jagged1 (sJ1) inhibits Notch signaling and induces FGF1 release. Thrombin, a key protease of the blood coagulation cascade and a potent inducer of angiogenesis, stimulates rapid FGF1 release through a mechanism dependent on the major thrombin receptor protease-activated receptor (PAR) 1. This study demonstrates that thrombin cleaves Jagged1 in its extracellular domain. The sJ1 form produced as a result of thrombin cleavage inhibits Notch-mediated CBF1/Suppressor of Hairless [(Su(H)]/Lag-1-dependent transcription and induces FGF1 expression and release. The overexpression of Jagged1 in PAR1 null cells results in a rapid thrombin-induced export of FGF1. These data demonstrate the existence of novel cross-talk between thrombin, FGF, and Notch signaling pathways, which play important roles in vascular formation and remodeling.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Fármacos Cardiovasculares/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Fragmentos de Peptídeos/metabolismo , Trombina/farmacologia , Animais , Proteínas de Ligação ao Cálcio/química , Linhagem Celular , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fator 1 de Crescimento de Fibroblastos/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/química , Proteína Jagged-1 , Proteínas de Membrana/química , Camundongos , Peso Molecular , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/efeitos dos fármacos , Células-Tronco Multipotentes/metabolismo , Crista Neural/citologia , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Receptor PAR-1/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Receptores Notch/metabolismo , Receptores de Trombina , Proteínas Serrate-Jagged , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
12.
Am J Pathol ; 173(3): 865-78, 2008 09.
Artigo em Inglês | MEDLINE | ID: mdl-18688026

RESUMO

We previously found that soluble forms of the Notch ligands Jagged1 and Delta1 induced fibroblast growth factor receptor-dependent cell transformation in NIH3T3 fibroblasts. However, the phenotypes of these lines differed, indicating distinct functional differences among these Notch ligands. In the present study, we used allografts to test the hypothesis that NIH3T3 fibroblasts that express soluble forms of Delta1 and Jagged1 accelerate tumorigenicity in vivo. With the exception of the full-length Jagged1 transfectant, all other cell lines, including the control, generated tumors when injected subcutaneously in athymic mice. Suppression of Notch signaling by the soluble ligands significantly increased tumor onset and growth, whereas full-length Jagged1 completely suppressed tumor development. In addition, there were striking differences in tumor pathology with respect to growth kinetics, vascularization, collagen content, size and number of necrotic foci, and invasiveness into the underlying tissue. Further, the production of angiogenic factors, including vascular endothelial growth factor, also differed among the tumor types. Lastly, both Jagged1- and Delta1-derived tumors contained phenotypically distinct populations of lipid-filled cells that corresponded with increased expression of adipocyte markers. The divergence of tumor phenotype may be attributed to ligand-specific alterations in Notch receptor responses in exogenous and endogenous cell populations within the allographs. Our findings demonstrate distinct functional properties for these Notch ligands in the promotion of tumorigenicity in vivo.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Transformação Celular Neoplásica/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias Experimentais/metabolismo , Fenótipo , Animais , Western Blotting , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Fibroblastos/metabolismo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intracelular , Proteína Jagged-1 , Imageamento por Ressonância Magnética , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neovascularização Patológica/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Serrate-Jagged , Transfecção
13.
Exp Cell Res ; 313(15): 3308-18, 2007 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-17643421

RESUMO

Sphingosine kinase 1 catalyzes the formation of sphingosine-1-phosphate, a lipid mediator involved in the regulation of angiogenesis. Sphingosine kinase 1 is constitutively released from cells, even though it lacks a classical signal peptide sequence. Because copper-dependent non-classical stress-induced release of FGF1 also regulates angiogenesis, we questioned whether sphingosine kinase 1 is involved in the FGF1 release pathway. We report that (i) the coexpression of sphingosine kinase 1 with FGF1 inhibited the release of sphingosine kinase 1 at 37 degrees C; (ii) sphingosine kinase 1 was released at 42 degrees C in complex with FGF1; (iii) sphingosine kinase 1 null cells failed to release FGF1 at stress; (iv) sphingosine kinase 1 is a high affinity copper-binding protein which formed a complex with FGF1 in a cell-free system, and (v) sphingosine kinase 1 over expression rescued the release of FGF1 from inhibition by the copper chelator, tetrathiomolybdate. We propose that sphingosine kinase 1 is a component of the copper-dependent FGF1 release pathway.


Assuntos
Cobre/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Células Cultivadas , Quelantes/farmacologia , Clonagem Molecular , Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Molibdênio/farmacologia , Células NIH 3T3 , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Transporte Proteico , Temperatura
14.
Biochem Biophys Res Commun ; 350(3): 604-9, 2006 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-17027650

RESUMO

Thrombin induces cell proliferation and migration during vascular injury. We report that thrombin rapidly stimulated expression and release of the pro-angiogenic polypeptide fibroblast growth factor 1 (FGF1). Thrombin failed to induce FGF1 release from protease-activated receptor 1 (PAR1) null fibroblasts, indicating that this effect was dependent on PAR1. Similarly to thrombin, FGF1 expression and release were induced by TRAP, a specific oligopeptide agonist of PAR1. These results identify a novel aspect of the crosstalk between FGF and thrombin signaling pathways which both play important roles in tissue repair and angiogenesis.


Assuntos
Fator 1 de Crescimento de Fibroblastos/metabolismo , Receptor PAR-1/metabolismo , Transdução de Sinais/fisiologia , Trombina/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Camundongos , Células NIH 3T3 , Transdução de Sinais/efeitos dos fármacos
15.
J Biol Chem ; 279(26): 27440-9, 2004 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-15084601

RESUMO

Mutations in the human endoglin gene result in hereditary hemorrhagic telangiectasia type 1, a vascular disorder characterized by multisystemic vascular dysplasia, arteriovenous malformations, and focal dilatation of postcapillary venules. Previous studies have implicated endoglin in the inhibition of cell migration in vivo and in vitro. In the course of studies to address the relationship of the conserved cytosolic domain to endoglin function, we identified zyxin, a LIM domain protein that is concentrated at focal adhesions, as an interactor with endoglin in human umbilical vein vascular endothelial cells. This interaction is localized within the 47-amino acid carboxyl-terminal cytosolic domain of endoglin, and maps within zyxin residues 326-572. The endoglin-zyxin interaction was found to be largely mediated by the third LIM domain of zyxin, and is specific for endoglin because the homologous cytosolic domain of the transforming growth factor-beta type III receptor, betaglycan, fails to interact with zyxin. Expression of endoglin is associated with reduction of zyxin, as well as its interacting proteins p130(cas) and CrkII, from a focal adhesion protein fraction, and this reduction is correlated with inhibition of cell migration. We also show that endoglin-dependent: (i) inhibition of cell migration, (ii) reduction of focal adhesion-associated p130(cas)/CrkII protein levels, (iii) tyrosine phosphorylation of p130(cas), and (iv) focal adhesion-associated endoglin levels are mediated by the cytosolic domain of endoglin. These results suggest a novel mechanism of endoglin function involving its interaction with LIM domain-containing proteins, and associated adapter proteins, affecting sites of focal adhesion.


Assuntos
Movimento Celular/fisiologia , Adesões Focais/fisiologia , Molécula 1 de Adesão de Célula Vascular/fisiologia , Animais , Antígenos CD , Linhagem Celular , Proteínas do Citoesqueleto , Citosol/metabolismo , Endoglina , Deleção de Genes , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Camundongos , Microscopia de Fluorescência , Microesferas , Mutagênese Sítio-Dirigida , Oligopeptídeos/metabolismo , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de Superfície Celular , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína do Retinoblastoma/metabolismo , Transfecção , Tirosina/metabolismo , Molécula 1 de Adesão de Célula Vascular/química , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo , Zixina
16.
J Biol Chem ; 278(18): 16405-13, 2003 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-12598523

RESUMO

Aberrant activations of the Notch and fibroblast growth factor receptor (FGFR) signaling pathways have been correlated with neoplastic growth in humans and other mammals. Here we report that the suppression of Notch signaling in NIH 3T3 cells by the expression of either the extracellular domain of the Notch ligand Jagged1 or dominant-negative forms of Notch1 and Notch2 results in the appearance of an exaggerated fibroblast growth factor (FGF)-dependent transformed phenotype characterized by anchorage-independent growth in soft agar. Anchorage-independent growth exhibited by Notch-repressed NIH 3T3 cells may result from prolonged FGFR stimulation caused by both an increase in the expression of prototypic and oncogenic FGF gene family members and the nonclassical export of FGF1 into the extracellular compartment. Interestingly, FGF exerts a negative effect on Notch by suppressing CSL (CBF-1/RBP-Jk/KBF2 in mammals, Su(H) in Drosophila and Xenopus, and Lag-2 in Caenorhabditis elegans)-dependent transcription, and the ectopic expression of constitutively active forms of Notch1 or Notch2 abrogates FGF1 release and the phenotypic effects of FGFR stimulation. These data suggest that communication between the Notch and FGFR pathways may represent an important reciprocal autoregulatory mechanism for the regulation of normal cell growth.


Assuntos
Transformação Celular Neoplásica , Fator 1 de Crescimento de Fibroblastos/fisiologia , Proteínas de Membrana/fisiologia , Proteínas , Células 3T3 , Animais , Proteínas de Ligação ao Cálcio , Divisão Celular , Proteínas de Drosophila , Fator 1 de Crescimento de Fibroblastos/genética , Fibroblastos/citologia , Peptídeos e Proteínas de Sinalização Intercelular , Proteína Jagged-1 , Glicoproteínas de Membrana/fisiologia , Camundongos , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Receptores Notch , Proteínas Serrate-Jagged , Proteínas de Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA