Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Sci ; 126(Pt 20): 4746-55, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23943874

RESUMO

FMS-like tyrosine kinase 3 with internal tandem duplication (FLT3 ITD) is an important oncoprotein in acute myeloid leukemia (AML). Owing to its constitutive kinase activity FLT3 ITD partially accumulates at endomembranes, a feature shared with other disease-associated, mutated receptor tyrosine kinases. Because Ras proteins also transit through endomembranes we have investigated the possible existence of an intracellular FLT3-ITD/Ras signaling pathway by comparing Ras signaling of FLT3 ITD with that of wild-type FLT3. Ligand stimulation activated both K- and N-Ras in cells expressing wild-type FLT3. Live-cell Ras-GTP imaging revealed ligand-induced Ras activation at the plasma membrane (PM). FLT3-ITD-dependent constitutive activation of K-Ras and N-Ras was also observed primarily at the PM, supporting the view that the PM-resident pool of FLT3 ITD engaged the Ras/Erk pathway in AML cells. Accordingly, specific interference with FLT3-ITD/Ras signaling at the PM using PM-restricted dominant negative K-RasS17N potently inhibited cell proliferation and promoted apoptosis. In conclusion, Ras signaling is crucial for FLT3-ITD-dependent cell transformation and FLT3 ITD addresses PM-bound Ras despite its pronounced mislocalization to endomembranes.


Assuntos
Leucemia Mieloide Aguda/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo , Proteínas ras/metabolismo , Animais , Processos de Crescimento Celular/fisiologia , Membrana Celular/genética , Membrana Celular/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Genes ras , Humanos , Leucemia Mieloide Aguda/genética , Camundongos , Fosforilação , Transdução de Sinais , Sequências de Repetição em Tandem , Células Tumorais Cultivadas , Tirosina Quinase 3 Semelhante a fms/genética , Proteínas ras/genética
2.
J Stroke Cerebrovasc Dis ; 24(4): 834-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25724246

RESUMO

BACKGROUND: Stroke can cause hemilateral paresis of the tongue. The present study investigated the functional consequences of a lingual hemiparalysis on the symmetry and the grooving of the tongue in the coronal plane during the production of vowel-consonant-vowel sequences. The hypotheses were that, because of the lingual hemiparalysis, the stroke patients' tongue shapes would be (1) more asymmetrical and (2) less grooved than the tongues of the control speakers. METHODS: The participants in this prospective data collection were 9 stroke patients with lingual hemiparalysis and 6 control speakers. All participants produced vowel-consonant-vowel sequences with the vowels [a, i, and u] and the target consonants [k, t, ∫, s, and r]. The tongue shape in the coronal plane was traced and measured. The outcome measures were asymmetry and midlingual concavity. The participants and controls were compared using repeated measures analyses of variance with post hoc Scheffé tests. RESULTS: There were no significant differences in asymmetry. There was significantly reduced midlingual concavity for the stroke patients (F[1, 13] = 8.78; P < .05). There was also a within-subjects effect for consonant (F[4, 50] = 14.26; P < .01). Post hoc testing with Scheffé tests indicated that the consonant [k] had significantly lower grooving than the other consonant sounds (P < .05). CONCLUSIONS: The hemilateral paresis affected not the symmetry but the midlingual grooving. Residual ipsilateral innervation in the hemiparalyzed tongue may help patients compensate. More research is needed to assess the impact of the intrinsic deformation of the tongue on speech acceptability and intelligibility in patients with a lingual hemiparalysis.


Assuntos
Paresia/etiologia , Paresia/patologia , Acidente Vascular Cerebral/complicações , Língua/diagnóstico por imagem , Língua/fisiopatologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Reprodutibilidade dos Testes , Ultrassonografia , Adulto Jovem
3.
EMBO J ; 29(8): 1377-88, 2010 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-20224550

RESUMO

The vascular endothelial growth factors VEGFA and VEGFC are crucial regulators of vascular development. They exert their effects by dimerization and activation of the cognate receptors VEGFR2 and VEGFR3. Here, we have used in situ proximity ligation to detect receptor complexes in intact endothelial cells. We show that both VEGFA and VEGFC potently induce formation of VEGFR2/-3 heterodimers. Receptor heterodimers were found in both developing blood vessels and immature lymphatic structures in embryoid bodies. We present evidence that heterodimers frequently localize to tip cell filopodia. Interestingly, in the presence of VEGFC, heterodimers were enriched in the leading tip cells as compared with trailing stalk cells of growing sprouts. Neutralization of VEGFR3 to prevent heterodimer formation in response to VEGFA decreased the extent of angiogenic sprouting. We conclude that VEGFR2/-3 heterodimers on angiogenic sprouts induced by VEGFA or VEGFC may serve to positively regulate angiogenic sprouting.


Assuntos
Células Endoteliais/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Embrião de Mamíferos/metabolismo , Endotélio Vascular/metabolismo , Humanos , Neovascularização Fisiológica , Multimerização Proteica , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
4.
Biochem Soc Trans ; 40(1): 20-5, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22260660

RESUMO

VEGFs (vascular endothelial growth factors) are master regulators of vascular development and of blood and lymphatic vessel function during health and disease in adults. This family of five mammalian ligands acts through three RTKs (receptor tyrosine kinases). In addition, co-receptors such as NRPs (neuropilins) associate with the ligand-receptor signalling complex and modulate the output. Therapeutics to block several of the VEGF signalling components as well as NRP function have been developed with the aim of halting blood vessel formation, angiogenesis, in diseases that involve tissue growth and inflammation, such as cancer. The present review outlines the current understanding of NRPs in relation to blood and lymphatic vessel biology.


Assuntos
Neovascularização Fisiológica , Neuropilinas/fisiologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Humanos , Terapia de Alvo Molecular , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neuropilinas/genética , Neuropilinas/metabolismo , Ligação Proteica , Isoformas de Proteínas/metabolismo
5.
Biochem J ; 437(2): 169-83, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21711246

RESUMO

VEGFs (vascular endothelial growth factors) control vascular development during embryogenesis and the function of blood vessels and lymphatic vessels in the adult. There are five related mammalian ligands, which act through three receptor tyrosine kinases. Signalling is modulated through neuropilins, which act as VEGF co-receptors. Heparan sulfate and integrins are also important modulators of VEGF signalling. Therapeutic agents that interfere with VEGF signalling have been developed with the aim of decreasing angiogenesis in diseases that involve tissue growth and inflammation, such as cancer. The present review will outline the current understanding and consequent biology of VEGF receptor signalling.


Assuntos
Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Transdução de Sinais/fisiologia , Animais , Heparitina Sulfato/metabolismo , Humanos , Integrinas/fisiologia , Neuropilinas/fisiologia , Fator de Crescimento Placentário , Proteínas da Gravidez/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/fisiologia
6.
Blood ; 113(15): 3568-76, 2009 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-19204327

RESUMO

The mechanism of cell transformation by Fms-like tyrosine kinase 3 (FLT3) in acute myeloid leukemia (AML) is incompletely understood. The most prevalent activated mutant FLT3 ITD exhibits an altered signaling quality, including strong activation of the STAT5 transcription factor. FLT3 ITD has also been found partially retained as a high-mannose precursor in an intracellular compartment. To analyze the role of intracellular retention of FLT3 for transformation, we have generated FLT3 versions that are anchored in the perinuclear endoplasmic reticulum (ER) by appending an ER retention sequence containing a RRR (R3) motif. ER retention of R3, but not of corresponding A3 FLT3 versions, is shown by biochemical, fluorescence-activated cell sorting, and immunocytochemical analyses. ER anchoring reduced global autophosphorylation and diminished constitutive activation of ERK1/2 and AKT of the constitutively active FLT3 versions. ER anchoring was, however, associated with elevated signaling to STAT3. Transforming activity of the FLT3 D835Y mutant was suppressed by ER anchoring. In contrast, ER-anchored FLT3 ITD retained STAT5-activating capacity and was transforming in vitro and in vivo. The findings highlight another aspect of the different signaling quality of FLT3 ITD: It can transform cells from an intracellular location.


Assuntos
Retículo Endoplasmático/metabolismo , Leucemia Mieloide Aguda/metabolismo , Transdução de Sinais/fisiologia , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo , Animais , Divisão Celular/fisiologia , Linhagem Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Duplicação Gênica , Humanos , Rim/citologia , Leucemia Mieloide Aguda/genética , Camundongos , Camundongos Endogâmicos C3H , Mutagênese , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/metabolismo
7.
Ann Hematol ; 89(7): 653-62, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20119833

RESUMO

Treatment of acute myeloid leukemia (AML) remains challenging with many patients harboring unfavorable prognostic parameters such as FLT3 internal tandem duplication (FLT3-ITD) mutations leading to a constitutively activated FLT3-receptor tyrosine kinase (RTK). Activation of proteins by phosphorylation of tyrosine residues is a common mechanism in leukemia development. Therefore, specific tyrosine kinase inhibitors (TKI) have been developed for AML therapy and are currently under investigation. The staurosporine derivate PKC412 (Midostaurin) was found to be an effective inhibitor of the FLT3-RTK and is currently undergoing clinical trials for FLT3-mutated AML patients. Since resistance towards TKIs has been observed in vitro and in clinical trials, we have generated a PKC412-resistant clone (MV4-11r) of the human myelomonoblastic cell line MV4-11, which carries a homozygous FLT3-ITD mutation. MV4-11r displayed higher vitality after addition of PKC412 compared with MV4-11 with a pronounced reduction of apoptotic cells. Cytogenetic characterization revealed the acquisition of additional aberrations in the resistant cell line such as clonal alterations at chromosome 13q with additional FLT3 signals. Microarray analysis revealed significant expression changes in several genes prior to and after incubation with PKC412. The expression status of candidate genes being regulated by FLT-ITD like JAG1, p53, MCL-1, C-KIT, and FLT3/-L was confirmed by real-time PCR. In summary, resistance against PKC412 appears to be mediated by up-regulation of anti-apoptotic genes and down-regulation of proapoptotic signals as well as genes that are involved in normal and malignant hematopoiesis.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Leucemia Mieloide Aguda/enzimologia , Mutação , Inibidores de Proteínas Quinases/farmacologia , Estaurosporina/análogos & derivados , Tirosina Quinase 3 Semelhante a fms/metabolismo , Linhagem Celular Tumoral , Cromossomos Humanos Par 13/genética , Cromossomos Humanos Par 13/metabolismo , Ensaios Clínicos como Assunto , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica/genética , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , Inibidores de Proteínas Quinases/análogos & derivados , Estaurosporina/farmacologia , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Tirosina Quinase 3 Semelhante a fms/genética
8.
Protein J ; 28(2): 57-65, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19184382

RESUMO

Flt3 ligand (FL) is an early-acting hematopoietic cytokine that stimulates the proliferation and differentiation of hematopoietic progenitor cells by activating its cognate receptor, Flt3. Recently, FL was shown to potently contribute to the development and expansion of antigen-presenting dendritic cells and CD34(+) natural killer cell progenitors in vivo. Here, we report a comprehensive method for the production of bioactive recombinant human FL (rhFL) in E. coli, suitable for structural, biophysical and physiological studies. A soluble form of human FL capable of binding to the Ftl3 receptor could be overexpressed in the E. coli strain Rosetta-gami(DE3) as inclusion bodies. We have established protocols for the efficient in vitro refolding and ensuing purification of rhFL to homogeneity (>95%), with yields approaching 5 mg of pure rhFL per liter of culture. The ability of rhFL to adopt a bioactive conformation was confirmed via a cell-proliferation assay and the activation of the Flt3 receptor in the human leukemic cell line, OCI-AML3.


Assuntos
Escherichia coli/genética , Proteínas de Membrana/biossíntese , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cromatografia por Troca Iônica , Dicroísmo Circular , Clonagem Molecular , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/farmacologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Dobramento de Proteína , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Espectrometria de Massas por Ionização por Electrospray
9.
Cells Tissues Organs ; 188(1-2): 225-35, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18303245

RESUMO

Aberrant subcellular localization of mutant transmembrane receptors is increasingly acknowledged as a possible mechanism for an altered signaling quality leading to transformation. There is evidence that mutated receptor tyrosine kinases of subclass III, for example the platelet-derived growth factor receptor (PDGFR) and KIT-protein, are aberrantly localized in human cancers. In order to further analyze this phenomenon, we investigated the localization of FLT3, a subclass III receptor tyrosine kinase frequently mutated in leukemia. By immunofluorescence staining and confocal laser scanning microscopy we found that in retrovirally transduced COS7 cells, wild type FLT3 receptor protein is localized primarily at the cell surface. In contrast, a mutant FLT3 receptor protein with an internal tandem duplication (ITD) accumulates in a perinuclear region and is not detectable at the plasma membrane. Surprisingly, and in contrast to previously published data, intracellular FLT3-ITD accumulation could neither be detected in the endoplasmic reticulum (ER) nor in the Golgi apparatus. Furthermore, transient overexpression per se leads to accumulation of wild type FLT3 receptor protein in the ER in addition to surface localization, probably due to inefficient intracellular transport by the overloaded sorting machinery of the secretory pathway. Based on our data and the immature glycosylation pattern of FLT3-ITD, we speculate that the mutant protein resides most probably in an unidentified compartment of the secretory pathway between the ER and the Golgi apparatus.


Assuntos
Leucemia/enzimologia , Proteínas Mutantes/metabolismo , Tirosina Quinase 3 Semelhante a fms/química , Tirosina Quinase 3 Semelhante a fms/metabolismo , Animais , Células COS , Membrana Celular/enzimologia , Núcleo Celular/enzimologia , Chlorocebus aethiops , Retículo Endoplasmático/enzimologia , Complexo de Golgi/enzimologia , Humanos , Estrutura Terciária de Proteína , Transporte Proteico , Retroviridae , Transdução Genética
10.
Methods Mol Biol ; 1447: 217-42, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27514809

RESUMO

Spatiotemporal aspects of protein-tyrosine phosphatase (PTP) activity and interaction partners for many PTPs are elusive. We describe here an elegant and relatively simple method, in situ proximity ligation assay (in situ PLA), which can be used to address these issues. The possibility to detect endogenous unmodified proteins in situ and to visualize individual interactions with spatial resolution is the major advantage of this technique. We provide protocols suitable to monitor association of the transmembrane PTPs PTPRJ/DEP-1/CD148 and PTPRB/VE-PTP with their substrates, the receptor tyrosine kinases FMS-like tyrosine kinase 3 (FLT3/CD135), and Tie2 and vascular endothelial growth factor receptor 2 (VEGFR2), respectively. Detailed description of method development and reagents as well as highlighting of critical factors will enable the reader to apply the method successfully to other PTP-protein interactions.


Assuntos
Mapeamento de Interação de Proteínas/métodos , Proteínas Tirosina Fosfatases/metabolismo , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Imagem Óptica/métodos , Mapas de Interação de Proteínas , Receptor TIE-2/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo
11.
Dev Cell ; 28(6): 633-46, 2014 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-24656741

RESUMO

Neuropilin 1 (NRP1) modulates angiogenesis by binding vascular endothelial growth factor (VEGF) and its receptor, VEGFR2. We examined the consequences when VEGFR2 and NRP1 were expressed on the same cell (cis) or on different cells (trans). In cis, VEGF induced rapid VEGFR2/NRP1 complex formation and internalization. In trans, complex formation was delayed and phosphorylation of phospholipase Cγ (PLCγ) and extracellular regulated kinase 2 (ERK2) was prolonged, whereas ERK1 phosphorylation was reduced. Trans complex formation suppressed initiation and vascularization of NRP1-expressing mouse fibrosarcoma and melanoma. Suppression in trans required high-affinity, steady-state binding of VEGF to NRP1, which was dependent on the NRP1 C-terminal domain. Compatible with a trans effect of NRP1, quiescent vasculature in the developing retina showed continuous high NRP1 expression, whereas angiogenic sprouting occurred where NRP1 levels fluctuated between adjacent endothelial cells. Therefore, through communication in trans, NRP1 can modulate VEGFR2 signaling and suppress angiogenesis.


Assuntos
Endocitose/fisiologia , Endotélio Vascular/patologia , Fibrossarcoma/irrigação sanguínea , Melanoma Experimental/irrigação sanguínea , Neovascularização Patológica/prevenção & controle , Neuropilina-1/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Comunicação Celular , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Endotélio Vascular/metabolismo , Fibrossarcoma/metabolismo , Fibrossarcoma/prevenção & controle , Imunofluorescência , Humanos , Melanoma Experimental/metabolismo , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Fosfolipase C gama/metabolismo , Fosforilação , Estereoisomerismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Exp Hematol ; 41(5): 444-461.e4, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23377000

RESUMO

Autotaxin (ATX) has been reported to act as a motility and growth factor in a variety of cancer cells. The ATX protein acts as a secreted lysophospholipase D by converting lysophosphatidylcholine (LPC) to lysophosphatidic acid (LPA), which signals via G-protein-coupled receptors and has important functions in cell migration and proliferation. This study demonstrates that ATX expression is specifically upregulated and functionally active in acute myeloid leukemia (AML) harboring an internal tandem duplication (ITD) mutation of the FLT3 receptor gene. Moreover, ATX expression was also found in normal human CD34+ progenitor cells and selected myeloid and lymphoid subpopulations. Enforced expression of mutant FLT3-ITD by retroviral vector transduction increased ATX mRNA in selected cell lines, whereas inhibition of FLT3-ITD signaling by sublethal doses of PKC412 or SU5614 led to a significant downregulation of ATX mRNA and protein levels. In the presence of LPC, ATX expression significantly increased proliferation. LPA induced proliferation, regardless of ATX expression, and induced chemotaxis in all tested human leukemic cell lines and human CD34(+) progenitors. LPC increased chemotaxis only in cells with high expression of endogenous ATX by at least 80%, demonstrating the autocrine action of ATX. Inhibition of ATX using a small molecule inhibitor selectively induced killing of ATX-expressing cell lines and reduced motility in these cells. Our data suggest that the production of bioactive LPA through ATX is involved in controlling proliferation and migration during hematopoiesis and that deregulation of ATX contributes to the pathogenesis of AML.


Assuntos
Movimento Celular/genética , Proliferação de Células , Células-Tronco Hematopoéticas/metabolismo , Leucemia Mieloide/genética , Diester Fosfórico Hidrolases/genética , Tirosina Quinase 3 Semelhante a fms/genética , Doença Aguda , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Humanos , Indóis/farmacologia , Células K562 , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Lisofosfatidilcolinas/metabolismo , Lisofosfatidilcolinas/farmacologia , Lisofosfolipídeos/metabolismo , Lisofosfolipídeos/farmacologia , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Diester Fosfórico Hidrolases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estaurosporina/análogos & derivados , Estaurosporina/farmacologia , Sequências de Repetição em Tandem/genética
13.
Nat Commun ; 4: 1672, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23575676

RESUMO

Vascular endothelial growth factor (VEGF) guides the path of new vessel sprouts by inducing VEGF receptor-2 activity in the sprout tip. In the stalk cells of the sprout, VEGF receptor-2 activity is downregulated. Here, we show that VEGF receptor-2 in stalk cells is dephosphorylated by the endothelium-specific vascular endothelial-phosphotyrosine phosphatase (VE-PTP). VE-PTP acts on VEGF receptor-2 located in endothelial junctions indirectly, via the Angiopoietin-1 receptor Tie2. VE-PTP inactivation in mouse embryoid bodies leads to excess VEGF receptor-2 activity in stalk cells, increased tyrosine phosphorylation of VE-cadherin and loss of cell polarity and lumen formation. Vessels in ve-ptp(-/-) teratomas also show increased VEGF receptor-2 activity and loss of endothelial polarization. Moreover, the zebrafish VE-PTP orthologue ptp-rb is essential for polarization and lumen formation in intersomitic vessels. We conclude that the role of Tie2 in maintenance of vascular quiescence involves VE-PTP-dependent dephosphorylation of VEGF receptor-2, and that VEGF receptor-2 activity regulates VE-cadherin tyrosine phosphorylation, endothelial cell polarity and lumen formation.


Assuntos
Polaridade Celular , Endotélio Vascular/citologia , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Animais , Endotélio Vascular/enzimologia , Endotélio Vascular/metabolismo , Junções Intercelulares , Camundongos , Fosforilação , Receptor TIE-2/metabolismo
14.
Cold Spring Harb Perspect Med ; 2(7): a006502, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22762016

RESUMO

Vascular endothelial growth factors (VEGFs) are master regulators of vascular development and of blood and lymphatic vessel function during health and disease in the adult. It is therefore important to understand the mechanism of action of this family of five mammalian ligands, which act through three receptor tyrosine kinases (RTKs). In addition, coreceptors like neuropilins (NRPs) and integrins associate with the ligand/receptor signaling complex and modulate the output. Therapeutics to block several of the VEGF signaling components have been developed with the aim to halt blood vessel formation, angiogenesis, in diseases that involve tissue growth and inflammation, such as cancer. In this review, we outline the current information on VEGF signal transduction in relation to blood and lymphatic vessel biology.


Assuntos
Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Transdução de Sinais , Animais , Humanos , Integrinas/metabolismo , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica , Neuropilinas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
Mol Aspects Med ; 32(2): 88-111, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21565214

RESUMO

Vascular endothelial growth factors (VEGFs) are critical regulators of vascular and lymphatic function during development, in health and in disease. There are five mammalian VEGF ligands and three VEGF receptor tyrosine kinases. In addition, several VEGF co-receptors that lack intrinsic catalytic activity, but that indirectly modulate the responsiveness to VEGF contribute to the final biological effect. This review describes the molecular features of VEGFs, VEGFRs and co-receptors with focus on their role in the treatment of cancer.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Células Endoteliais/metabolismo , Humanos , Inflamação/metabolismo , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica/fisiologia , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
16.
Exp Hematol ; 38(3): 180-90, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20035824

RESUMO

OBJECTIVE: Internal tandem duplication (ITD) mutations of the FLT3 receptor are associated with a high incidence of relapse in acute myeloid leukemia (AML). Expression of the CXCR4 receptor in FLT3-ITD-positive AML is correlated with poor outcome, and inhibition of CXCR4 was shown to sensitize AML blasts toward chemotherapy. The aim of this study was to evaluate the impact of FLT3-ITD on cell proliferation and CXCR4-dependent migration in human hematopoietic progenitor cells and to investigate their response to CXCR4 inhibition. MATERIALS AND METHODS: We used primary blasts from patients with FLT3-ITD or FLT3 wild-type AML. In addition, human CD34(+) hematopoietic progenitor cells were transduced to >70% with retroviral vectors containing human FLT3-ITD. RESULTS: We found that FLT3-ITD transgene overexpressing human hematopoietic progenitor cells show strongly reduced migration toward stromal-derived factor-1 in vitro and display significantly reduced bone marrow homing in nonobese diabetic severe combined immunodeficient mice. Cocultivation of FLT3-ITD-positive AML blasts or hematopoietic progenitor cells on bone marrow stromal cells resulted in a strong proliferation advantage and increased early cobblestone area-forming cells compared to FLT3-wild-type AML blasts. Addition of the CXCR4 inhibitor AMD3100 to the coculture significantly reduced both cobblestone area-forming cells and proliferation of FLT3-ITD-positive cells, but did not affect FLT3-wild-type cells-highlighting the critical interaction between CXCR4 and FLT3-ITD. CONCLUSION: CXCR4 inhibition to decrease cell proliferation and to control the leukemic burden may provide a novel therapeutic strategy in patients with advanced FLT3-ITD-positive AML.


Assuntos
Células da Medula Óssea/patologia , Leucemia Mieloide/patologia , Receptores CXCR4/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo , Doença Aguda , Animais , Benzilaminas , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Ciclamos , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Compostos Heterocíclicos/farmacologia , Humanos , Leucemia Experimental/genética , Leucemia Experimental/metabolismo , Leucemia Experimental/patologia , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mutação , Fosforilação , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/metabolismo , Transplante Heterólogo , Proteínas Supressoras de Tumor/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética
18.
Blood ; 107(10): 4011-20, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16455956

RESUMO

Mutations of the nucleophosmin (NPM1) gene have recently been described in patients with acute myeloid leukemia (AML). To clarify the prevalence as well as the clinical impact of this mutation, we investigated 1485 patients with AML for NPM1 exon 12 mutations using fragment analysis. A 4 bp insert was detected in 408 of 1485 patients (27.5%). Sequence analysis revealed known mutations (type A, B, and D) as well as 13 novel alterations in 229 analyzed cases. NPM1 mutations were most prevalent in patients with normal karyotype (NK) (324 of 709; 45.7%) compared with 58 of 686 with karyotype abnormalities (8.5%; P < .001) and were significantly associated with several clinical parameters (high bone marrow [BM] blasts, high white blood cell [WBC] and platelet counts, female sex). NPM1 alterations were associated with FLT3-ITD mutations, even if restricted to patients with NK (NPM1-mut/FLT3-ITD: 43.8%; versus NPM1-wt/FLT3-ITD: 19.9%; P < .001). The analysis of the clinical impact in 4 groups (NPM1 and FLT3-ITD single mutants, double mutants, and wild-type [wt] for both) revealed that patients having only an NPM1 mutation had a significantly better overall and disease-free survival and a lower cumulative incidence of relapse. In conclusion, NPM1 mutations represent a common genetic abnormality in adult AML. If not associated with FLT3-ITD mutations, mutant NPM1 appears to identify patients with improved response toward treatment.


Assuntos
Leucemia Mieloide Aguda/genética , Mutação , Proteínas Nucleares/genética , Adulto , Sequência de Aminoácidos , Sequência de Bases , Elementos de DNA Transponíveis , Éxons , Humanos , Cariotipagem , Leucemia Mieloide Aguda/mortalidade , Leucemia Mieloide Aguda/patologia , Microscopia Confocal , Dados de Sequência Molecular , Nucleofosmina , Reação em Cadeia da Polimerase , Prevalência , Prognóstico
19.
Biochem J ; 371(Pt 2): 443-9, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12519077

RESUMO

Prostanoid receptors belong to the class of heptahelical plasma membrane receptors. For the five prostanoids, eight receptor subtypes have been identified. They display an overall sequence similarity of roughly 30%. Based on sequence comparison, single amino acids in different subtypes of different species have previously been identified by site-directed mutagenesis or in hybrid receptors that appear to be essential for ligand binding or G-protein coupling. Based on this information, a series of mutants of the human FP receptor was generated and characterized in ligand-binding and second-messenger-formation studies. It was found that mutation of His-81 to Ala in transmembrane domain 2 and of Arg-291 to Leu in transmembrane domain 7, which are putative interaction partners for the prostanoid's carboxyl group, abolished ligand binding. Mutants in which Ser-263 in transmembrane domain 6 or Asp-300 in transmembrane domain 7 had been replaced by Ala or Gln, respectively, no longer discriminated between prostaglandins PGF(2alpha) and PGD(2). Thus distortion of the topology of transmembrane domains 6 and 7 appears to interfere with the cyclopentane ring selectivity of the receptor. PGF(2alpha)-induced inositol formation was strongly reduced in the mutant Asp-300Gln, inferring a role for this residue in agonist-induced G-protein activation.


Assuntos
Receptores de Prostaglandina/metabolismo , Substituição de Aminoácidos , Sequência de Bases , Sítios de Ligação , Primers do DNA , Dinoprosta/metabolismo , Feminino , Humanos , Cinética , Ligantes , Mutagênese Sítio-Dirigida , Placenta/metabolismo , Gravidez , Receptores de Prostaglandina/química , Receptores de Prostaglandina/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA